Neurodevelopment and Schizophrenia
Neurodevelopment and Schizophrenia
Since the early 1990s, developmental neurobiology has made important strides towards eluci-
dating the pathophysiology of psychiatric disorders. Nowhere has this link between basic science
and clinical insights become more evident than in the field of schizophrenia research. In this
volume, the editors bring together some of the most active investigators in this field. Each con-
tributor provides an up-to-date overview of the relevant research, including directions for further
investigation.
The book begins with a section on advances in developmental neurobiology. This is followed
by sections on etiological and pathophysiological developments, and models that integrate this
knowledge. The final section addresses the clinical insights that emerge from the developmental
models and sets the scene for future efforts at early detection and prevention of schizophrenia.
This book will be valuable to researchers in psychiatry and neurobiology, students in medicine
and psychology, and all mental health practitioners.
http://avaxhome.ws/blogs/ChrisRedfield
Neurodevelopment and
Schizophrenia
Edited by
Matcheri S. Keshavan
University of Pittsburgh and Western Psychiatric Institute and Clinic,
Pittsburgh, PA and Wayne State University, Detroit, MI, USA
James L. Kennedy
University of Toronto Centre for Mental Health and Addiction,
Toronto, Canada
and
Robin M. Murray
King’s College London and Institute of Psychiatry, London, UK
p u b l i s h e d b y t h e p r e s s s y n d i c at e o f t h e u n i v e r s i t y o f c a m b r i d g e
The Pitt Building, Trumpington Street, Cambridge, United Kingdom
cambrid ge university press
The Edinburgh Building, Cambridge CB2 2RU, UK
40 West 20th Street, New York, NY 10011–4211, USA
477 Williamstown Road, Port Melbourne, VIC 3207, Australia
Ruiz de Alarcón 13, 28014 Madrid, Spain
Dock House, The Waterfront, Cape Town 8001, South Africa
http://www.cambridge.org
C Cambridge University Press 2004
A catalog record for this book is available from the British Library
The publisher has used its best endeavors to ensure that the URLs for external websites referred to in this
book are correct and active at the time of going to press. However, the publisher has no responsibility for
the websites and can make no guarantee that a site will remain live or that the content is or will remain
appropriate.
Every effort has been made in preparing this book to provide accurate and up-to-date information that is
in accord with accepted standards and practice at the time of publication. Nevertheless, the authors, editors,
and publisher can make no warranties that the information contained herein is totally free from error, not
least because clinical standards are constantly changing through research and regulation. The authors,
editors, and publisher therefore disclaim all liability for direct or consequential damages resulting from the
use of material contained in this book. Readers are strongly advised to pay careful attention to information
provided by the manufacturer of any drugs or equipment that they plan to use.
Contents
5 Brain plasticity and long-term function after early cerebral insult: the
example of very preterm birth 89
Matthew Allin, Chiara Nosarti, Larry Rifkin, and Robin M. Murray
v
vi Contents
14 How does drug abuse interact with familial and developmental factors
in the etiology of schizophrenia? 248
Chih-Ken Chen and Robin M. Murray
Index 473
viii
ix List of contributors
James L. Kennedy
Jeffrey A. Lieberman
Centre for Addiction and Mental Health,
Department of Psychiatry, UNC Conte
University of Toronto, 250 College St,
Center for Neuroscience of Mental
Toronto, ON M5T1R8, Canada
Disorders, University of North Carolina
Matcheri S. Keshavan School of Medicine, Chapel Hill,
University of Pittsburgh School of NC 27599, USA
Medicine, Department of Psychiatry and
Western Psychiatric Institute and Clinic, Beatriz Luna
Pittsburgh and the Department of Laboratory of Neurocognitive
Psychiatry and Behavioral Neurosciences, Development, Western Psychiatric
Wayne State University School of Medicine, Institute and Clinic, University of
4201 St Antoine Boulevard, Pittsburgh Medical Center, 3501 Forbes
Detroit, MI 48201, Ave, Pittsburgh, PA 15213,
USA USA
Timothy A. Klempan
Sahebarao P. Mahadik
Centre for Addiction and Mental Health,
Department of Psychiatry and Health
University of Toronto, 250 College St,
Behavior, Medical College of Georgia and
Toronto, ON M5T1R8,
Medical Research Service, VA Medical
Canada
Center, 1 Freedom Way, Augusta,
Eugenia Kravariti GA 30904, USA
Division of Psychological Medicine,
Institute of Psychiatry, De Crespigny Park, Colm McDonald
Denmark Hill, London SE5 8AF, UK Division of Psychological Medicine,
Institute of Psychiatry, De Crespigny Park,
Christian W. Kreipke Denmark Hill, London SE5 8AF, UK
Cellular and Clinical Neurobiology,
Department of Psychiatry and Behavioral Darlene S. Melchitzky
Neurosciences, Wayne State University Department of Psychiatry, University of
School of Medicine, 540 E. Canfield, Pittsburgh, Pittsburgh, PA 15213 and
Detroit, MI 48201, USA Department of Biology, Mercyhurst
Stephen M. Lawrie College, Erie, PA 16546,
Division of Psychiatry, Royal Edinburgh USA
Hospital, Edinburgh EH10 5HF, UK
Karoly Mirnics
Pat Levitt Departments of Psychiatry and
Kennedy Center, Vanderbilt University, Neurobiology, University of Pittsburgh,
Nashville, TN 37235, USA Pittsburgh, PA 15213, USA
xi List of contributors
Ray Norbury
Michael A. Rosenthal
Division of Psychological Medicine,
Child Psychiatry Branch, National Institute
Institute of Psychiatry, De Crespigny Park,
of Mental Health, National Institutes of
Denmark Hill, London SE5 8AF, UK
Health, Bethesda, MD 20892, USA
Although both Kraepelin and Bleuler noted that premorbid abnormalities in child-
hood could be present many years before a schizophrenic psychosis developed
(Marenco and Weinberger, 2000), until the late 1980s and 1990s, most people
viewed schizophrenia as an adult-onset mental illness. As a result, most biologi-
cal studies focused on a search for possible neurodegenerative changes that might
account for the onset of the condition. During the 1960s and 1970s (see Garmezy,
1974; Offord and Cross, 1969), evidence began to accumulate from developmen-
tally oriented follow-up and follow-back studies that abnormalities in interpersonal
relationships, neurodevelopmental immaturities, and attentional deficits in child-
hood all predicted the later onset of schizophrenia (see Rutter and Garmezy, 1983).
However, it was not until 1987 (Murray and Lewis, 1987; Weinberger, 1987) that
psychiatrists concerned with adult patients firmly took on board the notion that
schizophrenia might be a neurodevelopmental disorder. Since then, there has been
a veritable explosion of research tackling this proposition using a variety of research
strategies. In parallel, there has been an upsurge in studies of brain development
and function, giving rise to a much better understanding of brain plasticity and of
the role of neurotransmitters in both normal functioning and disease states. Clearly,
the time is ripe for a book that brings together the findings and concepts deriving
from both basic and clinic neuroscience, in order to examine the neurodevelop-
mental hypothesis in a critical but constructive fashion and then to consider the
implications for clinical practice. This book does just that, and does it extremely
well.
There is abundant consistent evidence that there are strong genetic influences on
the underlying liability to develop schizophrenia. Accordingly, one key challenge for
the neurodevelopmental hypothesis is to make explicit how neurodevelopmental
risk factors relate to genetic risk for schizophrenia. It is appropriate, therefore, that
the book begins with an account of what is known about genetic influences on brain
development, followed by reviews of what has been learned about normal brain
development from structural and functional magnetic resonance imaging studies.
Contrary to some people’s view that brain development is confined to infancy, it is
evident that there are important neural changes that extend into late adolescence
xiii
xiv Foreword
and early adult life. It remains the case that we have much to learn still about how
these changes relate to both normal and abnormal development, but they provide
a possible basis for the transition from the premorbid features of childhood to the
overt psychosis seen in adult life. This transition is also discussed with respect to
the evidence that there may be new abnormal brain changes that occur at this time.
The velo-cardio-facial syndrome is put forward as a possible model for the inter-
play between genes, brain, behavior, and cognition, and it is also suggested that
structural magnetic resonance imaging could provide a useful endophenotype for
genetic studies. This reflects the growing awareness that susceptibility genes prob-
ably do not act directly on mental disorders and that, therefore, it may be useful to
examine genetic effects on neurobiological abnormalities representing more prox-
imal effects of genes.
The neurodevelopmental hypothesis has been accompanied by indications of
the possible contributory causal role of epigenetic factors, malnutrition, pre- and
perinatal risk factors (including infection), minor physical anomalies, and adverse
rearing environment. The evidence on all of these is succinctly reviewed, with the
conclusion that the risk effects are probably real even though their effect size is
small.
Initially, with the advent of persuasive findings in genetics and in biological psy-
chiatry more generally, it became unfashionable to consider either social risk factors
or the effects of drug abuse: both of which had constituted a major focus of interest
in the 1960s and 1970s. However, animal studies and migration studies of humans
have shown that there is now reasonable evidence that social factors are influen-
tial, even though we do not understand how they operate. What is quite different
from the 1960s and 1970s is the appreciation that it is necessary to understand
how social factors may impact on brain development and that there may well be
a synergistic interplay between environmental risk and genetic vulnerability, with
the risks largely dependent on the presence of genetic susceptibility. Much the same
message derives from the study of recreational drugs, with the specific suggestion
that the drug effects associated with heavy early usage affect neurotransmitters
in ways that may precipitate the onset of psychosis when combined with genetic
susceptibility.
Further chapters provide a more detailed consideration of the possible role of the
dopamine system, of mis-wired limbic lobe or thalamocortical circuitry, of estrogen
and X-chromosome effects on brain development, of premorbid structural brain
abnormalities, and of neurodegenerative models.
From a clinical perspective, it is crucial to know whether the neurodevelopmen-
tal features are specific to schizophrenia or apply to a broader range of psychiatric
conditions. In frustrating fashion, the evidence suggests both substantial common-
alities and important specificities. A key chapter considers whether the extensive
xv Foreword
Michael Rutter
REFERENCES
Garmezy, N. (1974). Children at risk: the search for the antecedents to schizophrenia. Schizophr
Bull 8: 14–55; 9: 90–125.
Marenco, S., Weinberger, D. R. (2000). The neurodevelopmental hypothesis of schizophrenia:
following a trail of evidence from cradle to grave. Dev Psychopath 12: 501–527.
Murray, R. M., Lewis, S. W. (1987). Is schizophrenia a neurodevelopmental disorder? Br Med
J 295: 681–682.
xvi Foreword
Offord, D. R., Cross, L. A. (1969). Behavioral anticedents of adult schizophrenia. Arch Gen
Psychiatry 21: 267–283.
Rutter, M., Garmezy, N. (1983). Developmental psychopathology. In Mussen’s Handbook of
Child Psychology, 4th edn, Vol. 4: Socialization, Personality, and Child Development, ed. M.
Hetherington, New York Wiley, pp. 775–911.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
Preface
This volume arises out of a widely perceived need to take stock of our new know-
ledge about the developmental pathophysiology of schizophrenia. In 1997, we pub-
lished our first book on this topic, entitled Neurodevelopment and Psychopathology.
That volume, which was surprisingly well received, covered neurodevelopmen-
tal approaches to adult psychopathology, though many of the chapters concerned
schizophrenia. The enormous progress made in the subsequent years, particularly
on schizophrenia, has led us to focus the current volume exclusively on this disorder,
arguably the most debilitating of all psychiatric illnesses.
Since the early 1990s, our understanding of the developmental origins of
schizophrenia has “come of age,” with impressive advances emerging from both
the basic sciences and clinical studies. In retrospect, it is clear that the early neu-
rodevelopmental models that emerged in the mid-1980s were extremely simplistic
and too often relied on speculations about cellular and molecular mechanisms that
were not subsequently confirmed. Furthermore, to a large extent, they ignored the
contribution of psychology and certainly they included no mention of the role of
the social environment. We believe that the recent explosion of knowledge in both
neuroscience and cognitive science, as well as in imaging and epidemiology, has
allowed us to begin to remedy such deficiencies. Therefore, this new book brings
together many of the most productive and admired investigators in those areas of
research, individuals who we believe have contributed most to contemporary devel-
opmental models of schizophrenia. Each of the chapters provides a state-of-the-art
overview of the authors’ area of expertise, including directions for the future.
We start with a section on recent advances in developmental neurobiology. The
current state of our knowledge of genetics is reviewed, including the recent identi-
fication and apparent replication of susceptibility genes for schizophrenia (Ch. 1).
The latter is no small cause for rejoicing as claims for the identification of genes had
previously materialized and dematerialized with disturbing regularity, rather like
the sightings of alien spacecraft. Then, progress in our understanding of the normal
development of the human brain and its structural (Ch. 2), functional (Ch. 3), and
cognitive (Ch. 4) properties is successively outlined. Such a perspective is vital since
schizophrenia researchers have all too often attempted to outline the abnormal
xvii
xviii Preface
psychology and physiology underlying the condition before the normal had been
charted. The important field of brain plasticity and its limits is then discussed in
Ch. 5 with its implications for long-term functioning and psychopathology.
The next section contains overviews of pathophysiology and etiology of
schizophrenia. The lessons for schizophrenia from the study of unusual genetic
disorders such as the velo-cardio-facial syndrome are discussed, as is the impact of
genetic loading for schizophrenia on brain structure (Chs. 7 and 8). The important
and now well-documented effects of early environmental factors such as perinatal
complications (Ch. 11) and nutritional anomalies (Ch. 9) on risk of schizophrenia
are reviewed. Then the newer field of the effects of risk factors nearer to the onset
of psychosis is considered, such as psychosocial adversity (Ch. 13) and drug abuse
(Ch. 14) in pathogenetics, their epigenetic interaction (Ch. 10), and their impact
on gene expression (Ch. 12). Chapter 6 outlines a novel theory on the etiological
role of stress on schizophrenia and how its effects may be mediated by gluco-
corticoids. At a pathophysiological level, the developmental dysregulation of the
neurotransmitter systems such as dopamine (Ch. 15), and the limbic (Ch. 16)
and thalamocortical (Ch. 17) circuitry are critically reviewed. The important
role of X chromosome and estrogens in brain development and its relevance for
schizophrenia are discussed in Ch. 18. The possible premorbid neurodegenerative
changes in the schizophrenic illness (Ch. 20) and the commonalities versus differ-
ences between developmental/degenerative changes in common neuropsychiatric
disorders beginning in childhood/adolescence (Ch. 21) are discussed.
In the final section, some of the important clinical questions that drive pathophys-
iological research are considered. Can we identify preschizophrenia children? What
do studies of those at high genetic risk of the disorder teach us? Does understand-
ing of pathophysiology lead to specific predictions for preventive and therapeutic
approaches?
For too long, the origins of schizophrenia have been considered to be shrouded
in mystery. However, the exciting advances that have been made in schizophre-
nia research in the past decades, captured in this volume, have made the disease
more comprehensible, even though the puzzle continues to unravel. We also hope
that this volume will be of value for both researchers and practicing clinicians.
Throughout this volume, the implications of research findings for clinical practice
are discussed. The book will have served its purpose if the topics discussed herein
provided stimulation and new learning for a new generation of researchers and
clinicians in their efforts to inch toward better scientific knowledge and therapeutic
possibilities as applied to the patient with schizophrenia.
We wish to express our sincere gratitude to Drs Vaibhav Diwadkar, Debra
Montrose, Raj Rajarethinam, and Vandana Shashi for providing peer reviews of
the chapters in this book. We are in particular grateful to Karol L. Rosengarth for
her painstaking efforts in formatting and proofreading this work.
Part I
Basic aspects
1
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
3
Table 1.1. Neurodevelopmental genes and reports of expression-based and genetic analysis
in schizophrenia
Mash1 Neurulation -
Notch NOTCH4 (6p21.3) (Wei & Hemmings, 2000 and
see text)
Delta DLL1 (6q27)
Neurogenin NGN1 (5q23-q31)
NeuroD NEUROD (2q32)
Sonic Hedgehog SHH (7q36)
Wnt WNT1 (12q12-q13) (Cotter et al., 1998; Miyaoka
et al., 1999)
Krox20 Patterning EGR2 (10q21.1-q22.1)
Hox HOXB (17q21.3) (Kennedy and Kidd,
unpublished)
Dlx DLX1 (2q32)
Emx EMX2 (10q26.1)
Gbx GBX2 (2q36-q37)
Nkx TITF1 (14q13)
Otx OTX2 (14q21-q22)
Pax PAX6 (11p13) (Stober et al., 1999)
POU POU3F3 (3p14.2)
NCAM Cell migration/ NCAM1 (11q23.1) (Vicente et al., 1997; Doherty
neurite extension et al., 1990)
L1CAM
N-Cadherin NCAD (18q11.2)
Reelin RELN (7q22) (Fatemi et al., 2000; Guidotti
et al., 2000)
NGF Neuronal NGFB (1p13.1)
BDNF Survival BDNF (11p13) (Muglia et al., 2003)
NT-3 NTF3 (12p13) (Nanko et al., 1994 and see text)
NT-4/5 NTF5 (19q13.3)
GDNF GDNF (5p13.1-p12) (Lee et al., 2001)
CNTF CNTF (11q12.2) (Thome et al., 1996 and see text)
SNAP25 Presynaptic/ SNAP25 (20p11.2-p12) (Tachikawa et al., 2001; Wong
exocytosis et al., 2003)
Syntaxin STX1A (7q11.23) (A. Wong et al., unpublished data)
Synaptobrevin VAMP1 (12p)
Synapsin SYN3 (22q12.3) (Ohmori et al., 2000) and see text
Complexin CPLX2 (5q35.3) (Harrison and Eastwood, 1998
and see text)
Synaptophysin SYP (Xp11.23-p11.22)
Synaptotagmin SYT1 (12cen-q21)
5 Genes and brain development
Neurulation
The coordinated development of the human brain, from a single cell to some 1012
neurons with a possible 1000 connections between each, is a feat of unimaginable
intricacy. The function of individual groups of neurons within this framework in
the control of essential processes such as learning and memory, perception, mood,
and motor activity adds a further level of complexity. While the differentiative
pathways followed by neurons are topologically similar to those in other cell types,
one unique aspect of nervous system morphogenesis is the essential connectivity
of its units through axonal and dendritic outgrowth, allowing for interactions
in a highly plastic system. Despite the apparent difficulties of dissecting such a
detailed system, the changes involved in neuronal specification and overall CNS
development are becoming increasingly well understood. Much of our present state
of awareness in the field has been gathered through research using simple organisms
such as sea slug, squid and nematode, while recombinant DNA technology has
allowed generalization to vertebrates through investigations in mice. The cascade
of events proceeding from early gastrulation of the embryo to maturation can now
be adequately described.
The events leading to formation of the neural tube are known collectively as
neurulation. Two distinct developmental programs may be used: primary neurula-
tion, during which the chordamesoderm instructs the overlying ectoderm to divide,
invaginate, and separate from the surface to form the neural tube, and secondary
neurulation, during which a cylindrical zone of cells descends into the embryo and
hollows to form the tube. In mammals, this appears to occur in regionally differ-
entiated processes, with primary neurulation in the anterior region and secondary
neurulation posterior to somite 35.
The first signals responsible for the determination of anterior/posterior identity
in the ectoderm during gastrulation emanate from Hensen’s node, a small group
of cells at the anterior end of the primitive streak. The neural ectoderm is induced
through vertical signals from the mesodermal tissues beneath and adjacent to it.
The neural ectoderm gives rise to the neural plate, which is polarized through the
dorsal–ventral and anterior–posterior axes. The edges of the plate fold upwards
to join at the dorsal midline, producing the neural tube. Cells overlying the dorsal
midline of the neural tube form neural crest cells and migrate to form the brain and
spinal cord from the anterior and posterior portions, respectively. A commitment
to cell fates is revealed with the closure of the tube, with ventricular CNS generated
from the interior of the tube and epithelial cells at the interior periphery eventually
forming neurons and glia.
The brain becomes further divided through a series of constrictions into
the forebrain (prosencephalon), midbrain (mesencephalon), and hindbrain
6 T. A. Klempan, P. Muglia, and J. L. Kennedy
is primarily endothelial and myocardial (Li et al., 1998) with minimal expression
in brain. A number of follow-up reports on NOTCH4 have failed to replicate the
original finding for both individual markers and haplotypes using case–control and
family-based association approaches (Fan et al., 2002; Imai et al., 2001b; Klempan
et al., 2001; McGinnis et al., 2001; Sklar et al., 2001; Swift-Scanlan et al., 2002; Ujike
et al., 2001). The extreme biases in transmission of NOTCH4 alleles witnessed in
the first study now appear more likely to be a false-positive association, although
an unusual population specific effect cannot be excluded since differences between
the African-American and European populations have been noted at the NOTCH4
locus (Luo et al., 2004).
Abnormalities of the Wnt signaling pathway have been suggested by several recent
studies which have described reductions of β-catenin and γ -catenin staining in the
CA3 and CA4 hippocampal subregions and increases in Wnt1 staining in these
regions of schizophrenic brains relative to controls (Cotter et al., 1998; Miyaoka
et al., 1999). Furthermore, levels of glycogen synthase kinase-3β (GSK-3β) are
significantly reduced in the prefrontal cortex of schizophrenia patients and Wnt
is known to act as a repressor of GSK-3β (Beasley et al., 2001). GSK-3β also par-
ticipates in apoptosis, a form of programmed cell death, and, therefore, aberrant
GSK-3β expression may provide a rationale for observations of irregular neuronal
distributions found in schizophrenia (Kozlovsky et al., 2002). Many of the signaling
components of the Wnt pathway have been localized and some of these map within
susceptibility regions for psychosis (Rhoads et al., 1999).
Segmentation
Patterning of the specialized cell groups that will eventually specify distinct regions
of the CNS is carried out by other groups of transcription factors, whose expression
is confined to discrete segments of the developing embryo. The compartmental-
ization granted by division of regions of the brain into segments (prosomeres in
the forebrain and rhombomeres in the hindbrain) prevents the mixing of various
lineages of cells and the activity of expressed genes and restricts the targets and
navigational properties of axons within these segments.
One gene, for example, that is known to be critical in the establishment of neural
tube boundaries is the zinc-finger transcription factor Krox20. Krox20 is expressed
in the neural plate in alternating segments (rhombomeres r3 and r5) prior to distinct
rhombomere formation, as revealed by lineage tracing studies in the chick. Mice that
are null for Krox20 display a disruption of segmental identity with a fused r2/r4/r6
region (Schneider-Manoury et al., 1997). The function of Krox20 is considered
comparable to that of the Drosophila pair-rule genes, which translate information
9 Genes and brain development
from previously expressed genes into periodic stripes of further expression. At the
early stages of segmental specification, however, there is very little conservation of
specific genes between flies and vertebrates.
One family of genes that is critical in the determination of regional identity along
the anterior–posterior axis is known as the homeobox (Hox) family. Based upon
homeotic genes in Drosophila, these master regulatory factors are strongly con-
served from fly to mammals. Homeobox proteins are characterized by a 60 amino
acid residue motif (the homeodomain), which binds to specific sequences of DNA.
The Hox genes show colinearity with positions of genes within clusters along the
chromosome corresponding to their domains of expression along the embryo.
Those genes located further 3 within the cluster are expressed both earlier during
development and in a more anterior location. The limits of rostral expression of
the Hox genes are strongly coordinated with the divisions between rhombomeres,
suggesting that they may be involved in the specification and/or maintenance of seg-
ment identity (Krumlauf, 1994). The influence of specific Hox members on actual
segment phenotype is illustrated by Hoxb1. The expression of Hoxb1 is particularly
strong in rhombomere r4 and loss of Hoxb1 in mutant mice transforms r4 to an r2
phenotype (Studer et al., 1996), while ectopic Hoxb1 expression in chick produces
the opposite result (Bell et al., 1999).
In addition to the Hox family of transcription factors, a number of other
homeodomain-containing proteins impart positional information to the devel-
oping brain. Mice lacking both Dlx1 and Dlx2, for instance, do not demonstrate
proper migration of cortical cells from subcortical regions and show disrupted cell
migration within the striatum (Anderson et al., 1997). In the absence of func-
tional Emx1 protein, the corpus callosum fails to develop, while Emx2 mutants
lack hippocampal dentate gyrus and Cajal–Retzius cells of the neocortex. These
Emx2 mutants are also similar to the reeler mouse, with disturbances of lamination
and neuronal migration (Mallamaci et al., 2000). The expression of Gbx2, required
in rostral hindbrain differentiation (Wassarman et al., 1997), acts in concert with
Otx2 to establish the isthmic signaling region (Broccoli et al., 1999). The function
of Otx2 is perhaps even more critical in neural induction, as targeted mutations of
Otx2 result in absence of rostral brain areas.
Another family of transcriptional regulatory factors expressed in the mammalian
forebrain with persistence into adulthood is known as the POU domain fam-
ily. Members of this group contain a POU homeodomain and a POU specific
domain and are generally expressed in restricted regions late in forebrain develop-
ment. The severe defects produced by mutations in many early patterning genes
(such as Hox) make these unlikely candidates for the subtle structural alterations
witnessed in schizophrenia; however, many members of the POU class of factors
10 T. A. Klempan, P. Muglia, and J. L. Kennedy
Cell adhesion
In the developing brain, once a neuron has become committed to its phenotype it
must migrate to its proper layer of the maturing brain (Ruiz i Altaba, 1994). Cell
adhesion molecules (CAMs) are cell membrane proteins that mediate adhesion
between neural cells, exerting a key role in the morphogenesis, differentiation, and
migration of neurons as well as in the guidance of outgrowing axons in the develop-
ing brain (see Fig. 1.1). CAMs can be classified functionally into calcium-dependent
and calcium-independent groups. The calcium-dependent category contains at
least 80 proteins belonging to the cadherin superfamily, while the Ca2+ -independent
CAMs comprise the immunoglobulin (Ig) superfamily (IgSF). Most of the cadherin
11 Genes and brain development
NCAM
120
NCAM
180
NgCAM
L1
N-cadherin
CNR cadherin
Fig. 1.1. Members of the cadherin and immunoglobulin superfamilies of cell adhesion molecules.
Several prominent members of these transmembrane protein families are depicted, with
known roles in neurite outgrowth and morphogenesis. CNR, cadherin neuronal-related
receptor; NCAM, neuronal cell adhesion molecule; Ng CAM, neurogenin cell adhesion
molecule.
superfamily genes are expressed in the brain, and the protein structure is character-
ized by a unique domain named the cadherin motif, which is involved in calcium
binding (Takeichi, 1990). The cadherin motif (also called the EF motif) is repeated
a variable number of times in the different members of the superfamily (Yagi and
Takeichi, 2000). In addition to the classic cadherins, other members of the pro-
tocadherins superfamily are cadherin neuronal-related receptors (CNRs) and the
so-called seven-pass transmembrane cadherins. The seven-pass transmembrane
cadherins have a transmembrane structure similar to G-protein-coupled recep-
tors and appear to be involved in polarity orientation of the developing neuron,
as shown in Drosophila (Usui et al., 1999). The CNRs are coded by a cluster of
13 genes that map to 5q31.1 in humans and play a role in both the formation of
neuronal circuits at the synaptic level and the strengthening of the synapse during
long-term potentiation (LTP) (Yagi and Takeichi, 2000). It is interesting to note that
a significant linkage region for schizophrenia, derived from meta-analysis of data
from 20 genome scans, is located in a 30 cM stretch across 5q23–34 (Lewis et al.,
2003), which includes the CNR gene cluster. CNRs are distinguished from the other
12 T. A. Klempan, P. Muglia, and J. L. Kennedy
cadherins that exhibit homophilic interactions since they show etherophilic inter-
actions. They have been observed to bind to the Reelin protein and to the integrins
(Karecla et al., 1996; Senzaki et al., 1999). The genetic structure of the CNRs resem-
ble the organization of genes of the immunologic system, such as the T cell receptor
genes (Wu and Maniatis, 1999).
The calcium-independent CAMs (the IgSF) are characterized by a large amino-
terminal extracellular domain that contains a variable number of Ig motifs and
fibronectin repeats, which characterize the various members of the family sharing
a similar organization (Fields and Itoh, 1996). NCAM is the most extensively stud-
ied and characterized protein within the IgSF. NCAM contains five Ig folds and two
fibronectin repeats in the extracellular part of the protein (Crossin and Krushel,
2000). Further to its role during CNS development, NCAM appears to be involved
in the remodeling, regeneration, and maintenance of neuronal connections and
pathways in the adult brain (Kiss et al., 2001; Murase and Schuman, 1999). NCAM
also plays a role in learning and memory, and in guidance of brain development
in response to environmental stimuli (Fields and Itoh, 1996). Changes in NCAM
expression are also observed during synaptic remodeling and LTP in the rat hip-
pocampus (Fields and Itoh, 1996). NCAM is encoded by a single gene (NCAM1)
consisting of 26 exons located at 11q23.1 (Bello et al., 1989), which undergoes
alternative splicing generating at least 30 different forms with different spatial and
temporal patterns of expression in vertebrates (Akbarian et al., 1996). Three NCAM
isoforms (NCAM 180, NCAM 140 and NCAM 120) represent the most common
variants. NCAM 180 knockout mice have minor brain development abnormal-
ities, such as decreased size of the olfactory bulb, anterior ventricle enlargement,
and hippocampal dentate gyrus thinning (Andrews and Wood, 1986; Treloar et al.,
1997). In addition, the NCAM 180 knockouts exhibit behavioural deficits during
learning tasks and in the prepulse inhibition of startle (Wood et al., 1998). Both Hox
and Pax transcription factors appear to have a regulatory role on NCAM transcrip-
tion (Jones et al., 1993). NCAM functions as a cell–cell and cell–matrix homophilic
and heterophilic adhesion receptor; however, the mechanisms of interaction are not
completely understood. The binding of NCAM activates transmembrane-signaling
reactions and contributes to the initiation of cellular responses implicated in synap-
tic plasticity. The signaling downstream of neural NCAM not only impacts on
morphological changes but also can affect transcription factor activation and gene
induction (Krushel et al., 1999). Two main pathways are activated in response
to NCAM-mediated interactions, represented by the mitogen-activated protein
kinase and phospholipase C cascades (Crossin and Krushel, 2000). Other pathways
involve the interaction of the NCAM molecule with tyrosine kinase-linked recep-
tors (Beggs et al., 1997). The function of NCAM is also regulated by the molecule’s
content of polysialic acid (Fields and Itoh, 1996). The PSA residue makes the NCAM
13 Genes and brain development
Neuronal survival
With the development of the nervous system, as axons innervate their appropriate
target regions and connections become established, a naturally occurring cell death
takes place to refine the complex network of connections. This process serves to
establish a balance between the requirements of the target tissue and the characteris-
tics of the innervating neuronal population (Purves, 1988). The primary molecules
involved in this target-derived regulation of neuronal survival and differentiation
15 Genes and brain development
BDNF
NGF NT-4/5 NT-3 ALL
N T R
are the neurotrophins (see Fig. 1.2). The neurotrophin family of growth factors
display functional similarity through the promotion of neuronal survival, as first
seen with nerve growth factor (NGF) (Levi-Montalcini, 1987) and later for BDNF
(Leibrock et al., 1989). Neurotrophins now also include neurotrophin 3 (NT-3)
(Ernfors et al., 1990; Hohn et al., 1990), neurotrophin 4/5 (NT-4/5) (Berkmeier
et al., 1991; Ip et al., 1992), and neurotrophin 6 (NT-6) (Gotz et al., 1994), all cloned
through homology to NGF and BDNF. These also play key roles in neuronal differ-
entiation, cell body growth, promotion of neurite sprouting/extension, and, in the
mature CNS, the control of short-term synaptic transmission and LTP, considered
a mechanism for the processes of memory and learning (Thoenen, 1995).
Neurotrophin signaling is thought to occur primarily through the Trk family
of tyrosine kinase receptors. Binding of neurotrophin ligand induces transphos-
phorylation of Trk receptors, initiating signal transduction through phosphory-
lation of further intracellular proteins. Specificity is exhibited in the binding of
neurotrophins for individual Trk receptors: TrkA is a receptor for NGF, TrkB is a
receptor for BDNF and NT-4/5, and TrkC is a receptor for NT-3. A further receptor
known as p75NTR binds all neurotrophins with low affinity and acts selectively to
increase or decrease the response to different neurotrophins in neurons express-
ing Trk receptors. In the absence of Trk receptors, p75NTR controls an apoptotic
response to both NGF and BDNF in some neurons.
16 T. A. Klempan, P. Muglia, and J. L. Kennedy
Neurotransmission
The process of neurotransmission at its most fundamental level involves regulated
exocytosis through recruitment of synaptic vesicles, fusion to the plasma mem-
brane and neurotransmitter release, and eventual re-establishment of the vesicle
pool through endocytosis. The processes and molecules associated with vesicu-
lar trafficking are also critical for cell division, fertilization, and secretion of hor-
mones and other proteins, and they are strongly conserved from yeast to human.
Exocytosis is regulated most centrally by a group of proteins known as soluble N-
ethylmaleimide-sensitive attachment factor protein receptors (SNAREs), which are
small (18–42 kDa) integral (or anchored) membrane proteins that interact with
each other through amphipathic helical domains known as SNARE motifs. The
three core SNARE proteins can be categorized into target-SNAREs (t-SNAREs)
such as SNAP-25 (synaptosomal-associated protein of 25 kDa) and the syntaxins,
and vesicle-SNAREs (v-SNAREs) such as the synaptobrevins (or VAMPs). While
this core complex of SNARE proteins is known to regulate vesicular fusion (Hodel,
1998), the cyclical and quantal aspects of neurotransmitter release are facilitated
through a host of synaptic protein interactions, which are seen to be quite strongly
location-specific among regions of the brain (Bark and Wilson, 1994). The array
of synaptic terminal proteins involved in generalized neurotransmission includes
the core complex and interactors, which are either soluble or bound to the synaptic
vesicle or plasma membrane, such as soluble SNAPs, N-ethylmaleimide sensitive
19 Genes and brain development
Synaptic
Synapsin
vesicle
ATP ADP
NSF
Synaptobrevin/VAMP
Syntaxin SNAP-25
Presynaptic membrane
Fig. 1.3. Synaptic terminal proteins involved in exocytosis. The core complex required for vesicle
fusion to the presynaptic membrane includes synaptosomal associated protein of 25 kDa
(SNAP-25), syntaxins, and synaptobrevins (or VAMP). Proteins such as N-ethylmaleimide-
sensitive factor (NSF), which recycle this complex, and synapsins, which interact between
the vesicle and the cytoskeleton, are also involved, as well as numerous others.
pontine nuclei, and cerebellar granule cells (also pancreatic β-cells and adrenal
chromaffin cells), and it is associated with regions of synaptic plasticity. Antisense
oligonucleotides to SNAP-25 inhibit axonal elongation in rat cortical neurons,
specifically preventing complete terminal differentiation (Osen-Sand et al., 1993).
A strain of mouse designated coloboma, with a 2 cM deletion of a region of chro-
mosome 2 containing the Snap-25 gene, exhibits hyperactivity and delayed devel-
opment (Hess et al., 1996; Heyser et al., 1995). This strain also has abnormalities
in dopaminergic, serotonergic, and glutamatergic transmission in dorsal striatum
(Raber et al., 1997). This suggests that it may act as a model of attention-deficit
hyperactivity disorder and other psychiatric conditions. Hyperactivity is corrected
on introduction of a Snap-25 transgene into the strain, indicating the specificity of
this feature to the gene (Hess et al., 1996).
Postmortem assays of SNAP-25 immunoreactivity in schizophrenia have revealed
changes in the inferior temporal and prefrontal association cortices (Thompson
et al., 1998), with decreased expression in Brodmann areas 10 and 20 and increased
expression in area 9. A study of hippocampal connectivity found reduced cortical
SNAP-25 protein, most notably in the terminal fields of entorhinal cortex pro-
jections of schizophrenics (Young et al., 1998). More recently, decreased SNAP-25
expression has been seen in the cerebellum of schizophrenics (Mukaetova-Ladinska
et al., 2002), further implicating this molecule in the etiology of the disease. Altered
levels of SNAP-25 have also been witnessed in cerebral spinal fluid by quantitative
dot blotting (Thompson et al., 1999), with significant elevation of SNAP-25 over
that in control subjects and those with headaches. A further study was able to con-
firm the original SNAP-25 findings in area 10 (along with reduced synaptophysin
expression) yet showed no alteration in SNAP-25 mRNA expression (Karson et al.,
1999). Enhanced cortical expression of SNAP-25 has been identified through sub-
traction suppression polymerase chain reaction (PCR) in several rodent models of
schizophrenia, including haloperidol-treated versus control and Fischer 344 versus
Lewis rats (Wong et al., 2003).
The human SNAP-25 gene has been localized to 20p11.2-12 (Maglott et al., 1996),
near a region identified in a multi-center schizophrenia genome scan (Moises et al.,
1995). Three single nucleotide polymorphisms have been identified (Barr et al.,
2000) in the 3 untranslated region of SNAP-25, and the transmission of alleles
of these polymorphisms have been evaluated in 200 small, nuclear schizophrenia
families. Preliminary results indicate unbiased transmission of SNAP-25 alleles and
haplotypes within these families (Wong et al., 2003). A polymorphic tetranucleotide
repeat identified in the SNAP-25 promoter region also shows no association with
affection, subtypes, or family history in a recent case–control study (Tachikawa et al.,
2001); consequently, further investigation of additional markers and sample groups
is warranted.
21 Genes and brain development
highlights the relative non-specificity of this process and suggests that changes may
arise from an abnormality of synaptic connectivity determined much earlier in
life. These quantifiable changes in protein levels reflect actual biochemical changes
at the presynaptic terminal, potentially leading to synaptic dysfunction, as loss of
neuropil is not evident in these regions. An imbalance in stoichiometry between
critical regulatory elements may have severe consequences for both response capa-
city following calcium influx and rates of vesicle fusion and recycling, leading to an
increase or deficit in neurotransmission at the synapse. These changes in neuronal
phenotype or maturational state may account for the disorganization and volume
loss witnessed in various brain regions.
Apart from the recent microarray-based analysis described (Mirnics et al., 2000;
also see Ch. 12), other reports are providing increasing evidence for cortical presy-
naptic dysfunction in schizophrenia, including findings of synaptic vesicle cluster-
ing (Soustek, 1989) and alterations of many presynaptic protein and mRNA levels.
In addition to a fundamental role in neurotransmission, these molecules are crit-
ical during early embryonic development, influence neurite outgrowth and growth
cone maturation, and regulate morphological plasticity (Hepp and Langley, 2001).
These changes in presynaptic protein expression are also affected by neurotrophic
factors such as BDNF (Tartaglia et al., 2001), thus pointing to the need for further
elaboration of epistasis in these pathways when examining genetic variants.
Conclusions
While a staggering number of genes are involved in the patterning and function of
the human brain, the rapid pace of human genome sequence analysis has facilitated
the isolation and further characterization of brain-expressed sequences. Many of the
proteins encoded by these genes will participate in complex biological activities
integral to the establishment and maintenance of proper neuronal connectivity,
which, in turn, provides a template for higher-order cognitive function. It is natural
to consider, therefore, given the demonstrated genetic liability for schizophrenia,
that a number of these loci are likely to be involved as either direct etiological
determinants or phenotypic modifiers of the disease. As most researchers would
now agree that schizophrenia is inherited in a decidedly non-Mendelian fashion,
the best question that we can ask at present would be what set of DNA alterations are
consistently linked to a particular phenotype of schizophrenia? Possible modifiers
of cognitive abilities are beginning to be found through assessment of candidate
gene influence on neuropsychological tests (Bilder et al., 2002; Egan et al., 2003)
and further genetic determinants of individual disease components will likely be
identified as the complexities of the human genome are unraveled.
24 T. A. Klempan, P. Muglia, and J. L. Kennedy
This chapter has outlined the role played in the developing brain by various
genes within broad functional categories. A myriad of other factors (both genetic
and environmental) await investigation, and the final understanding of schizophre-
nia etiology may only be known with improvements in methodologies for detection
of gene–gene and gene–environment interaction. Until fairly recently, the relative
permanency of individual cells and populations/networks within the brain was
undisputed; however, we now know that the CNS is highly plastic and responsive,
with the capacity for neurogenesis throughout life from discrete sets of neural stem
cells (Eriksson et al., 1998). We should perhaps not be surprised, therefore, to learn
that the complex set of phenotypes collectively known as schizophrenia is antici-
pated by an equally complex set of genetic alterations, from which only particular
combinations may be identified within any given individual at risk for the disorder.
Of particular interest in this regard is the recent description of epistasis between
alleles of the G72 and d-amino acid oxidase (DAOO) genes in conferring risk for
schizophrenia (Chumakov et al., 2002). Together with neuregulin 1 and dysbindin,
these genes may induce schizophrenia susceptibility through a common N-methyl-
d-aspartate receptor pathway (Cloninger, 2002), a pathway that could alter the
neurodevelopmental trajectory of the brain. If replicated, the finding of Chumakov
and colleagues would constitute the first significant observation of gene–gene inter-
action in the etiology of schizophrenia, with likely expansion as the interactions
between various susceptibility factors are uncovered in the years to come.
Acknowledgements
We thank the Canadian Institutes of Health Research for grant support and Mary
Smirniw for her assistance with this manuscript.
REFERENCES
Akatone, A., Kunugi, H., Tanaka, H., Nanko, S. (2002). Association analysis of polymorphic CGG
repeat in 5 UTR of the reelin and VLDLR genes with schizophrenia, Schizophr Res 58: 37–41.
Akbarian, S., Kim, J. J., Potkin, S. G., et al. (1996). Maldistribution of interstitial neurons
in prefrontal white matter of the brains of schizophrenic patients. Arch Gen Psychiatry 53:
425–436.
Anderson, S. A., Eisenstat, D. D., Shi, L., Rubenstein, J. L. R. (1997). Interneuron migration from
basal forebrain to neocortex: dependence on Dlx genes. Science 278: 474–476.
Andrews, P. L., Wood, K. L. (1986). Systemic baclofen stimulates gastric motility and secretion
via a central action in the rat. Br J Pharmacol 89: 461–467.
Angelucci, F., Mathe, A. A., Aloe, L. (2000). Brain-derived neurotrophic factor and tyrosine
kinase receptor TrκB in rat brain are significantly altered after haloperidol and risperidone
administration. J Neurosci Res 60: 783–794.
25 Genes and brain development
Arenas, E., Trupp, M., Akerud, P., Ibanez, C. F. (1995). GDNF prevents degeneration and promotes
the phenotype of brain noradrenergic neurons in vivo. Neuron 15: 1465–1473.
Arinami, T., Toru, M. (1996). No evidence for association between CNTF null mutant allele and
schizophrenia. Br J Psychiatry 169: 253.
Arinami, T., Takekoshi, K., Itokawa, M., Hamaguchi, H., Toru, M. (1996). Failure to find associ-
ations of the CA repeat polymorphism in the first intron and the Gly-63/Glu-63 polymor-
phism of the neurotrophin-3 gene with schizophrenia. Psychiatry Genet 6: 13–15.
Artavanis-Tsakonis, S., Muskavitch, M. A., Yedvobnick, Y. (1983). Molecular cloning of Notch,
a locus affecting neurogenesis in Drosophila melanogaster. Proc Natl Acad Sci USA 80:
1977–1981.
Barbeau, D., Liang, J. J., Robitalille, Y., Quirion, R., Srivastava, L. K. (1995). Decreased expression
of the embryonic form of the neural cell adhesion molecule in schizophrenic brains. Proc
Natl Acad Sci USA 92: 2785–2789.
Bark, I. C., Wilson, M. C. (1994). Regulated vesicular fusion in neurons: snapping together the
details. Proc Natl Acad Sci USA 91: 4621–4624.
Barr, C. L., Feng, Y., Wigg, K., et al. (2000). Identification of DNA variants in the SNAP-25 gene
and linkage study of these polymorphisms and attention-deficit hyperactivity disorder. Mol
Psychiatry 5: 405–409.
Beasley, C., Cotter, D., Khan, N., et al. (2001). Glycogen synthase kinase-3beta immunoreactivity
is reduced in the prefrontal cortex in schizophrenia. Neurosci Lett 302: 117–120.
Beggs, H. E., Baragona, S. C., Hemperly, J. J., Maness, P. F. (1997). NCAM140 interacts with the
focal adhesion kinase p125(fak) and the SRC-related tyrosine kinase p59(fyn). J Biol Chem
272: 8310–8319.
Bell, E., Wingate, R. J. T., Lumsden, A. (1999). Homeotic transformation of rhombomere identity
after localized Hoxb1 misexpression. Science 284: 2168–2171.
Bello, M. J., Salagnon, N., Rey, J. A., et al. (1989). Precise in situ localization of NCAM, ETS1,
and D11S29 on human meiotic chromosomes. Cytogenet Cell Genet 52: 7–10.
Berkmeier, L. R., Winslow, J. W., Kaplan, D. R., et al. (1991). Neurotrophin-5: a novel neurotrophic
factor that activates trk and trkB. Neuron 4: 189–201.
Bilder, R. M., Volavka, J., Czobor, P., et al. (2002). Neurocognitive correlates of the COMT
Val(158)Met polymorphism in chronic schizophrenia. Biol Psychiatry 52: 701–707.
Broccoli, V., Boncinelli, E., Wurst, W. (1999). The caudal limit of Otx2 expression positions the
isthmic organizer. Nature 401: 164–168.
Brouha, A. K., Weickert, C. S., Hyde, T. M., et al. (1996). Reductions in brain derived neurotrophic
factor mRNA in the hippocampus of patients with schizophrenia. In Proceedings of the Society
for Neuroscience. Washington, DC: Society for Neuroscience, pp. 1680.
Browning, M. D., Dudek, E. M., Rapier, J. L., Leonard, S., Freedman, R. (1993). Significant reduc-
tions in synapsin but not synaptophysin specific activity in the brains of some schizophrenics.
Biol Psychiatry 34: 529–535.
Casarosa, S., Fode, C., Guillemot, F. (1999). Mash1 regulates neurogenesis in the ventral telen-
cephalon. Development 126: 525–534.
Catsicas, S., Larhammar, D., Blomqvist, A., et al. (1991). Expression of a conserved cell-type-
specific protein in nerve terminals coincides with synaptogenesis. Proc Natl Acad Sci USA
88: 785–789.
26 T. A. Klempan, P. Muglia, and J. L. Kennedy
Chapouton, P., Schuurmans, C., Guillemot, F., Gotz, M. (2001). The transcription factor neuro-
genin 2 restricts cell migration from the cortex to the striatum. Development 128: 5149–5159.
Chumakov, I., Blumenfeld, M., Guerassimenko, O., et al. (2002). Genetic and physiological data
implicating the new human gene G72 and the gene for d-amino acid oxidase in schizophre-
nia. Proc Natl Acad Sci USA 99: 13675–13680.
Cloninger, C. R. (2002). The discovery of susceptibility genes for mental disorders. Proc Natl
Acad Sci USA 99: 13365–13367.
Cotter, D., Kerwin, R., al-Sarraji, S., et al. (1998). Abnormalities of Wnt signalling in schizophre-
nia: evidence for neurodevelopmental abnormality. Neuroreport 9: 1379–1383.
Crossin, K. L., Krushel, L. A. (2000). Cellular signaling by neural cell adhesion molecules of the
immunoglobulin superfamily. Dev Dyn 218: 260–279.
Cunningham, B. A. (1995). Cell adhesion molecules as morphoregulators. Curr Opin Cell Biol 7:
628–633.
D’Arcangelo, G., Curran, T. (1998). Reeler: new tales on an old mutant mouse. Bioessays 20:
235–244.
Dawson, E., Powell, J. F., Sham P. C., et al. (1995). An association study of a neurotrophin-3
(NT-3) gene polymorphism with schizophrenia. Acta Psychiatr Scand 92: 425–428.
de la Pompa, J. L. , Wakeham, A., Correia, K. M., et al. (1997). Conservation of the Notch signalling
pathway in mammalian neurogenesis. Development 124: 1139–1148.
DeSilva, U., D’Arcangelo, G., Braden, V. V., et al. (1997). The human reelin gene: isolation,
sequencing, and mapping on chromosome 7. Genome Res 7: 157–164.
Doherty, P., Walsh, F. S. (1996). CAM–FGF receptor interactions: a model for axonal growth.
Mol Cell Neurosci 8: 99–111.
Doherty, P., Fruns, M., Seaton, P., et al. (1990). A threshold effect of the major isoforms of NCAM
on neurite outgrowth. Nature 343: 464–466.
Durany, N., Michel, T., Zochling, R., et al. (2001). Brain-derived neurotrophic factor and neu-
rotrophin 3 in schizophrenic psychoses. Schizophr Res 52: 79–86.
Eastwood, S. L., Harrison, P. J. (2000). Hippocampal synaptic pathology in schizophrenia, bipolar
disorder and major depression: a study of complexin mRNAs. Mol Psychiatry 5: 425–432.
(2001). Synaptic pathology in the anterior cingulate cortex in schizophrenia and mood dis-
orders. A review and a Western blot study of synaptophysin, GAP-43 and the complexins.
Brain Res Bull 55: 569–578.
Eastwood, S. L., Burnet, P. W., Harrison, P. J. (2000). Expression of complexin I and II mRNAs
and their regulation by antipsychotic drugs in the rat forebrain. Synapse 36: 167–177.
Eastwood, S. L., Cotter, D., Harrison, P. J. (2001). Cerebellar synaptic protein expression in
schizophrenia. Neuroscience 105: 219–229.
Eckhardt, M., Muhlenhoff, M., Bethe, A., et al. (1995). Molecular characterization of eukaryotic
polysialyltransferase-1. Nature 373: 715–718.
Egan, M. F., Kojima, M., Callicott, J. H., et al. (2003). The BDNF val66met polymorphism affects
activity-dependent secretion of BDNF and human memory and hippocampal function. Cell
112: 257–269.
Ericson, J., Murh, J., Placzek, M., et al. (1995). Sonic hedgehog induces the differentiation of
ventral forebrain neurons: a common signal for ventral patterning within the neural tube.
Cell 81: 747–756.
27 Genes and brain development
Eriksson, P. S., Perfilieva, E., Bjork-Eriksson, T., et al. (1998). Neurogenesis in the adult human
hippocampus. Nat Med 4: 1313–1317.
Ernfors, P., Ibanez, C. F., Ebendal, T., Olson, L., Persson, H. (1990). Molecular cloning and neu-
rotrophic activities of a protein with structural similarities to nerve growth factor: develop-
mental and topographical expression in brain. Proc Natl Acad Sci USA 87: 5454–5458.
Falconer, D. S. (1951). Two new mutants trembler and reeler, with neurological actions in the
house mouse. J Genet 50: 192–201.
Fan, J. B., Tang, J. X., Gu, N. F., et al. (2002). A family-based and case–control association study
of the NOTCH4 gene and schizophrenia. Mol Psychiatry 7: 100–103.
Fatemi, S. H., Earle, J. A., McMenomy, T. (2000). Reduction in Reelin immunoreactivity in
hippocampus of subjects with schizophrenia, bipolar disorder and major depression. Mol
Psychiatry 5: 654–663, 571.
Ferreiro, B., Kintner, C., Zimmerman, K., Anderson, D. J., Harris, W. (1994). XASH1 genes
promote neurogenesis in Xenopus embryos. Development 120: 3649–3655.
Fields, R. D., Itoh, K. (1996). Neural cell adhesion molecules in activity-dependent development
and synaptic plasticity. Trends Neurosci 19: 473–480.
Gabriel, S. M., Haroutunian, V., Powchik, P., et al. (1997). Increased concentrations of presynaptic
proteins in the cingulate cortex of subjects with schizophrenia. Arch Gen Psychiatry 54:
559–566.
Gelernter, J., Van Dyck, C., van Kammen, D. P., et al. (1997). Ciliary neurotrophic factor null
allele frequencies in schizophrenia, affective disorders, and Alzheimer’s disease. Am J Med
Genet 74: 497–500.
Goffinet, A. M., Dernoncourt, C. (1991). Localization of the reeler gene relative to flanking loci
on mouse chromosome 5. Mamm Genome 1: 100–103.
Goggi, J., Pullar, I. A., Carney, S. L., Bradford, H. F. (2002). Modulation of neurotransmitter
release induced by brain-derived neurotrophic factor in rat brain striatal slices in vitro.
Brain Res 941: 34–42.
Gotz, R., Koster, R., Winkler, C., et al. (1994). Neurotrophin-6 is a new member of the nerve
growth factor family. Nature 372: 266–269.
Gower, H. J., Barton, C. H., Elsom, V. L., et al. (1988). Alternative splicing generates a secreted
form of N-CAM in muscle and brain. Cell 55: 955–964.
Grebb, J. A., Greengard, P. (1990). An analysis of synapsin II, a neuronal phosphoprotein, in
postmortem brain tissue from alcoholic and neuropsychiatrically ill adults and medically ill
children and young adults. Arch Gen Psychiatry 47: 1149–1156.
Greengard, P., Valtorta, F., Czernik, A. J., Benfenati, F. (1993). Synaptic vesicle phosphoproteins
and regulation of synaptic function. Science 259: 780–785.
Guidotti, A., Auta, J., Davis, J. M., et al. (2000). Decrease in reelin and glutamic acid decar-
boxylase67 (GAD67) expression in schizophrenia and bipolar disorder: a postmortem brain
study. Arch Gen Psychiatry 57: 1061–1069.
Guillin, O., Diaz, J., Carroll, P., et al. (2001). BDNF controls dopamine D3 receptor expression
and triggers behavioural sensitization. Nature 411: 86–89.
Ha, D. H., Robertson, R. T., Ribak, C. E., Weiss, J. H. (1996). Cultured basal forebrain cholinergic
neurons in contact with cortical cells display synapses, enhanced morphological features,
and decreased dependence on nerve growth factor. J Comp Neurol 373: 451–465.
28 T. A. Klempan, P. Muglia, and J. L. Kennedy
Han, H. Q., Bahler, M., Greengard, P., Nichols, R. A., Rubin, M. R. (1991). Induction of formation
of presynaptic terminals in neuroblastoma cells by synapsin IIb. Nature 349: 697–700.
Harrison, P. J., Eastwood, S. L. (1998). Preferential involvement of excitatory neurons in medial
temporal lobe in schizophrenia. Lancet 352: 1669–1673.
Hattori, M., Nanko, S. (1995). Association of neurotrophin-3 gene variant with severe forms of
schizophrenia. Biochem Biophys Res Commun 209: 513–518.
Hawi, Z., Straub, R. E., O’Neill, A., et al. (1998). No linkage or linkage disequilibrium
between brain-derived neurotrophic factor (BDNF) dinucleotide repeat polymorphism and
schizophrenia in Irish families. Psychiatry Res 81: 111–116.
Hepp, R., Langley, K. (2001). SNAREs during development. Cell Tissue Res 305: 247–253.
Hess, E. J., Collins, K. A., Wilson, M. C. (1996). Mouse model of hyperkinesis implicates SNAP-25
in behavioral regulation. J Neurosci 16: 3104–3111.
Heyser, C. J., Wilson, M. C., Gold, L. H. (1995). Coloboma hyperactive mutant exhibits delayed
neurobehavioral developmental milestones. Brain Res Dev Brain Res 89: 264–269.
Hodel, A. (1998). Snap-25. Int J Biochem Cell Biol 30: 1069–1073.
Hohn, A., Leibrock, J., Bailey, K., Barde, Y. A. (1990). Identification and characterization of a
novel member of the nerve growth factor/brain-derived neurotrophic factor family. Nature
344: 339–341.
Honer, W. G., Falkai, P., Young, C., et al. (1997). Cingulate cortex synaptic terminal proteins and
neural cell adhesion molecule in schizophrenia. Neuroscience 78: 99–110.
Honer, W. G., Falkai, P., Bayer, T. A., et al. (2002). Abnormalities of SNARE mechanism proteins
in anterior frontal cortex in severe mental illness. Cereb Cortex 12: 349–356.
Hynes, M., Rosenthal, A. (1999). Specification of dopaminergic and serotonergic neurons in the
vertebrate CNS. Curr Opin Neurobiol 9: 26–36.
Imai, C., Sugai, T., Iritani, S., et al. (2001a). A quantitative study on the expression of synapsin II
and N-ethylmaleimide-sensitive fusion protein in schizophrenic patients. Neurosci Lett 305:
185–188.
Imai, K., Harada, S., Kawanishi, Y., et al. (2001b). The (CTG)n polymorphism in the NOTCH4
gene is not associated with schizophrenia in Japanese individuals. BMC Psychiatry 1: 1.
(2001c). Polymorphisms in the promoter and coding regions of the synapsin III gene. A lack
of association with schizophrenia. Neuropsychobiology 43: 237–241.
Ip, N. Y., Ibanez, C. F., Nye, S. H., et al. (1992). Mammalian neurotrophin-4: structure, chromo-
somal localization, tissue distribution, and receptor specificity. Proc Natl Acad Sci USA 89:
3060–3064.
Jones, F. S., Holst, B. D., Minowa, O., De Robertis, E. M., Edelman, G. M. (1993). Binding and
transcriptional activation of the promoter for the neural cell adhesion molecule by HoxC6
(Hox-3.3). Proc Natl Acad Sci USA 90: 6557–6561.
Jonsson, E., Brene, S., Zhang, X. R., et al. (1997). Schizophrenia and neurotrophin-3 alleles. Acta
Psychiatr Scand 95: 414–419.
Jovanovic, J. N., Czernik, A. J., Fienberg, A. A., Greengard, P., Sihra, T. S. (2000). Synapsins as
mediators of BDNF-enhanced neurotransmitter release. Nat Neurosci 3: 323–329.
Kao, H. T., Porton, B., Czernik, A. J., et al. (1998). A third member of the synapsin gene family.
Proc Natl Acad Sci USA 95: 4667–4672.
29 Genes and brain development
Karecla, P. I., Green, S. J., Bowden, S. J., Coadwell, J., Kilshaw, P. J. (1996). Identification of a
binding site for integrin alphaEbeta7 in the N-terminal domain of E-cadherin. J Biol Chem
271: 30909–30915.
Karson, C. N., Mrak, R. E., Schluterman, K. O., et al. (1999). Alterations in synaptic proteins
and their encoding mRNAs in prefrontal cortex in schizophrenia: a possible neurochemical
basis for “hypofrontality”. Mol Psychiatry 4: 39–45.
Kiss, J. Z., Troncoso, E., Djebbara, Z., Vutskits, L., Muller, D. (2001). The role of neural cell
adhesion molecules in plasticity and repair. Brain Res Brain Res Rev 36: 175–184.
Klempan, T. A., Trakalo, J. M., Pato, C. N., et al. (2001). Polymorphisms of the NOTCH4 gene
and schizophrenia. Am J Hum Genet 69 (Suppl. 4): 567.
Klempan, T. A., Trakalo, J., King, N., et al. (2002). Transmission of SYNAPSIN3 gene variants in
schizophrenia families. In Proceeding of the 52nd Annual Meeting of the American Society for
Human Genetics. Bethesda, MD: American Society for Human Genetics, p. 489.
Kohtz, J. D., Baker D. P., Corte, G., Fishell, G. (1998). Regionalization within the mammalian
telencephalon is mediated by changes in responsiveness to Sonic hedgehog. Development
125: 5079–5089.
Kortschak, R. D., Tamme, R., Lardelli, M. (2001). Evolutionary analysis of vertebrate Notch genes.
Dev Genes Evol 211: 350–354.
Kozlovsky, N., Belmaker, R. H., Agam, G. (2002). GSK-3 and the neurodevelopmental hypothesis
of schizophrenia. Eur Neuropsychopharmacol 12: 13–25.
Krebs, M. O., Guillin, O., Bourdell, M. C., et al. (2000). Brain derived neurotrophic factor (BDNF)
gene variants association with age at onset and therapeutic response in schizophrenia. Mol
Psychiatry 5: 558–562.
Kristiansen, L. V., Marques F. A., Soroka, V., et al. (1999). Homophilic NCAM interactions
interfere with L1 stimulated neurite outgrowth. FEBS Lett 464: 30–34.
Krumlauf, R. (1994). Hox genes in vertebrate development. Cell 78: 191–201.
Krushel, L. A., Cunningham, B. A., Edelman, G. M., Crossin, K. L. (1999). NF-kappaB activity
is induced by neural cell adhesion molecule binding to neurons and astrocytes. J Biol Chem
274: 2432–2439.
LaMantia, A. S. (1999). Forebrain induction, retinoic acid, and vulnerability to schizophrenia:
insights from molecular and genetic analysis in developing mice. Biol Psychiatry 46: 19–30.
Lawrie, S. M., Abukmeil, S. S. (1998). Brain abnormality in schizophrenia. A systematic and
quantitative review of volumetric magnetic resonance imaging studies. Br J Psychiatry 172:
110–120.
Lee, J. E. (1997). Basic helix–loop–helix genes in neural development. Curr Opin Neurobiol 7:
13–20.
Lee, J. E., Hollenberg, S. M., Lipnick, N., et al. (1995). Conversion of Xenopus ectoderm into
neurons by NeuroD, a basic helix–loop–helix protein. Science 268: 836–844.
Lee, K., Kunugi, H., Nanko, S. (2001). Glial cell line-derived neurotrophic factor (GDNF) gene
and schizophrenia: polymorphism screening and association analysis. Psychiatry Res 104:
11–17.
Leibrock, J., Lottspeich, F., Hohn, A., et al. (1989). Molecular cloning and expression of brain-
derived neurotrophic factor. Nature 341: 149–152.
30 T. A. Klempan, P. Muglia, and J. L. Kennedy
Levi-Montalcini, R. (1987). The nerve growth factor 35 years later. Science 237: 1154–1162.
Lewis, C. M., Levinson, D. F., Wise, L. H., et al. (2003). Genome scan meta-analysis of schizophre-
nia and bipolar disorder, part II: schizophrenia. Am J Hum Genet 73: 34–48.
Li, L., Huang, G. M., Banta, A. B., et al. (1998). Cloning, characterization, and the complete
56.8-kilobase DNA sequence of the human NOTCH4 gene. Genomics 51: 45–58.
Lin, L. H., Doherty, D. H., Lile, J. D., Bektesh, S., Collins, F. (1993). GDNF: a glial cell-derived
neurotrophic factor for midbrain dopaminergic neurons. Science 260: 1130–1132.
Lou, X. J., Bixby, J. L. (1995). Patterns of presynaptic gene expression define two stages of synaptic
differentiation. Mol Cell Neurosci 6: 252–262.
Luo, X. J., Klempan, T. A., Lappalainen, J., et al. (2004). NOTCH4 gene haplotype is associated
with schizophrenia in African-Americans. Biol Psychiatry 55: 112–117.
Maglott, D. R., Feldblyum, T. V., Durkin, A. S., Nierman, W. C. (1996). Radiation hybrid mapping
of SNAP, PCSK2, and THBD (human chromosome 20p). Mamm Genome 7: 400–401.
Maisonpierre, P., Belluscio, L., Friedman, B., et al. (1990). NT-3, BDNF, and NGF in the devel-
oping rat nervous system: parallel as well as reciprocal patterns of expression. Neuron 5:
501–509
Mallamaci, A., Mercurio, S., Muzio, L., et al. (2000). The lack of Emx2 causes impairment of Reelin
signaling and defects of neuronal migration in the developing cerebral cortex. J Neurosci 20:
1109–1118.
McEvilly, R. J., de Diaz, M. O., Schonemann, M. D., Hooshmand, F., Rosenfeld, M. G. (2002).
Transcriptional regulation of cortical neuron migration by POU domain factors. Science
295: 1528–1532.
McGinnis, R. E., Fox, H., Yates, P., et al. (2001). Failure to confirm NOTCH4 association with
schizophrenia in a large population-based sample from Scotland. Nat Genet 28: 128–129.
McMahon, A. P., Bradley, A. (1990). The Wnt-1 (int-1) proto-oncogene is required for the
development of a large region of the mouse brain. Cell 62: 1073–1085.
Mirnics, K., Middleton, F. A., Marquez, A., Lewis, D. A., Levitt, P. (2000). Molecular characteriza-
tion of schizophrenia viewed by microarray analysis of gene expression in prefrontal cortex.
Neuron 28: 53–67.
Mirnics, K., Middleton, F. A., Lewis D. A., Levitt, P. (2001). Analysis of complex brain disorders
with gene expression microarrays: schizophrenia as a disease of the synapse. Trends Neurosci
24: 479–486.
Miyaoka, T., Seno, H., Ishino, H. (1999). Increased expression of Wnt-1 in schizophrenic brains.
Schizophr Res 38: 1–6.
Moises, H. W., Kristbjarnarson, H., Wiese, C., et al. (1995). An international two-stage genome-
wide search for schizophrenia susceptibility genes. Nat Genet 11: 321–324.
Morris, P. J., Dawson, S. J., Wilson, M. C., Latchman, D. S. (1997). A single residue within the
homeodomain of the Brn-3 POU family transcription factors determines whether they
activate or repress the SNAP-25 promoter. Neuroreport 8: 2041–2045.
Morton, A. J., Faull, R. L. M., Edwardson, J. M. (2001). Abnormalities in the synaptic vesicle
fusion machinery in Huntington’s disease. Brain Res Bull 56: 111–117.
Muglia, P., Vicente, A. M., Verga, M., et al. (2003). Association between the BDNF gene and
schizophrenia. Mol Psychiatry 8: 147–148.
31 Genes and brain development
Mukaetova-Ladinska, E. B., Hurt, J., Honer, W. G., Harrington, C. R., Wischik, C. M. (2002).
Loss of synaptic but not cytoskeletal proteins in the cerebellum of chronic schizophrenics.
Neurosci Lett 317: 161–165.
Muller, D., Djebbara-Hannas, Z., Jourdain, P., et al. (2000). Brain-derived neurotrophic factor
restores long-term potentiation in polysialic acid-neural cell adhesion molecule-deficient
hippocampus. Proc Natl Acad Sci USA 97: 4315–4320.
Murase, S., Schuman, E. M. (1999). The role of cell adhesion molecules in synaptic plasticity and
memory. Curr Opin Cell Biol 11: 549–553.
Murphy, K. C., Jones, L. A., Owen, M. J. (1999). High rates of schizophrenia in adults with
velo-cardio-facial syndrome. Arch Gen Psychiatry 56: 940–945.
Nakayama, J., Fukuda, M. N., Fredette, B., Ranscht, B., Fukuda, M. (1995). Expression cloning of
a human polysialyltransferase that forms the polysialylated neural cell adhesion molecule
present in embryonic brain. Proc Natl Acad Sci USA 92: 7031–7035.
Nanko, S., Hattori, M., Kuwata, S., et al. (1994). Neurotrophin-3 gene polymorphism associated
with schizophrenia. Acta Psychiatr Scand 89: 390–392.
Nguyen Ba-Charvet, K. T., von Boxberg, Y., Godement, P. (1999). The mouse homeodomain
protein OTX2 regulates NCAM promoter activity. Brain Res Mol Brain Res 67: 292–295.
Nimgaonkar, V. L., Zhang, X. R., Brar, J. S., DeLeo, M., Ganguli, R. (1995). Lack of association
of schizophrenia with the neurotrophin-3 gene locus. Acta Psychiatr Scand 92: 464–466.
Ohmori, O., Shinkai, T., Hori, H., Kojima, H., Nakamura, J. (2000). Synapsin III gene polymor-
phisms and schizophrenia. Neurosci Lett 279: 125–127.
Ohtsuki, T., Ichiki, R., Toru, M., Arinami, T. (2000). Mutational analysis of the synapsin III gene
on chromosome 22q12-q13 in schizophrenia. Psychiatry Res 94: 1–7.
Osen-Sand, A., Catsicas, M., Staple, J. K., et al. (1993). Inhibition of axonal growth by SNAP-25
antisense oligonucleotides in vitro and in vivo. Nature 364: 445–448.
Parras, C. M., Schuurmans, C., Scardigli, R., et al. (2002). Divergent functions of the proneural
genes Mash1 and Ngn2 in the specification of neuronal subtype identity. Genes Dev 16:
324–338.
Persico, A. M., D’Agruma, L., Maiorano, N., et al. (2001). Reelin gene alleles and haplotypes as
a factor predisposing to autistic disorder. Mol Psychiatry 6: 150–159.
Proschel, M., Saunders, A., Roses, A. D., Muller, C. R. (1992). Dinucleotide repeat polymorphism
at the human gene for the brain-derived neurotrophic factor (BDNF). Hum Mol Genet 1:
353.
Purves, D. (1988) Body and Brain: A Trophic Theory of Neural Connections. Cambridge, MA:
Harvard University Press.
Raber, J., Mehta, P. P., Kreifeldt, M., et al. (1997). Coloboma hyperactive mutant mice exhibit
regional and transmitter-specific deficits in neurotransmission. J Neurochem 68: 176–186.
Rakic, P., Caviness, V. S., Jr. (1995). Cortical development: view from neurological mutants two
decades later. Neuron 14: 1101–1104.
Rhoads, A. R., Karkera, J. D., Detera-Wadleigh, S. D. (1999). Radiation hybrid mapping of genes
in the lithium-sensitive wnt signaling pathway. Mol Psychiatry 4: 437–442.
Rice, D. S., Curran, T. (2001). Role of the reelin signaling pathway in central nervous system
development. Annu Rev Neurosci 24: 1005–1039.
32 T. A. Klempan, P. Muglia, and J. L. Kennedy
Rodriguez, M. A., Pesold, C., Liu, W. S., et al. (2000). Colocalization of integrin receptors and
Reelin in dendritic spine postsynaptic densities of adult nonhuman primate cortex. Proc
Natl Acad Sci USA 97: 3550–3555.
Rosahl, T. W., Spillane, D., Missler, M., et al. (1995). Essential functions of synapsins I and II in
synaptic vesicle regulation. Nature 375: 488–493.
Ruiz i Altaba, A. (1994). Pattern formation in the vertebrate neural plate. Trends Neurosci 17:
233–243.
Sasaki, T., Dai, X. Y., Kuwata, S., et al. (1997a). Brain-derived neurotrophic factor gene and
schizophrenia in Japanese subjects. Am J Med Genet 74: 443–444.
Sakai, T., Sasaki, T., Tatsumi, M., et al. (1997b). Schizophrenia and the ciliary neurotrophic factor
(CNTF) gene: no evidence for association. Psychiatry Res 71: 7–10.
Schneider-Manoury, S., Seitanidou, T., Charnay, P., Lumsden, A. (1997). Segmental and neuronal
architecture of the hindbrain of Krox-20 mouse mutants. Development 124: 1215–1226.
Schonemann, M. D., Ryan A. K., McEvilly, R. J., et al. (1995). Development and survival of the
endocrine hypothalamus and posterior pituitary gland requires the neuronal POU domain
factor Brn-2. Genes Dev 9: 3122–3135.
Schwab, S. G., Albus, M., Hallmayer, J., et al. (1995). Evaluation of a susceptibility gene for
schizophrenia on chromosome 6p by multipoint affected sib-pair linkage analysis. Nat
Genet 11: 325–327.
Selemon, L. D., Rajkowska, G., Goldman-Rakic, P. S. (1995). Abnormally high neuronal density
in the schizophrenic cortex. A morphometric analysis of prefrontal area 9 and occipital area
17. Arch Gen Psychiatry 52: 805–818.
Sendtner, M., Carroll, P., Holtmann, B., Hughes, R., Thoenen, H. (1994). Ciliary neurotrophic
factor. J Neurobiol 25: 1436–1453.
Senzaki, K., Ogawa, M., Yagi, T. (1999). Proteins of the CNR family are multiple receptors for
Reelin. Cell 99: 635–647.
Sklar, P., Schwab, S. G., Williams, N. M., et al. (2001). Association analysis of NOTCH4 loci in
schizophrenia using family and population-based controls. Nat Genet 28: 126–128.
Snider, W. (1994). Functions of the neurotrophins during nervous system development: what
the knockouts are teaching us. Cell 77: 627–638.
Sokolov, B. P., Tcherepanov, A. A., Haroutunian, V., Davis, K. L. (2000). Levels of mRNAs
encoding synaptic vesicle and synaptic plasma membrane proteins in the temporal cortex
of elderly schizophrenic patients. Biol Psychiatry 48: 184–196.
Soustek, Z. (1989). Ultrastructure of cortical synapses in the brain of schizophrenics. Zentralbl
Allg Pathol 135: 25–32.
Steindler, D. A., Colwell, S. A. (1976). Reeler mutant mouse: maintenance of appropriate and
reciprocal connections in the cerebral cortex and thalamus. Brain Res 113: 386–393.
Stober, G., Syagailo, Y. V., Okladnova, O., et al. (1999). Functional PAX-6 gene-linked polymor-
phic region: potential association with paranoid schizophrenia. Biol Psychiatry 45: 1585–
1591.
Stober, G., Meyer, J., Nanda, I., et al. (2000). Linkage and family-based association study of
schizophrenia and the synapsin III locus that maps to chromosome 22q13. Am J Med Genet
96: 392–397.
33 Genes and brain development
Straub, R. E., MacLean, C. J., O’Neill, F. A., et al. (1995). A potential vulnerability locus for
schizophrenia on chromosome 6p24–22: evidence for genetic heterogeneity. Nat Genet 11:
287–293.
Studer, M., Lumsden, A., Ariza-McNaughton, L., Bradley, A., Krumlauf, R. (1996). Altered seg-
mental identity and abnormal migration of motor neurons in mice lacking Hoxb1. Nature
384: 630–634.
Sun, Y., Nadal-Vicens, M., Misono, S., et al. (2001). Neurogenin promotes neurogenesis and
inhibits glial differentiation by independent mechanisms. Cell 104: 365–376.
Swift-Scanlan, T., Lan, T. H., Fallin, M. D., et al. (2002). Genetic analysis of the (CTG)n NOTCH4
polymorphism in 65 multiplex bipolar pedigrees. Psychiatr Genet 12: 43–47.
Tachikawa, H., Harada, S., Kawanishi, Y., Okubo, T., Suzuki, T. (2001). Polymorphism of the 5 -
upstream region of the human SNAP-25 gene: an association analysis with schizophrenia.
Neuropsychobiology 43: 131–133.
Takahashi, M., Shirakawa, O., Toyooka, K., et al. (2000). Abnormal expression of brain-derived
neurotrophic factor and its receptor in the corticolimbic system of schizophrenic patients.
Mol Psychiatry 5: 293–300.
Takei, K., Chan, T. A., Wang, F. S., Deng, H., Rutishauser, U., Jay, D. G. (1999). The neural cell
adhesion molecules L1 and NCAM-180 act in different steps of neurite outgrowth. J Neurosci
19: 9469–9479.
Takeichi, M. (1990). Cadherins: a molecular family important in selective cell–cell adhesion.
Annu Rev Biochem 59: 237–252.
Tanaka, Y., Ujike, H., Fujiwara, Y., et al. (1998). Schizophrenic psychoses and the CNTF null
mutation. Neuroreport 9: 981–983.
Tartaglia, N., Du, J., Tyler, W. J., et al. (2001). Protein synthesis-dependent and -independent
regulation of hippocampal synapses by brain-derived neurotrophic factor. J Biol Chem 276:
37585–37593.
Tcherepanov, A. A., Sokolov, B. P. (1997). Age-related abnormalities in expression of mRNAs
encoding synapsin 1A, synapsin 1B, and synaptophysin in the temporal cortex of
schizophrenics. J Neurosci Res 49: 639–644.
Thoenen, H. (1995) Neurotrophins and neuronal plasticity. Science 270: 593–598.
Thome, J., Durany, N., Harsanyi, A., et al. (1996). A null mutation allele in the CNTF gene and
schizophrenic psychoses. Neuroreport 7: 1413–1416.
Thome, J., Durany, N., Palomo, A., et al. (1997a). Variants in neurotrophic factor genes and
schizophrenic psychoses: no associations in a Spanish population. Psychiatry Res 71: 1–5.
Thome, J., Jonsson, E., Foley, P., et al. (1997b). Ciliary neurotrophic factor null mutation and
schizophrenia in a Swedish population. Psychiatr Genet 7: 79–82.
Thompson, P. M., Sower, A. C., Perrone-Bizzozero, N. I. (1998). Altered levels of the synaptosomal
associated protein SNAP-25 in schizophrenia. Biol Psychiatry 43: 239–243.
Thompson, P. M., Rosenberger, C., Qualls, C. (1999). CSF SNAP-25 in schizophrenia and bipolar
illness. A pilot study. Neuropsychopharmacology 21: 717–722.
Treloar, H., Tomasiewicz, H., Magnuson, T., Key, B. (1997). The central pathway of primary
olfactory axons is abnormal in mice lacking the N-CAM-180 isoform. J Neurobiol 32: 643–
658.
34 T. A. Klempan, P. Muglia, and J. L. Kennedy
Tsai, M. T., Hung, C. C., Tsai, C. Y., et al. (2002). Mutation analysis of synapsin III gene in
schizophrenia. Am J Med Genet 114: 79–83.
Turner, E. E., Fedtsova, N., Jeste, D. V. (1997). Cellular and molecular neuropathology of
schizophrenia: new directions from developmental neurobiology. Schizophr Res 27: 169–
180.
Ujike, H., Takehisa, Y., Takaki, M., et al. (2001). NOTCH4 gene polymorphism and susceptibility
to schizophrenia and schizoaffective disorder. Neurosci Lett 301: 41–44.
Usui, T., Shima, Y., Shimada, Y., et al. (1999). Flamingo, a seven-pass transmembrane cadherin,
regulates planar cell polarity under the control of Frizzled. Cell 98: 585–595.
van Kammen, D. P., Poltorak, M., Kelley, M. E., et al. (1998). Further studies of elevated cere-
brospinal fluid neuronal cell adhesion molecule in schizophrenia. Biol Psychiatry 43: 680–
686.
Vicente, A. M., Macciardi, F., Verga, M., et al. (1997). NCAM and schizophrenia: genetic studies.
Mol Psychiatry 2: 65–69.
Virgos, C., Martorell, L., Valero, J., et al. (2001). Association study of schizophrenia with poly-
morphisms at six candidate genes. Schizophr Res 49: 65–71.
Wassarman, K. M., Lewandoski, M., Campbell, K., et al. (1997). Specification of the anterior
hindbrain and establishment of a normal mid/hindbrain organizer is dependent on Gbx2
gene function. Dev Suppl 124: 2923–2934.
Wassink, T. H., Nelson J. J., Crowe, R. R., Andreasen, N. C. (1999). Heritability of BDNF alleles
and their effect on brain morphology in schizophrenia. Am J Med Genet 88: 724–728.
Wei, J., Hemmings, G. P. (2000). The NOTCH4 locus is associated with susceptibility to
schizophrenia. Nat Genet 25: 376–377.
Wong, A., Macciardi, F., Klempan, T., et al. (2003). Identification of candidate genes for psychosis
in rat models, and possible association between schizophrenia and the 14-3-3η gene. Mol
Psychiatry 8: 156–166.
Wood, G. K., Tomasiewicz, H., Rutishauser, U., et al. (1998). NCAM-180 knockout mice display
increased lateral ventricle size and reduced prepulse inhibition of startle. Neuroreport 9:
461–466.
Wu, Q., Maniatis, T. (1999). A striking organization of a large family of human neural cadherin-
like cell adhesion genes. Cell 97: 779–790.
Yagi, T., Takeichi, M. (2000). Cadherin superfamily genes: functions, genomic organization, and
neurologic diversity. Genes Dev 14: 1169–1180.
Young, C. E., Arima, K., Xie, J., et al. (1998). SNAP-25 deficit and hippocampal connectivity in
schizophrenia. Cereb Cortex 8: 261–268.
2
Any parent of a teenager can tell you that the brain of a 13 year old is different
than the brain of an 8 year old; yet, actually pinning down the neuroanatomical
substrates of those differences has proved elusive. Nature has gone through a great
deal of trouble to protect the brain. It is wrapped in a tough leathery membrane,
surrounded by a protective moat of fluid, and completely encased in bone. This has
shielded the brain from falls or attacks from predators, but it has also shielded the
brain from scientists.
Magnetic resonance imaging (MRI) has changed that. It allows for the acquisition
of exquisitely accurate pictures of the living growing human brain and it does so
without the use of ionizing radiation. This has opened the door not only for scanning
healthy children but also for acquiring repeated scans throughout development.
Using MRI, the team at the Child Psychiatry Branch of the National Institute
of Mental Health has been collecting brain MRI scans on healthy children and
adolescents since 1989. As of 2003, we have acquired over 300 scans from 150
healthy subjects. The data presented in this chapter will be largely drawn from this
cohort unless otherwise stated.
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
35
36 J. N. Giedd, M. A. Rosenthal, A. B. Rose, et al.
can have a 50% difference in brain volume, (b) there are robust differences in brain
sizes between males and females with similar functional capacity, and (c) there is
a paucity of established correlations between the size of any given brain structure
and a specific cognitive ability. However, from a computational science perspective
it seems likely that the number of neuronal connections in a structure reflects its
information processing capacity.
Also, modest positive correlation between IQ and total cerebral volume and
a possible relationship between hippocampal size and memory recall have been
reported. The intricacy of various neurochemical systems and the diversity of affer-
ent and efferent connections to the many distinct nuclei of most brain structures
make straightforward relationships between volumes of a single structure and per-
formance on a particular cognitive task uncommon. This supports the concept of
distributed neural systems, whereby functional attributes are not thought to lie so
much within a single structure as within a network of structures. Across species there
is a strong correlation between body size and brain size (Jerison, 1991), and animals
with large brains such as elephants and dolphins demonstrate greater behavioral
complexity.
Total brain volume is approximately 90% of its adult size by age six years. Data
of total brain size, as is the case for data of the subcomponents of the brain, is char-
acterized by a high degree of variability (Fig. 2.1). This large variability necessitates
large sample sizes to detect group differences (Lange et al., 1997).
MRI is adept at discerning gray matter, white matter, and fluid on brain images.
These boundaries are used to define the size and shape of a variety of brain structures
or regions.
White matter
White matter consists of myelinated axons (Fig. 2.2). The amount of white matter
in the brain increases throughout childhood and adolescence. Myelination greatly
speeds up transmission between neurons, up to 100 times the speed in unmyelinated
fibers. The greater speed of neuronal processing may facilitate cognitive complexity.
The myelinated axons comprising white matter can be projectional, connecting the
brain to the brainstem or spinal cord; associational, connecting one type of brain
part to another; or commissural, connecting like parts of the brain in the left and
right hemispheres. Diffusion tensor imaging, a structural brain imaging technique,
may help to discern these different types of white matter connection although our
group has not yet acquired diffusion tensor imaging scans on pediatric populations
(see Ch. 3 for a discussion of this approach).
The most conspicuous white matter component of the brain is the corpus cal-
losum (CC). It is the main connection between the left and right hemispheres and
37 Brain development in healthy children
1800
1400
1200
1000
Female
Male
800
4 6 8 10 12 14 16 18 20 22
Age in years
NIH, DHHS.
Fig. 2.1. Total cerebral volume for 145 children and adolescents (ages 4–22) based on 243 brain
magnetic resonance imaging scans. (Jay N. Giedd, Child Psychiatry Branch, NIMH, NIH,
DHHS.)
Dendrites
Axon
Cell body
(the cell’s life
support center) Terminal
branches of
axon
Neuronal impulse
Myelin
sheath
Fig. 2.2. The neuron. (Donald Bliss, NIH Medical Arts and Photography Branch for Jay N. Giedd, Child
Psychiatry Branch, NIMH, NIH, DHHS.) (See plate section for color version.)
38 J. N. Giedd, M. A. Rosenthal, A. B. Rose, et al.
115
Parietal
110
Percentage Frontal
105
Temporal
100
Total brain
95
90
4 6 8 10 12 14 16 18 20 22
Age (in years)
Fig. 2.3. Standardized regional brain development for 145 children and adolescents (ages 4–22)
based on 243 brain magnetic resonance imaging scans. (Jay N. Giedd, Child Psychiatry
Branch, NIMH, NIH, DHHS.)
Gray matter
Gray matter consists largely of cell bodies and is home to the nucleus and
the dendrites, the antennae-like connections that receive input from other cells
(Fig. 2.2). Gray matter comprises the cortex (Latin for “bark”), the outermost layer
of the brain, and certain subcortical structures such as the basal ganglia and the
thalamus.
Initial cross-sectional studies showed a general decrease in the amount of cortical
gray matter during childhood, beginning at the earliest ages of the study design,
which was often around age 5 years (Jernigan et al., 1991; Pfefferbaum et al., 1994;
Sowell et al., 2002; Steen et al., 1997). However, when scans were acquired in the
same individuals at approximately two year intervals, cortical gray matter was shown
to follow an inverted U-type pattern (Fig. 2.3). In the frontal lobes, involved in
planning, organizing, strategizing, and other “executive” functions, the cortical
gray matter reaches its maximal thickness at 11.0 years in girls and 12.1 years
in boys (Giedd et al., 1997, 1999). Temporal lobe cortical gray matter peaks at
16.7 years in girls and 16.2 years in boys. Parietal lobe cortical gray matter peaks at
10.2 years in girls and 11.8 years in boys.
The thickening and thinning of gray matter is thought to reflect changes in the
size and complexity of neurons, not a change in the actual number. The increasing
size may reflect a process called arborization, as the cells grow extra branches, twigs,
and roots, thus growing “bushier” and making a greater number of connections
to other cells. The decreasing amount of gray matter may reflect the process of
pruning where certain connections are eliminated.
40 J. N. Giedd, M. A. Rosenthal, A. B. Rose, et al.
The forces guiding these processes of arborization and pruning are not well
understood. Genetics, nutrition, toxins, bacteria, viruses, hormones, and many
other factors have been shown to have an effect. One hypothesis for the pruning
phase is the “use it or lose it” principle, in which those connections that are used
well survive and flourish whereas those connections that are not used will wither
and die. If this hypothesis is correct, the activities of the child or teenager may have
a powerful influence on the ultimate physical structure of the brain.
Sex differences
Sex differences in the brain are particularly intriguing to psychiatrists studying chil-
dren and adolescents since nearly all psychiatric disorders of childhood onset have
different ages of onset, prevalence, and symptomatology between boys and girls.
Whether environmental or other genetic factors interact with normal differences
between boy and girl brains to account for some of these clinical differences is a
topic of active research (see Ch. 18 for a review of this topic).
In an analysis of 71 male and 50 female healthy subjects between the ages of 4
and 18 years, males had a 9% larger cerebral volume. This difference was consistent
across the entire age range of 4 to 18 years and is similar to that found in adult
postmortem brain (Dekaban and Sadowsky, 1978; Ho et al., 1980) and in vivo
imaging studies (Flaum et al., 1995).
Given the myriad of factors that determine brain structure size, and findings
of enlarged brains in patient groups such as those with fragile X syndrome (Reiss
et al., 1994) and autism (Piven et al., 1995), size difference should not be inter-
preted as imparting any sort of functional advantage or disadvantage. The fact that
gross structural size may be insensitive to sexual differences in receptor density
or connectivity between different neurons further emphasizes the complexity of
interpreting the implications of size differences in brain structures.
Other structures examined in that study included the lateral ventricles, caudate,
putamen, globus pallidus temporal lobe, amygdala, and hippocampus. Of these
structures, when adjusted for total cerebral volume difference by analysis of covari-
ance, only the basal ganglia demonstrated sex differences in mean volume, with the
caudate being relatively larger in females and the globus pallidus being relatively
larger in males.
The sexual dimorphism of the basal ganglia is interesting in light of the frequent
implication of basal ganglia involvement in neuropsychiatric disorders such as
attention-deficit hyperactivity disorder (Castellanos et al., 1994; Hynd et al., 1991)
and Tourette’s syndrome (Hyde et al., 1995; Peterson et al., 1993; Singer, 1993),
which have a higher incidence in males. Obsessive–compulsive disorder, which
41 Brain development in healthy children
has an approximately equal incidence for males and females in late adolescence
and adulthood, may be more common in boys in early childhood (Swedo et al.,
1989). Also, a developmental subtype of this disorder, which demonstrates a more
treatment-refractory course and neurological/basal ganglia abnormalities, is more
common in young boys (Blanes and McGuire, 1997).
Amygdala and hippocampal volume increased for both sexes, but with the amyg-
dala increasing significantly more in males than females and hippocampal volume
increasing more in females. The amygdala and hippocampus findings are consis-
tent with the preponderance of androgen receptors in the amygdala (Clark et al.,
1988) and of estrogen receptors in the hippocampus of rhesus monkeys (Sholl and
Kim, 1989). Further evidence of a relationship between estrogen and hippocampal
volume in rodents is the decreased fiber outgrowth and altered density of dendritic
spines in the hippocampus of gonadectomized adult females, which is reversed with
hormone replacement (Gould et al., 1990; Morse et al., 1986). Similarly, in humans,
women with gonadal hypoplasia have been found to have smaller hippocampi than
controls (Murphy et al., 1993).
Conclusions
MRI has allowed for the safe acquisition of brain scans in healthy children and
has helped to launch a new era of pediatric neuroscience. Characterization of nor-
mal brain development is imperative to assess the hypothesis that many of the
most severe neuropsychiatric disorders of childhood onset are manifestations of
deviations from that normative path. White matter volumes tend to increase in a
roughly linear fashion from ages 4 to 18 years and slopes do not differ significantly
by lobe of the brain. Gray matter volumes, by comparison, tend to follow an inverted
U-shaped developmental curve and differ by lobe. Sexual dimorphism in healthy
brain development may lead to differential vulnerability, which would account for
some of the clinical differences in childhood neuropsychiatric disorders.
REFERENCES
Bogen, J. E., Bogen, G. M. (1969). The other side of the brain. 3, The corpus callosum and
creativity. Bull Los Ang Neurol Soc 34: 191–220.
Castellanos, F. X., Giedd, J. N., Eckburg, P., et al. (1994). Quantitative morphology of the caudate
nucleus in attention deficit hyperactivity disorder. Am J Psychiatry 151: 1791–1796.
Clark, A. S., MacLusky, N. J., Goldman-Rakic, P. S. (1988). Androgen binding and metabolism
in the cerebral cortex of the developing rhesus monkey. Endocrinology 123: 932–
940.
Cook, N. D. (1986). The Brain Code: Mechanisms of Information Transfer and the Role of the Corpus
Callosum. London: Methuen.
Dekaban, A. S., Sadowsky, D. (1978). Changes in brain weight during the span of human life:
relation of brain weights to body heights and body weights. Ann Neur 4: 345–356.
Flaum, M., Swayze, V. W., O’Leary, D. S., et al. (1995). Brain morphology in schizophrenia: effects
of diagnosis, laterality and gender. Am J Psychiatry 152: 704–714.
Giedd, J. N., Castellanos, F. X., Casey, B. J., et al. (1994). Quantitative morphology of the corpus
callosum in attention deficit hyperactivity disorder. [See comments] Am J Psychiatry 151:
665–669.
Giedd, J. N., Rumsey, J. M., Castellanos, F. X., et al. (1996). A quantitative MRI study of the
corpus callosum in children and adolescents. Brain Res Dev Brain Res 91: 274–280.
Giedd, J. N., Castellanos, F. X., Rajapakse, J. C., Vaituzis, A. C., Rapoport, J. L. (1997). Sexual
dimorphism of the developing human brain. Prog Neuropsychopharmacol Biol Psychiatry 21:
1185–1201.
Giedd, J. N., Blumenthal, J., Jeffries, N. O., et al. (1999). Brain development during childhood
and adolescence: a longitudinal MRI study. Nat Neurosci 2: 861–863.
Gould, E., Allan, M. D., McEwen, B. S. (1990). Dendritic spine density of adult hippocampal
pyramidal cells is sensitive to thyroid hormone. Brain Res 525: 327–329.
Hellige, J. B., Cox, J. P., Litvac, L. (1979). Information processing in the hemispheres: selective
hemisphere activation and capacity limitations. J Exp Psychol Gen 108: 251–259.
Ho, K. C., Roessmann, U., Straumfjord, J. V., Monroe, G. (1980). Analysis of brain weight. I.
Adult brain weight in relation to sex, age, and race. Arch Pathol Lab Med 104: 635–639.
Hyde, T. M., Stacey, M. E., Coppola, R., et al. (1995). Cerebral morphometric abnormalities in
Tourette’s syndrome: a quantitative MRI study of monozygotic twins. Neurology 45: 1176–
1182.
Hynd, G. W., Semrud-Clikeman, M., Lorys, A. R., Novey, E. S., Eliopulos, D. (1990). Brain
morphology in developmental dyslexia and attention deficit disorder/hyperactivity. Arch
Neurol 47: 919–926.
(1991). Corpus callosum morphology in attention deficit–hyperactivity disorder: morphome-
tric analysis of MRI. J Learn Disabil 24: 141–146.
Innocenti, G. M., Manzoni, T., Spidalieri, G. (1974). Patterns of somesthetic messages transferred
through the corpus callosum. Exp Brain Res 19: 447–466.
Jerison, H. J. (1991). Brain Size and the Evolution of Mind. New York: American Museum of
Natural History.
Jernigan, T. L., Trauner, D. A., Hesselink, J. R., Tallal, P. A. (1991). Maturation of human
cerebrum observed in vivo during adolescence. Brain 114: 2037–2049.
43 Brain development in healthy children
Joseph, R. (1980). Awareness, the origin of thought, and the role of conscious self-deception in
resistance and repression. Psychol Rep 46: 767–781.
Kappers, C. U. A., Huber, G. C., Crosby, C. C. (1936). The Comparative Anatomy of the Nervous
System of Vertebrates including Man, Vol. 2. New York: MacMillan.
Lange, N., Giedd, J. N., Castellanos, F. X., Vaituzis, A. C., Rapoport, J. L. (1997). Variability of
human brain structure size: ages 4 to 20. Psychiatry Res Neuroimaging 74: 1–12.
Levy, J. (1985). Interhemispheric collaboration: single mindedness in the asymmetric brain. In
Hemisphere Function and Collaboration in the Child, ed. C. T. Best. New York: Academic
Press, pp. 11–32.
Levy, J., Trevarthen, C. (1981). Color-matching, color naming and color memory in split brain
patients. Neuropsychology 19: 523–541.
Morse, J. K., Scheff, S. W., DeKosky, S. T. (1986). Gonadal steroids influence axonal sprouting in
the hippocampal dentate gyrus: a sexually dimorphic response. Exp Neur 94: 649–658.
Murphy, D. G. M., DeCarli, C. D., Daly, E., et al. (1993). Effects of the X chromosome on female
brain: a study of turner syndrome using quantitative magnetic resonance imaging. Lancet
342: 1197–1200.
Njiokiktjien, C. (1991). Pediatric Behavioral Neurology, Vol. 3 The Child’s Corpus Callosum.
Amsterdam: Suyi Publications.
Parashos, I. A., Wilkinson, W. E., Coffey, C. E. (1995). Magnetic resonance imaging of the corpus
callosum: predictors of size in normal adults. J Neuropsychiatry Clin Neurosci 7: 35–41.
Paus, T., Zijdenbos, A., Worsley, K., et al. (1999). Structural maturation of neural pathways in
children and adolescents: in vivo study. Science 283: 1908–1911.
Peterson, B., Riddle, M. A., Cohen, D. J., et al. (1993). Reduced basal ganglia volumes in Tourette’s
syndrome using three-dimensional reconstruction techniques from magnetic resonance
images. Neurology 43: 941–949.
Peterson, B. S., Leckman, J. F., Duncan, J. S., Wetzles, R., Riddle, M. A. (1994). Corpus callo-
sum morphology from magnetic resonance images in Tourette’s syndrome. Psychiatry Res
Neuroimaging 55: 85–99.
Pfefferbaum, A., Mathalon, D. H., Sullivan, et al. (1994). A quantitative magnetic resonance
imaging study of changes in brain morphology from infancy to late adulthood. Arch Neurol
51: 874–887.
Piven, J., Arndt, S., Bailey, J., et al. (1995). An MRI study of brain size in autism. Am J Psychiatry
152: 1145–1149.
Rapoport, S. I. (1990). Integrated phylogeny of the primate brain, with special reference to
humans and their diseases. Brain Res Rev 15: 267–294.
Reiss, A. L., Lee, J., Freund, L. (1994). Neuroanatomy of fragile X syndrome: the temporal lobe.
Neurology 44: 1317–1324.
Rosenthal, R. Bigelow, L. (1972). Quantitative brain measurements in chronic schizophrenia.
Br J Psychiatry 121: 259–264.
Shanks, M. F., Rockel, A. J., Powel, T. P. S. (1975). The commissural fiber connections of the
primary somatic sensory cortex. Brain Res 98: 166–171.
Sholl, S. A., Kim, K. L. (1989). Estrogen receptors in the rhesus monkey brain during fetal
development. Dev Brain Res 50: 189–196.
44 J. N. Giedd, M. A. Rosenthal, A. B. Rose, et al.
Singer, H., Reiss, A. L., Brown, J. E., et al. (1993). Volumetric MRI changes in basal ganglia of
children with Tourette’s syndrome. Neurology 43: 950–956.
Sowell, E., Trauner, D. A., Gamst, A., Jernigan, T. L. (2002). Development of cortical and subcor-
tical brain structures in childhood and adolescence: a structural MRI study. Dev Med Child
Neurol 44: 4–16.
Spidalieri, G., Franchi, G., Guandalini, P. (1985). Somatic receptive-field properties of single
fibers in the rostral portion of the corpus callosum in awake cats. Exp Brain Res 58: 75–81.
Steen, R. G., Ogg, R. J., Reddick, W. E., Kingsley, P. B. (1997). Age-related changes in the pedi-
atric brain: quantitative MR evidence of maturational changes during adolescence. Am J
Neuroradiol 18: 819–828.
Swedo, S. E., Rapoport, J. L., Leonard, H., Lenane, M., Cheslow, D. (1989). Obsessive–compulsive
disorder in children and adolescents. Clinical phenomenology of 70 consecutive cases. Arch
Gen Psychiatry 46: 335–341.
Thompson, P., Giedd, J. N., Blanton, R. E., et al. (2000). Growth patterns in the developing brain
detected by using continuum mechanical tensor maps. Nature 404: 190–192.
Tomasch, J. (1954). Size, distribution and number of fibers in the human corpus callosum. Anat
Rec 119: 119–135.
Zaidel, D., Sperry, R. W. (1974). Memory impairment after commissurotomy in man. Brain 97:
263–272.
(1977). Some long-term effects of cerebral commissurotomy in man. Neuropsychology 15:
193–204.
3
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
45
46 B. Luna and J. A. Sweeney
capacity. Brain weight is more than 90% of the adult level by school age (Caviness
et al., 1996); however, myelination and synaptic pruning continue throughout this
period without dramatic consequences on gross brain anatomy. Investigating brain
function allows us to see the effects of these structurally subtle, but significant,
processes. The elimination of redundant synaptic connections supports quicker
and more efficient regional neuronal processes, allowing for more focused brain
function with the increased capacity for more complicated neuronal operations.
Myelination speeds up neuronal transmission and allows distant brain regions to
participate more effectively in widely distributed circuitry, which, allows for top-
down, prefrontal executive control of behavior.
It has only recently become feasible to use neuroimaging procedures in healthy
pediatric populations. The available neuroimaging methods, such as positron emis-
sion tomography (PET) and single photon emission tomography, depend upon
ionizing radiation. Consequently, these two lines of investigation, cognitive devel-
opment and brain maturation, had remained separate with limited efforts at rec-
onciling or integrating them. With the advent of functional MRI (fMRI), which
allows non-invasive in vivo investigation of brain processes that underlie cognitive
function, we can now study the link between these psychological and neurobi-
ological phenomena. This technique has been used to investigate how the func-
tional organization of brain regions contributing to cognitive abilities changes with
development to support the emergence of adult-level cognition. Initial studies have
provided crucial information for delineating the brain circuitry underlying healthy
cognitive development.
Electroencephalography and PET were initially used to characterize developmen-
tal changes in brain function. Thatcher (1991) measured electroencephalography
coherence among neocortical regions and reported an increase in coherence activity
throughout adolescence, especially between frontal and other cortical areas. PET
results support these findings by indicating that local cerebral resting metabolic
rates decrease in frontal, parietal, and temporal regions throughout childhood
and only reach adult levels in adolescence (Chugani, 1998). These are significant
findings indicating a late integration of frontal regions to widely distributed brain
function; however, there are limitations to these methods and findings. The elec-
troencephalography studies lack sufficient spatial resolution to localize adequately
developmental changes in specific brain regions. Because PET is invasive, it has
rarely been used to study healthy pediatric populations.
Stimulus
Mirror IR Mirror
Head Coil
Fig. 3.1. Schematic of the MR environment. IR, infrared; H.R., heart rate.
health risk from repeated MRI studies also permits longitudinal fMRI studies of
developmental changes in brain–behavior systems. This is especially important as it
provides a basis for actually following developmental trajectories in individual sub-
jects, rather than utilizing only cross-sectional studies to investigate developmental
trends. Since development often proceeds in bursts rather than in a smooth, grad-
ual way (Thatcher, 1991), longitudinal studies provide a better way to characterize
patterns of development. For these reasons, fMRI is a valuable tool for bridging
cognitive and biological studies of brain development.
The principle of fMRI is that neuronal activation associated with cognitive
activity results in an increased metabolic demand; that metabolic demand, in turn,
brings increased blood flow to the active region. The increase in blood flow produces
a change in the ratio of oxygenated to deoxygenated blood, which changes the
magnetic properties of blood in a way that can be detected. Using sophisticated
image reconstruction and analysis procedures, these changes in regional magnetic
properties can be used to provide a detailed picture of brain regions involved in
performing a particular cognitive act. The psychological tasks subjects perform in
the MRI scanner need to be carefully selected to probe specific cognitive processes
of interest. A diagram of the MRI environment is presented in Fig. 3.1.
There are two primary fMRI methods used for identifying brain regions under-
lying specific cognitive processes: block design and event-related procedures. The
block design is a boxcar method used to identify the group of regions participating
in a cognitive process. Blocks of experimental trials are contrasted with alternating
blocks of baseline brain function; analyses identify the voxels where signal fluctuates
in accordance with changing block trial types. Baseline trials can vary from eyes
closed or fixation to trials identical to experimental trials except for the variable
of interest. The boxcar design provides the best characterization of specific brain
regions. The advantage of this method is that it is relatively easy to implement and
analyze, and it generates a relatively high signal-to-noise ratio, resulting in robust
49 Cognitive development: fMRI studies
brain activation. The limitation to this method is that one cannot disassociate the
different cognitive processes occurring in each experimental trial since these are
averaged across blocks, limiting the ability to investigate the relative contributions
of each of the brain regions in a circuit. Event-related designs are used to answer
this concern by acquiring brain images time-locked to the occurrence of each cog-
nitive event in a trial. In this manner, different brain regions can be associated with
specific cognitive steps in a task and regions that are primary to each cognitive pro-
cess can be identified. This method, however, typically has a significantly reduced
signal-to-noise ratio compared with block design studies, generating modest areas
of brain activation. Because of the limitations of each method, investigators will
often use both designs. A block design is used to identify all the brain regions in a
circuit; event-related designs are then used to disentangle the specific contributions
of each region to a cognitive process of interest.
Studies using fMRI to characterize brain function in healthy adults are relatively
straightforward. However, pediatric studies necessitate the comparison of brain
activation between different age groups, which requires additional methodological
considerations and statistical analyses. Initial studies reported differences in brain
activation determined from visual inspection between a group of children and
separate, but similar, adult studies (Casey et al., 1995; Hertz-Pannier et al., 1997).
Presently, pediatric studies study children and adult comparison groups in the same
study. Testing subjects ranging in age from childhood to adulthood provides the
greatest power, as regression analyses can identify brain regions that change with
age.
Working memory
Working memory is the ability to maintain and manipulate information online
in order to guide goal-directed behavior (Baddeley, 1983). This circuitry has been
well delineated in the single-cell literature and is known to be subserved by a widely
50 B. Luna and J. A. Sweeney
distributed circuitry in which prefrontal regions play a primary role (Fuster, 1997;
Goldman-Rakic, 1992). Delay-dependent cells found in prefrontal cortex have the
unique feature of responding exclusively to the maintenance of information, as
distinct from sensory and motor responses. Brain regions with neurons having this
property and that are part of a circuitry that supports these processes are critical
for working memory.
Several pediatric studies have compared brain activation between children and
adults during performance of spatial and non-spatial working memory tasks. Work-
ing memory studies require that subjects retain a spatial location or a sequence of
non-spatial stimuli in memory and use this information to guide a future response.
These studies have either found similar brain function in children and adults or
increased activation in children relative to adults. In one spatial working memory
task, six children aged 6–11 years and six adults were asked to recall the location
of a visual stimulus that occurred in previous trials (Thomas et al., 1999). Despite
differences in performance, children demonstrated similar patterns of activation as
that in adults in their prefrontal and parietal regions. They did not, however, show
bilateral activation of cingulate cortex, temporal regions, and premotor regions,
which are known to underlie response preparation and were recruited by adults. In
another study, nine children aged 8–11 years performed a spatial working memory
task where they had to press a button corresponding to the location of a previ-
ous prompt (Nelson et al., 2000). The children activated the same brain regions as
adults, including dorsal prefrontal, anterior cingulate, and posterior parietal cor-
tices. These results suggest that the brain circuitry underlying working memory is
mainly in place by 8–11 years of age; however, differences in the relative contri-
bution of different regions of this circuitry may change with age. In a non-spatial
working memory n-back task, six children aged 9–11 years were required to press
a button when a letter was repeated with one intervening non-match letter (Casey
et al., 1995). Brain activation during these test trials were compared with blocks of
trials where subjects pressed a button whenever the letter “X” appeared. Coronal
slices focused on prefrontal regions. Results indicated a greater magnitude of acti-
vation in prefrontal cortex in children than adults. These results suggest that, while
the correct circuitry is accessed by children when they maintain information in
working memory, children rely on a greater contribution of prefrontal cortex than
do adults. Increased activation of prefrontal cortex could indicate either an imma-
ture specialization of the underlying neuronal processes, such as synaptic pruning,
or a higher reliance on prefrontal cortex during a task that is harder for children
to perform. (See below for issues regarding age-related performance differences in
developmental fMRI studies.) Increases in task difficulty are known to generate
increases in brain activation (Szameitat et al., 2002).
51 Cognitive development: fMRI studies
Other studies have found evidence for increased participation of brain regions
with age during working memory tasks. Thirteen subjects aged 9–18 years per-
formed a spatial working memory task during whole-brain imaging (Klingberg
et al. 2002). Subjects were presented with stimuli that appeared sequentially in a
4 by 4 grid for three or five steps. After a 1.5 second delay, subjects had to respond
if the present stimulus was in a location prompted in the previous sequence. Age-
related increases in activation were found in the superior frontal sulcus and intra-
parietal sulcus, which corresponded to increased working memory capacity. Similar
results were found in another spatial working memory task that was used to char-
acterize changes in brain function related to age progression in eight children, eight
adolescents, and seven adults (Kwon et al., 2002). A block design compared acti-
vation during trials where subjects had to press a key if an “O” appeared in the
same location as two previous trials (2-back) compared with rest trials where sub-
jects responded when the “O” stimulus appeared at center. Reaction time decreased
with age while accuracy remained stable. Multiple regression analyses were used
to correlate the magnitude of brain function with age. Results showed that brain
function increased with age throughout prefrontal and premotor regions as well as
throughout posterior parietal regions, while no age-related decreases in activation
were found. These results remained after factoring out performance differences
between age groups. These two studies are unique in that they looked at age as
a continuous variable from childhood to adulthood and, therefore, may be more
sensitive than studies on discrete age groups in identifying brain regions that show
subtle changes with age.
Taken together, these studies indicate that the basic brain circuitry underlying
spatial working memory is in place by childhood; however, with increasing age, there
are changes in the degree that different regions in this circuitry are recruited. The
prefrontal cortex, which is known to play a primary role in mature working memory,
demonstrates the most consistent change with age in these different studies.
cortex was imaged in nine children aged 7–12 years and nine adults while they
performed a go–no-go task (Casey et al., 1997). Subjects had to button press in
response to a centrally presented letter (go-trials) except when the letter was an “X”
(no-go trials). Children made more false-alarm errors than adults. Children acti-
vated the middle frontal gyrus (Broca’s area 9, 10, 46) as adults did; however, they
showed a greater volume of activation. This study, as in the working memory results,
provides evidence that children rely on increased participation of prefrontal cortex
during response suppression, which could be a result of changes in the underlying
anatomy or of increased effort by children to complete the task.
Evidence for increasing participation of multiple brain regions with age
during performance of tasks requiring voluntary response suppression has also been
provided by whole-brain fMRI. In one study, 16 children aged 8–12 years and 16
adults performed a combination task with flanker and go–no-go trials (Bunge et al.,
2001). In the flanker trials, subjects pressed a button corresponding to the direction
of a central arrow flanked on both sides by two arrows pointing in the opposite
direction to the central arrow (response interference), two arrows pointing in the
same direction as the central arrow (no response interference), diamond shapes
(no response interference), or “X” symbols to indicate no response (no-go trials).
Children made more interference and response-suppression errors than adults.
Results from correct trials only indicated that children, as well as adults, recruited
prefrontal cortex during successful interference suppression, but in the opposite
hemisphere. During successful response inhibition, children, unlike adults, relied
more heavily on posterior rather than prefrontal regions. Children did not recruit
right ventrolateral prefrontal cortex as adults did during both tasks. These results
indicate that, with age, there is increased participation of prefrontal regions during
response-suppression tasks. A study on adolescents showed similar results. Nine
subjects aged 12–19 years were compared with eight healthy adults aged 22–40 years.
Dorsal cortical regions were imaged while subjects performed a stop task, where
they were required to press a button only if a picture of an airplane was not followed
by a picture of a bomb (Rubia et al., 2000). Performance did not differ between
age groups; however, adolescents demonstrated reduced activation of prefrontal
regions and increased activation in caudate and inferior frontal gyrus relative to
adults. Age-related increases in prefrontal function were also found during a Stroop
interference task (Adleman et al., 2002). Thirty subjects aged 7–22 years performed
a Stroop test where subjects had to read a list of color words printed in black
font, name the color of a series of “X” symbols, and say the color of the ink of
color words that were discordant for color font. Dorsal cortical regions of the
brain were imaged. Results indicated that parietal involvement in the Stroop task is
mature by adolescence, while prefrontal activation continues to increase into adult-
hood. These results support findings from working memory studies indicating that
53 Cognitive development: fMRI studies
studies indicate that the increased integration of distant brain regions underlie
improved efficiency in the voluntarily cognitive control of behavior that occurs
throughout childhood and adolescence.
These results support other studies indicating that the increased participation
of brain regions that underlie executive and attentional control are important in
the maturation of voluntary response suppression. We extend these findings by
proposing that the recruitment of regions that underlie preparatory and refined
responses are also crucial to adult-level performance. Children are able to suppress
prepotent responses and interference, but what improves with age is the consis-
tency in which this behavioral state is maintained. We propose that this stage of
maturation, encompassing adolescence, may be characterized by increased reliance
on more widely distributed brain function that incorporates regions which are
not primary in the generating of a voluntary response but, instead, improve the
consistent control of behavior by a more concerted effort of the whole brain.
Developmental fMRI studies of both working memory and voluntary-response
suppression indicate that brain function does change with age and with increases
in performance; however, these changes are subtle and require further study. We
still need to characterize the relative contributions of localized brain regions and
the activities that are dependent on the integration of whole-brain processes. These
initial studies do show us that there are important changes occurring during adol-
escence that could be vulnerable either to emergence of schizophrenia or the neu-
rodevelopmental pathology that precedes it. With the background of methods and
findings from normative psychological developmental research, we can now begin
to explore possible failures in development that are associated with this disorder.
The occurrence of abnormalities of prefrontal processes in schizophrenia is now
widely supported by a range of histological, neuroimaging, and behavioral studies.
We have seen that this region is particularly sensitive to developmental changes and
may be vulnerable to impairment. Additionally, with neuroimaging techniques, we
can characterize the integrity of integrated brain function and begin to understand
the implications of localized impairment or failure to integrate the whole brain
and the relationship these might have to neurodevelopmental psychiatric disorders
(Luna and Sweeney, 1999; Luna et al., 2002). By studying at-risk populations, we
can examine when the developmental dysmaturation of executive abilities begins to
diverge most dramatically from normal processes (Ch. 23 reviews high-risk stud-
ies). As discussed below, we are still refining fMRI tools, especially with relevance
to pediatric populations; however, well-defined developmental experiments with
pediatric patient populations can begin to elucidate the nature of an impairment.
In the next section, we present how pediatric fMRI studies have informed us of
the development of other abilities and what they have shown us about impaired
development.
56 B. Luna and J. A. Sweeney
Abnormal populations
Pediatric fMRI studies have already provided insight into the possible factors under-
lying the etiology of developmental abnormalities such as ADHD and dyslexia.
Dyslexia
Dyslexia is an impairment in the ability to read that is not caused by reduced
intellectual capabilities. Neuroimaging studies indicate abnormalities in the
58 B. Luna and J. A. Sweeney
relative to the healthy group. These results suggest that abnormalities in frontal and
parietal heteromodal association cortex may precede illness onset and represent
a neurodevelopmental failure that contributes to risk for the disorder, indicating
that studies of the high-risk group may be valuable in testing neurodevelopmental
models of schizophrenia.
not tolerate the discomfort. Less-restrictive systems include using pillows to pack
the head into the head coil, minimizing range of movement, and visual feedback
of head location to help subjects to reposition themselves to their original position
(Thulborn and Shen, 1999). Training subjects to keep their heads still through sen-
sory feedback in a mock scanner also helps to minimize head motion (Slifer, 1996).
Pediatric neuroimaging studies must report the amount of head movement found
in their subjects lest their results be confounded by this artifact. This is of par-
ticular concern when studies find a reduction of activation throughout the brains
of children relative to adults with no regions demonstrating similar magnitude of
activation.
Children have faster respiratory and heart rate cycles than adults. Respiration can
lead to physiological artifact by introducing variations in the static magnetic field,
which can lead to spurious and decreased activation (Kemna and Posse, 2001; Raj
et al., 2001). Increased pulse rate can result in motion within the brain, affecting
brainstem structures the most, but also basal ganglia and, to a lesser degree, the
cortical surface (Gaillard et al., 2001b). Algorithms are being devised to remove
this artifact (Frank et al., 2001) but there is still limited experience in using such
approaches with pediatric samples.
A great misconception is that there are dramatic differences in the gross mor-
phology of the child and adult brains that make any direct comparison of functional
responses between age groups seriously flawed. This view implies that children’s
brains would be warped to a higher degree than adults when warping brain struc-
ture into a common adult brain atlas such as the Talairach atlas (Talairach and
Tournoux, 1988). Studies have shown, however, that brain size and gross morphol-
ogy at age 5 is not significantly different from that of adults (Caviness et al., 1996;
Giedd et al., 1996; Reiss et al., 1996) and the variability in brain structure between
children and adults is less than the normal variance between adult subjects. Nasal
sinuses continue to form into adolescence, perhaps resulting in increasing sources
of artifacts in brain regions bordering these areas. Neck length also continues to
increase developmentally, raising concerns regarding the optimal placement of a
child’s head on a head coil designed for an adult (Gaillard et al., 2001b). This could
result in children not reaching the “sweet” spot of the head coil where optimal
images are obtained.
Differences in performance
A major concern of pediatric neuroimaging is understanding if age differences in
brain function result from differences in performance or actually reflect differences
in functional brain systems (Bookheimer, 2000). Some investigators choose to use
tasks where children perform at the same levels as adults (Casey et al., 1995).
The primary result in these studies, perhaps not surprisingly, has been similar
61 Cognitive development: fMRI studies
activation across ages. When differences in activation are found, these may reflect
underlying differences in anatomy or differences in the computational demand
required to equate performance across age groups. However, age differences in
brain activation where performance levels of the task differ does not exclude the
possibility that adult patterns of activation would be evident in the child brain if
performance was equated. These two approaches are useful in answering different
questions regarding development. The former approach is needed to identify the
presence of mature brain circuitry, while the latter allows the characterization of
the immature aspects of the brain systems supporting a specific cognitive process.
Although mature circuitry may be accessed by children given optimal conditions,
it is important to characterize the brain regions that are not being accessed when
performance fails.
There are methods available to gain the benefits of both approaches. One
approach is to identify the brain regions that demonstrate age-related changes owing
to differences in performance and those that are independent of performance dif-
ferences. One study identified performance-related activity by identifying regions
that had greater activation in children that performed worse than adults but not
in children with equivalent performance levels as adults (Schlaggar et al., 2002).
Brain regions that show age-related differences in children with both poor and
equivalent performance as adults are independent from performance differences
and may reflect differences in the underlying neuroanatomy. Another approach is
to change task difficulty parametrically. As task difficulty increases, differences in
brain activation resulting from age differences should emerge. The variables that
consistently differ between ages across different task difficulties can be identified
as demonstrating developmental changes. In this manner, we can identify regions
that demonstrate age differences owing to performance differences and those that
are independent of performance and may indicate differences in neuroanatomy
(Benson et al., 1996).
Future directions
The goal of pediatric neuroimaging is to characterize the interaction between brain
maturation and cognitive development. We still need to characterize how synaptic
pruning and myelination processes contribute to age-related changes in the func-
tional organization of brain systems. The next step in pediatric neuroimaging is to
gain more direct characterization of brain maturation while obtaining indices of
brain function.
Diffusion tensor imaging (DTI) provides an indirect measure of myelination in
vivo by distinguishing the motion properties of water molecules inside axonal tracts
(Basser et al., 1994; Conturo et al., 1996; Le Bihan et al., 2001). The confinement
62 B. Luna and J. A. Sweeney
produced by myelin and tract tissue forces water molecules to diffuse rapidly in
an anisotropic manner in the direction of the fibers, in contrast to non-directional
isotropic diffusion outside fiber tracts (Peled and Yeshurun, 2001). Similar to fMRI,
DTI is a non-invasive and relatively short procedure (about 10 minutes) and can be
acquired using the same echo planar imaging sequence as fMRI, which enables
the direct overlay of images, permitting high resolution for localizing regions.
Initial DTI studies indicated a continued increase in frontal white matter anisotropy
throughout childhood (Klingberg et al., 1999). The high resolution of DTI allows
the tracking of subtle white matter differences that are not evident in white matter
volume, as well as sensitivity to subtle and dynamic changes in brain disorders.
Decreased anisotropy in left parietotemporal regions have been reported for adult
subjects with reading disabilities, providing evidence for impaired communication
between visual- and language-processing regions (Klingberg et al., 2000) and a pos-
sible mechanism for the impaired brain function demonstrated in these regions (see
above). DTI has also demonstrated reduced anisotropy in schizophrenia patients,
indicating a lack of connective integrity in white matter volumes throughout the
neocortex (Agartz et al., 2001; Foong et al., 2000; Lim et al., 1999). Understand-
ing the changes in myelination and other potential changes in white matter tracts
that concur with cognitive development will be pivotal in characterizing possible
dysmaturation in schizophrenia.
Phosphorus MR spectroscopy (31 P MRS) allows non-invasive in vivo investi-
gation of phosphorus-containing metabolites, which provide an indirect measure
of synaptic pruning (Keshavan et al., 1991; Stanley et al., 1996). MRS probes the
integrity of cell membranes by indicating the levels of phosphomonoesters (PMEs),
which are membrane phospholipid precursors, and phosphodiesters (PDEs), which
are phospholipid breakdown products (Keshavan et al., 1991). This can provide
an indirect measure of age-related changes in synaptic integrity. MRS has pro-
vided evidence for decreases in PMEs and increases in PDEs in prefrontal cortex
in schizophrenia (Pettegrew et al., 1993). The concurrent acquisition of fMRI and
MRS data could potentially reveal the contribution changes in synaptic integrity to
age-related changes in brain functional organization.
Finally, in order to increase power, validity, and reliability, pediatric studies
should include a larger number of subjects with equal distribution of individu-
als across each age group. Given the variability in rates of maturation, sources of
individual difference should be reduced. Variables such as gender, intelligence quo-
tient, and puberty would decrease intersubject variability. Control groups should be
carefully characterized and be free of any neurobiological abnormalities, including
metabolic and neurological disorders as well as psychiatric illness in the subject and
first-degree relatives. Most importantly, longitudinal studies should be performed
in a well-characterized group of healthy subjects who could be tested annually
63 Cognitive development: fMRI studies
with the same cognitive tasks. In this manner, developmental trajectories can be
delineated and also stage-like characterization of development can be understood.
Only with carefully designed studies with a large number of subjects, as suggested,
will we be able to generate normative data that is sensitive to subtle dysmaturation
processes present in psychiatric disorders.
Conclusions
We have recently been able to characterize the interface between cognitive devel-
opment and brain maturation using fMRI, a non-invasive neuroimaging method.
This is important to the understanding of neurodevelopmental models of dysmat-
uration in brain disorders such as schizophrenia. Initial pediatric fMRI studies have
shown that the basic circuitry supporting the capacity to perform a task is estab-
lished by childhood. Changes in the participation of frontal cortex and the ability
to recruit widely distributed brain circuitry support increases in the efficiency of the
cognitive control of behavior with age. This pattern was observed in studies inves-
tigating the cognitive development of working memory and voluntary response
suppression, and language processing in healthy individuals. Studies of abnormal
pediatric populations demonstrate impairments in frontal circuitry in ADHD and
parietotemporal regions in dyslexia. When using fMRI with children, special pre-
cautions must be taken to eliminate factors that have the potential to confound the
results. The future direction of fMRI includes the ability to probe brain anatom-
ical changes supporting the brain functional changes that are observed during
development.
REFERENCES
Adleman, N. E., Menon, V., Blasey, C. M., et al. (2002). A developmental fMRI study of the Stroop
color–word task. NeuroImage 16: 61–75.
Agartz, I., Andersson, J. L. R., Skare, S. (2001). Abnormal brain white matter in schizophrenia: a
diffusion tensor imaging study. Neuroreport 12: 2251–2254.
Armstrong, E., Schleicher, A., Omran, H., Curtis, M., Zilles, K. (1995). The ontogeny of human
gyrification. Cereb Cortex 5: 56–63.
Baddeley, A. D. (1983). Working memory. Philos Trans R Soc Lond B 302: 311–324.
Basser, P. J., Mattiello, J., Lebihan, D. (1994). Estimation of the effective self-diffusion tensor from
the NMR spin echo. J Magn Reson 103: 247–254.
Benson R. R., Logan W. J., Cosgrove G. R., et al. (1996). Functional MRI localization of language
in a 9-year-old child. Can J Neurol Sci 23: 213–219.
Bjorklund, D. F., Harnishfeger, K. K. (1990). The resources construct in cognitive development:
diverse sources of evidence and a theory of inefficient inhibition. Dev Rev 10: 48–71.
64 B. Luna and J. A. Sweeney
(1995). The evolution of inhibition mechanisms and their role in human cognition and behav-
ior. In Interference and Inhibition in Cognition, ed. F. N. Dempster, C. J. Brainerd. San Diego,
CA: Academic Press, pp. 141–173.
Bookheimer, S. Y. (2000). Methodological issues in pediatric neuroimaging. Mental Retard Dev
Disabil Res Rev 6: 161–165.
Booth, J. R., MacWhinney, B., Thulborn, K. R., et al. (1999). Functional organization of activation
patterns in children: whole brain fMRI imaging during three different cognitive tasks. Prog
Neuropsychopharmacol Biol Psychiatry 23: 669–682.
Bunge, S. A., Dudukovic, N. M., Thomason, M. E, Vaidya, C. J., Gabrieli, J. D. E. (2001). Immature
frontal lobe contributions to cognitive control in children: evidence from fMRI. Neuron 33:
301–311.
Case, R. (1992). The role of the frontal lobes in the regulation of cognitive development. Brain
Cogn 20: 51–73.
Casey, B. J., Cohen, J. D., Jezzard, P., et al. (1995). Activation of prefrontal cortex in children
during a nonspatial working memory task with functional MRI. NeuroImage 2: 221–229.
Casey, B. J., Trainor, R. J., Orendi, J. L., et al. (1997). A developmental functional MRI study of
prefrontal activation during performance of a go-no-go task. J Cogn Neurosci 9: 835–847.
Caviness, V. S., Kennedy, D. N., Bates, J. F., Makris, N. (1996). The developing human brain: a
morphometric profile. In Developmental Neuroimaging: Mapping the Development of Brain
and Behavior, ed. R. W. Thatcher, G. Reid Lyon, J. Rumsey, N. A. Krasnegor. New York:
Academic Press, pp. 3–14.
Changeux, J. P., Danchin, A. (1976). Selective stabilization of developing synapses as a mechanism
for the specification of neuronal networks. Nature 264: 705–712.
Chugani, H. T. (1998). A critical period of brain development: studies of cerebral glucose utiliza-
tion with PET. Prev Med 27: 184–188.
Cohen, J. D., Barch, D. M., Carter, C., Servan-Schreiber, D. (1999). Context-processing deficits
in schizophrenia: converging evidence from three theoretically motivated cognitive tasks.
J Abnorm Psychol 108: 120–133.
Conturo, T. E., McKinstry, R. C., Akbudak, E., Robinson, B. H. (1996). Encoding of anisotropic
diffusion with tetrahedral gradients: a general mathematical diffusion formalism and experi-
mental results. Magn Reson Med 35: 399–412.
Cox, R. W. (1996). AFNI: Software for analysis and visualization of functional magnetic resonance
neuroimages. Comput Biomed Res 29: 162–173.
Diamond, A., Goldman-Rakic, P. S. (1989). Comparison of human infants and rhesus monkeys
on Piaget’s AB task: evidence for dependence on dorsolateral prefrontal cortex. Exp Brain
Res 74: 24–40.
Eddy, W. F., Fitzgerald, M., Genovese, C. R., Mockus, A., Noll, D. C. (1996). Functional image
analysis software: computational olio. In Proceedings in Computational Statistics, ed. A. Prat.
Heidelberg: Physica-Verlag, pp. 39–49.
Eden, G. F., Van Meter, J. W., Rumsey, J. M., et al. (1996). Abnormal processing of visual motion
in dyslexia revealed by functional brain imaging. [See comments] Nature 382: 66–69.
Everling, S., Munoz, D. P. (2000). Neuronal correlates for preparatory set associated with pro-
saccades and anti-saccades in the primate frontal eye field. J Neurosci 20: 387–400.
65 Cognitive development: fMRI studies
Everling, S., Dorris, M. C., Klein, R. M., Munoz, D. P. (1999). Role of primate superior colliculus
in preparation and execution of anti-saccades and pro-saccades. J Neurosci 19: 2740–2754.
Fischer, B., Biscaldi M, Gezeck, S. (1997). On the development of voluntary and reflexive com-
ponents in human saccade generation. Brain Res 754: 285–297.
Foong, J., Maier, M., Clark, C. A., et al. (2000). Neuropathological abnormalities of the corpus
callosum in schizophrenia: a diffusion tensor imaging study. J Neurol Neurosurg Psychiatry
68: 242–244.
Frank, L. R., Buxton, R. B., Wong, E. C. (2001). Estimation of respiration-induced noise fluctu-
ations from undersampled multislice fMRI data. Magn Reson Med 45: 635–644.
Fry, A. F., Hale, S. (1996). Processing speed, working memory, and fluid intelligence: evidence
for a developmental cascade. Psychol Sci 7: 237–241.
Fuster, J. M. (1997). The Prefrontal Cortex. New York: Raven Press.
Gaillard, W. D., Hertz-Pannier, L., Mott, S. H., et al. (2000). Functional anatomy of cognitive
development: fMRI of verbal fluency in children and adults. Neurology 54: 180–185.
Gaillard, W. D., Pugliese, M., Grandin, C. B., et al. (2001a). Cortical localization of reading in
normal children: an fMRI language study. Neurology 57: 47–54.
Gaillard, W. D., Grandin, C. B., Xu, B. (2001b). Developmental aspects of pediatric fMRI: con-
siderations for image acquisition, analysis, and interpretation. NeuroImage 13: 239–249.
Georgiewa, P., Rzanny, R., Gaser, C., et al. (2002). Phonological processing in dyslexic children:
a study combining functional imaging and event related potentials. Neurosci Lett 318: 5–8.
Giedd, J. N., Snell, J. W., Lange, N., et al. (1996). Quantitative magnetic resonance imaging of
human brain development: ages 4–18. Cereb Cortex 6: 551–560.
Giedd, J. N., Blumenthal, J., Jeffries, N. O., et al. (1999). Brain development during childhood
and adolescence: a longitudinal MRI study. Nat Neurosci 2: 861–863.
Goldman-Rakic, P. S. (1988). Topography of cognition: parallel distributed networks in primate
association cortex. Annu Rev Neurosci 11: 137–156.
(1990). Parallel systems in the cerebral cortex: the topography of cognition. In Natural and
Artificial Parallel Computation, ed. M. A. Arbib, J. A. Robinson. New York: MIT Press,
pp. 155–176.
(1992). Working memory and the mind. Sci Am 267: 111–117.
Gottesman, I. I., Shields, J., Hanson, D. (1982). Schizophrenia: The Epigenetic Puzzle. New York:
Cambridge University Press.
Hertz-Pannier, L., Gaillard, W. D., Mott, S. H., et al. (1997). Noninvasive assessment of language
dominance in children and adolescents with functional MRI: a preliminary study. Neurology
48: 1003–1012.
Hertz-Pannier, L., Chiron, C., Vera, P., van de Morteele, P. F., et al. (2001). Functional imaging
in the work-up of childhood epilepsy. Childs Nerv Syst 17: 223–228.
Holland, S. K., Plante, E., Byars, A. W., et al. (2001). Normal fMRI brain activation patterns in
children performing a verb generation task. NeuroImage 14: 837–843.
Huttenlocher, P. R. (1990). Morphometric study of human cerebral cortex development.
Neuropsychologia 28: 517–527.
Hyde, T. M., Ziegler, J. C., Weinberger, D. R. (1992). Psychiatric disturbances in metachromatic
leukodystrophy. Arch Neurol 49: 401–406.
66 B. Luna and J. A. Sweeney
Jernigan, T. L., Trauner, D. A., Hesselink, J. R., Tallal, P. A. (1991). Maturation of human cerebrum
observed in vivo during adolescence. Brain 114: 2037–2049.
Kemna, L. J., Posse, S. (2001). Effect of respiratory CO2 changes on the temporal dynamics of the
hemodynamic response in functional MR imaging. NeuroImage 14: 642–649.
Keshavan, M. S., Kapur, S., Pettegrew, J. W. (1991). Magnetic resonance spectroscopy in psychi-
atry: potential, pitfalls, and promise. Am J Psychiatry 148: 976–985.
Keshavan, M. S., Anderson, S., Pettegrew, J. W. (1994). Is schizophrenia due to excessive synaptic
pruning in the prefrontal cortex? The Feinberg hypothesis revisited. J Psychiatr Res 28:
239–265.
Keshavan, M., Diwadkar, V., Spencer, S. M., et al. (2002). A preliminary functional magnetic
resonance imaging study in offspring of schizophrenic parents. Prog Neuropsychopharmacol
Biol Psychiatry 26: 1143–1149.
Kim, S. G., Ugurbil, K., Strick, P. L. (1994). Activation of a cerebellar output nucleus during
cognitive processing. Science 265: 949–951.
Klein, C., Heinks, T., Andresen, B., Berg, P., Moritz, S. (2000). Impaired modulation of the saccadic
contingent negative variation preceding antisaccades in schizophrenia. Biol Psychiatry 47:
978–990.
Klingberg, T., Vaidya, C. J., Gabrieli, J. D. E., Moseley, M. E., Hedehus, M. (1999). Myelination
and organization of the frontal white matter in children: a diffusion tensor MRI study.
Neuroreport 10: 2817–2821.
Klingberg, T., Hedehus, M., Temple, E., et al. (2000). Microstructure of temporo-parietal white
matter as a basis for reading ability: evidence from diffusion tensor magnetic resonance
imaging. Neuron 25: 493–500.
Klingberg, T., Forssberg, H., Westerberg, H. (2002). Increased brain activity in frontal and pari-
etal cortex underlies the development of visuospatial working memory capacity during
childhood. J Cogn Neurosci 14: 1–10.
Kwon, H., Reiss, R. L., Menon, V. (2002). Neural basis of protracted developmental changes in
visuo-spatial working memory. Proc Natl Acad Sci USA 99: 13336–13341.
Le Bihan, D., Mangin, J. F., Poupon, C., et al. (2001). Diffusion tensor imaging: concepts and
applications. J Magn Reson 13: 534–546.
Levin, H. S., Culhane, K. A., Hartmann, J., Evankovich, K., Mattson, A. J. (1991). Developmental
changes in performance on tests of purported frontal lobe functioning. Dev Neuropsychol
7: 377–395.
Lim, K. O., Hedehus, M., Moseley, M., et al. (1999). Compromised white matter tract integrity
in schizophrenia inferred from diffusion tensor imaging. Arch Gen Psychiatry 56: 367–374.
Logan, W. J. (1999). Functional magnetic resonance imaging in children. Semin Pediatr Neurol
6: 78–86.
Luciana, M., Nelson, C. A. (1998). The functional emergence of prefrontally-guided working
memory systems in four- to eight-year-old children. Neuropsychologia 36: 273–293.
Luna, B., Sweeney, J. A. (1999). Cognitive functional magnetic resonance imaging at very-high-
field: eye movement control. Top Magn Reson Imaging 10: 3–15.
(2001). Studies of brain and cognitive maturation through childhood and adolescence: a
strategy for testing neurodevelopmental hypotheses. Schizophr Bull 27: 443–455.
67 Cognitive development: fMRI studies
Luna, B., Thulborn, K. R., Munoz, D. P., et al. (2001). Maturation of widely distributed brain
function subserves cognitive development. NeuroImage 13: 786–793.
Luna, B., Minshew, N. J., Garver, K. E., et al. (2002). Neocortical system abnormalities in autism:
an fMRI study of spatial working memory. Neurology 59: 834–840.
McDowell, J. E., Clementz, B. A. (2001). Behavioral and brain imaging studies of saccadic per-
formance in schizophrenia. Biol Psychol 57: 5–22.
Merriam, E. P., Thase, M. E., Haas, G. L., Keshavan, M. S., Sweeney, J. A. (1999). Prefrontal cortical
dysfunction in depression determined by Wisconsin Card Sorting Test performance. Am J
Psychiatry 156: 780–782.
Munoz, D. P., Broughton, J. R., Goldring, J. E., Armstrong, I. T. (1998). Age-related performance
of human subjects on saccadic eye movement tasks. Exp Brain Res 217: 1–10.
Nelson, C. A., Monk, C. S., Lin, J., et al. (2000). Functional neuroanatomy of spatial working
memory in children. Dev Psychol 36: 109–116.
Park, S., Holzman, P. S. (1992). Schizophrenics show spatial working memory deficits. Arch Gen
Psychiatry 49: 975–982.
Peled, S., Yeshurun, Y. (2001). Superresolution in MRI: application to human white matter fiber
tract visualization by diffusion tensor imaging. Magn Reson Med 42: 29–35.
Pettegrew, J. W., Keshavan, M. S., Minshew, N. J. (1993). 31 P nuclear magnetic resonance spec-
troscopy: neurodevelopment and schizophrenia. Schizophr Bull 19: 35–53.
Pfefferbaum, A., Mathalon, D. H., Sullivan, E. V., et al. (1994). A quantitative magnetic resonance
imaging study of changes in brain morphology from infancy to late adulthood. Arch Neurol
51: 874–887.
Poldrack, R. A., Pare-Blagoev, E. J., Grant, P. E. (2002). Pediatric functional magnetic resonance
imaging: progress and challenges. Top Magn Reson Imaging 13: 61–70.
Raj, D., Anderson, A. W., Gore, J. C. (2001). Respiratory effects in human functional magnetic
resonance imaging due to bulk susceptibility changes. Phys Med Biol 46: 3331–3340.
Rakic, P., Bourgeois, J. P., Eckenhoff, M. F., Zecevic N, Goldman-Rakic, P. S. (1986). Concurrent
overproduction of synapses in diverse regions of the primate cerebral cortex. Science 232:
232–235.
Reiss, A. L., Abrams, M. T., Singer, H. S., Ross, J. L., Denckla, M. B. (1996). Brain development,
gender, and IQ in children: a volumetric imaging study. Brain 119: 1763–1774.
Ridderinkhof, K. R., van der Molen, M. W. (1997). Mental resources, processing speed, and
inhibitory control: a developmental perspective. Biol Psychol 45: 241–261.
Rosenberg, D. R., Sweeney, J. A., Gillen, J., et al. (1997). Magnetic resonance imaging of children
without sedation: preparation with simulation. J Am Acad Child Adolesc Psychiatry 36: 853–
859.
Rubia, K., Overmeyer S, Taylor, E., et al. (2000). Functional frontalisation with age: mapping
neurodevelopmental trajectories with fMRI. Neurosci Biobehav Rev 24: 13–19.
Schlaggar, B. L., Brown, T. T., Lugar, H. M., et al. (2002). Functional neuroanatomical differences
between adults and school-age children in the processing of single words. Science 296: 1476–
1479.
Sereno, A. B., Holzman, P. S. (1995). Antisaccades and smooth pursuit eye movements in
schizophrenia. Biol Psychiatry 37: 394–401.
68 B. Luna and J. A. Sweeney
Shaywitz, B. A., Shaywitz, S. E., Pugh, K. R., et al. (2002). Disruption of posterior brain systems
for reading in children with developmental dyslexia. Biol Psychiatry 52: 101–110.
Slifer, K. J. (1996). A video system to help children cooperate with motion control for radiation
treatment without sedation. J Pediatr Oncol Nurs 13: 91–97.
Stanley, J. A., Williamson, P. C., Drost, D. J., et al. (1996). An in vivo proton magnetic resonance
spectroscopy study of schizophrenia patients. Schizophr Bull 22: 597–609.
Stapleton, S. R., Kiriakopoulos, E., Mikulis, D., et al. (1997). Combined utility of functional MRI,
cortical mapping, and frameless stereotaxy in the resection of lesions in eloquent areas of
brain in children. Pediatr Neurosurg 26: 68–82.
Szameitat, A. J., Schubert, T., Mueller, K., von Cramon, D. Y. (2002). Localization of executive
functions in dual-task performance with fMRI. J Cogn Neurosci 14: 1184–1199.
Talairach, J., Tournoux, P. (1988). Co-Planar Stereotaxic Atlas of the Human Brain. New York:
Thieme Medical.
Temple, E., Poldrack, R. A., Salidis, J., et al. (2001). Disrupted neural responses to phonological
and orthographic processing in dyslexic children: an fMRI study. Neuroreport 12: 299–307.
Thatcher, R. W. (1991). Maturation of the human frontal lobes: physiological evidence for staging.
Dev Neuropsychol 7: 397–419.
Thomas, K. M., King, S. W., Franzen, P. L., et al. (1999). A developmental functional MRI study
of spatial working memory. NeuroImage 10: 327–338.
Thulborn, K. R., Shen, G. X. (1999). An integrated head immobilization system and high-
performance RF coil for fMRI of visual paradigms at 1.5 T. J Magn Reson 139: 26–34.
Vaidya, C. J., Austin, G., Kirkorian, G., et al. (1998). Selective effects of methylphenidate in
attention deficit hyperactivity disorder: a functional magnetic resonance study. Proc Natl
Acad Sci USA 95: 14494–14499.
Waddington, J. L., Torrey, E. F., Crow, T. J., Hirsch, S. R. (1991). Schizophrenia, neurodevelopment
and disease. Arch Gen Psychiatry 48: 271–273.
Weinberger, D. R., Lipska, B. K. (1995). Cortical maldevelopment, anti-psychotic drugs, and
schizophrenia: a search for common ground. Schizophr Res 16: 87–110.
Weinberger, D. R., Berman, K. F., Zec, R. F. (1986). Physiologic dysfunction of dorsolateral pre-
frontal cortex in schizophrenia: I. Regional cerebral blood flow evidence. Arch Gen Psychiatry
43: 114–124.
Welsh, M. C., Pennington, B. F., Groisser, D. B. (1991). A normative–developmental study of
executive function: a window on prefrontal function in children. Dev Neuropsychol 7: 131–
149.
Wilson, S. P., Kipp, K. (1998). The development of efficient inhibition: evidence from directed-
forgetting tasks. Dev Rev 18: 86–123.
Woods, R. P., Mazziotta, J. C., Cherry, S. R. (1993). Automated image registration. In Quantifi-
cation of Brain Function: Tracer Kinetics and Image Analysis in Brain PET, ed. K. Uemura,
N. A. Lassen, T. Jones, et al. Amsterdam: Elsevier Science, pp. 391–400.
Yakovlev P. I., Lecours A. R. (1967). Regional Development of the Brain in Early Life. Oxford:
Blackwell Scientific, pp. 3–70.
4
Children are not born with the full complement of cognitive skills they need for
adulthood; instead, they need to develop these abilities as they grow. Initial theor-
ies about this development espoused a behaviorist idea of the child as a passive
participant shaped by external forces, with more recent views considering the child
as developing through invariant and universal cognitive stages in an active quest to
make sense of their experience (Piaget, 1965). In this scheme, adolescence heralded
the final developmental progression in cognitive development, with the transition
from concrete to formal operational thought. This capacity to deal with abstract
concepts, “operating on operations” rather than reality, what is now referred to as
meta-cognition, or encompassed in the notion of executive functions, was consid-
ered to become available to the child around the age of 11 or 12 years (Stuss, 1992;
Travis, 1998).
Much of the detail of the influential cognitive model proposed by Piaget and
his proponents has now been amended and extended. Nevertheless, the notion
of a sequential and gradual accumulation of cognitive processes, along with the
limitations and capabilities of the child as they mature, has been invaluable to
the study of child development. An understanding of the normal development of
cognitive function is important in order to appreciate the pattern of impairments in
children and adolescents who suffer from illnesses of putative neurodevelopmental
origin. In particular, we need to know when cognitive functions are due to come “on-
line” during maturation, and how this differs across cognitive domains. Information
on the normal progression of skill acquisition allows for better characterization of
whether the onset of behavioral disturbances or symptoms of psychiatric or other
disorders reflect a deviation in functioning or a failure to make age-appropriate
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
69
70 S. J. Wood, C. R. De Luca, V. Anderson, and C. Pantelis
gains. In this regard, the observation that the acute symptoms of schizophrenia
arise during this final phase of cognitive transition raises interesting questions
about the etiology of the disorder and how the underlying substrates of psychosis
are expressed.
Adolescence
Adolescence is a loose descriptive term referring to the period of transition from
childhood to adulthood. Although it comprises the teenage years and is heralded
by the onset of puberty, it is not defined by specific events. Indeed, while puberty
and adolescence are often used synonymously (Petersen, 1998), the two do not
necessarily co-occur (Dubas, 1991). For the purposes of this review, adolescence
will be taken to cover the years from ages 12 to 20, as distinct cognitive advances
in higher-order thinking are seen to occur around the age of 12, with a general
stabilization of most executive processes as individuals enter their twenties.
Adolescence is a time of major upheaval in behavioral and social domains as
the developing child seeks to attain the skills necessary for independence. The
major changes tend to be in the areas of social interaction, risk-taking behavior,
and intellectual expansion. The first two are more comprehensively reviewed by
Spear (2000), while the last is covered in detail. The greater emphasis placed on
exploring cognitive developments unique to adolescence derives from the belief that,
ultimately, the emergence of independent thinking, social and moral awareness,
impulsivity, and challenging of conceptual ideas are a reflection of the higher-order
executive abilities that become available to the child during the teenage years.
Cognitive changes
Cognition generally refers to “knowing” or “thinking” and includes a broad range
of abilities over and above what is thought of as intelligence. These include memory,
attention, executive functions, plus less-obvious skills such as appreciating humour
and perceiving another’s motivation: skills that are often deficient in individuals
with neurodevelopmental disorders such as schizophrenia. Basic cognitive pro-
cesses are in place by early childhood, and it has been shown that certain abilities
are inborn (Meltzoff and Moore, 1977). However, more complex behaviors develop
at a slower rate and some functions are not mastered until late in adulthood. This
holds implications for the observation of deficits in areas of higher-order func-
tions as individuals enter the adolescent period. Studies of cognitive development
tend to have proceeded with little attention to studies of brain development; how-
ever, given that the predominant brain regions developing during adolescence are
the frontal lobes, it is sensible to examine the cognitive development of functions
thought to be mediated by these regions. The frontal lobes have been given par-
ticular attention in the mediation of executive functions, that is, those interrelated
skills required for successful independent, volitional behavior in response to novelty
(Anderson, 1998). The functions subsumed under the executive umbrella include
higher-order processes such as strategic planning, problem solving, inhibitory con-
trol, cognitive flexibility, abstract thinking, concept formation, working memory,
and self-monitoring.
Cerebral organization and cognitive functioning models initially presented
executive function as an adult capacity that reaches maturity during adolescence,
approximating the stage of Piaget’s predicted transition from concrete to formal
operational thinking (Golden, 1981; Stuss, 1992; Travis, 1998). Research has now
identified a stage-like sequence of executive function development characterized by
“spurts” in executive abilities beginning from as young as 12 months of age, with
the majority of functions coming “online” around the age of 8 (Ardila and Rosselli,
1994; Case, 1992; Luciana and Nelson, 1998). These higher-order processes are
73 Cognitive development in adolescence
adolescence and is at its height between the ages of 20 and 29 years. Success on these
tasks is thought to result at least in part from the achievement and maintenance of
maximal short-term memory capacity in the 15–19 age range, since maturation of
this “capacity resource” contributes to a greater ability to define and order a strategic
plan (Hitch et al., 2001). However, while increases in abilities such as memory span,
inhibitory control, and speed of processing may allow for more efficient executive
control, advances in the last have been dissociated from the maturation of these
types of general, capacity-limited factor (Ridderinkhof and van der Molen, 1997).
In our study, attentional set-shifting ability was also tested using the intradimen-
sional/extradimensional task from the CANTAB, which assesses the ability of an
individual to establish the correct response set to positive and negative feedback, to
reverse this rule, and later to shift this learning to new exemplars within a previously
learned set. This measure of cognitive flexibility was found to follow a swifter mat-
urational path, and performance on this task did not differentiate any of the groups
(De Luca et al., 2003), confirming documented adult levels of set-shifting compet-
ence in 8 and 10 year old children (Anderson et al., 2001a; Chelune and Baer, 1986;
Luciana and Nelson, 1998). This would suggest that the ability to adjust responses
to feedback, and to shift them to new exemplars within a previously learned set,
is an executive skill acquired early in development, indicating that the purported
neural circuitry subserving this ability in the lateral and orbital prefrontal cortex
is connected and activated earlier than other executive systems (Rezai et al., 1993;
Roberts et al., 1998; Stuss and Benson, 1987). However, our use of an endpoint
score for this task may have obscured subtle differences in performance. Previous
research would suggest that children do in fact make a greater number of errors in
learning a response set, but they do not differ in the number of trials necessary to
advance at either level (Luciana and Nelson, 1998; Shanab and Yasin, 1979). Con-
sequently, they can competently complete such tasks by responding to the absolute
properties of the stimuli without relevant understanding of the underlying contin-
gency being tested. Adolescents and adults, by comparison, are found to experience
greater difficulty performing a shift in set because of their more profound under-
standing of the dimension characteristics of the task. Having learned the relevant
dimension governing the response rule, their performance is potentially impeded
as they are required to extinguish the original response set and transfer this learning
to new exemplars (Owen et al., 1993; Whitney and White, 1993). Indeed, patients
with schizophrenia are seen to perseverate on previously learned sets and perform
poorly on similar tests such as the Wisconsin Card Sorting Task. Their inability
to dissociate from one paradigm and shift to a new exemplar highlights deficits in
attentional control and, therefore, implicates dysfunction in the lateral and orbital
frontal systems that mediate this function. It is interesting to note that this skill
appears to be relatively unimpaired in first-episode schizophrenia, suggesting that
75 Cognitive development in adolescence
these brain areas, which develop earlier in life, are in fact well connected and
activated appropriately during development but may be affected later by the con-
tinued insult of acute psychotic episodes.
While the set-shifting component of the attentional system appears to develop
relatively early in childhood, the literature suggests that other attentional processes,
such as the ability to attend to meaningful stimuli and actively inhibit distractors,
continue to improve during adolescence (Anderson et al., 2001a). Greater control
in the triage of incoming information with internal needs and desires places less
demand on the capacity-limited attentional system, which, in turn, results in the
more efficient processing of relevant information. While attentional components
are found to dissociate both anatomically and in terms of their maturational course,
gradual increments in performance from 6 to 15 years of age have been found for
a range of these skills across modalities (Anderson et al., 2001a).
As is the case with the development of executive abilities, the prolonged trajectory
of attentional skills implies that deficits may not be obvious until later in devel-
opment. Within a developmental framework, the cumulative effects of attention
deficits, such as distractibility or the inability to shift-set, may have serious implica-
tions for the learning of new information and coordination of internal desires with
external demands at a time when these skills appear to be critical to the success of
the individual. In addition, impaired executive function will affect the adolescent’s
ability to plan homework or find classrooms, resulting in poorer scholastic perfor-
mance over and above its effect on actual learning. These practical consequences
highlight the dynamic interplay that exists between cognitive and psychological
factors, and how cognitive inefficiencies that arise during adolescence, already a
time of considerable emotional upheaval, feed feelings of personal inadequacy and
disrupt the usual process of self-discovery. It is not surprising then that the current
drive in the treatment of schizophrenia is to treat aggressively first-episode patients,
both to prevent the purported neurotoxic effects of repeated acute episodes and to
halt the snowballing effects of chronic illness and perceived personal failure.
adolescent period are thought to reflect maturation of the connections in this func-
tional system. Myelination of the cingulate gyrus and prefrontal cortex does not
fully occur until at least the second decade of life (Giedd et al., 1999), and these
are key areas involved in the integration of emotional/biological information with
intentional, cognitively driven plans (Benes, 1989; Paus et al., 2001). Conversely,
reductions in the gray matter of the parietal and frontal cortices during late ado-
lescence are considered a marker of the refinement of these anterior–posterior
connections (Giedd et al., 1999; Sowell et al., 1999).
Functional magnetic resonance imaging (fMRI) studies looking at development
of selective attention and response inhibition skills have reported variable find-
ings, with both hypo- and hyperactivation found in these brain regions in children
and adolescents compared with adults (see Ch. 3). A steady increase in activation
of frontal, parietal, cingulate, striatal, and thalamic areas has been shown from
childhood to adulthood for tasks assessing response inhibition (Luna and Sweeney,
2001). These studies suggested that the power of activation corresponds to the
extent of functional maturation and, therefore, voluntary control over one’s actions
(Kwon et al., 2002; Rubia et al., 2000). In contrast, other fMRI studies have indi-
cated that essentially the same circuits are activated in children and adults, although
the activation is greater in children, both for the volume of cortex engaged to per-
form the task and the percentage signal change (Casey et al., 2000). These authors
argue that, taken together, the decrease in structural and functional areas mediating
attentional processes suggests that improvements in these skills during adolescence
correspond to anatomical maturation of frontal and parietal areas. These physio-
logical processes are thought to strengthen and enhance stable connections, leading
to a decrease in the brain tissue required to perform a task and removal of compet-
ing inputs that interfere with direct communication between these interconnected
regions (Klingberg et al., 2002). It is specifically these later maturing systems that
are found to be dysfunctional in both first-episode and chronic schizophrenia; they
will be discussed in greater detail in the section below dealing with schizophrenia.
Developmental lesions
Research on the impact of developmental lesions supports the protracted and dis-
tinct consequences of disruption to the developing neural system. Of the limited
studies examining the outcome of frontal lesions in the young child, pervasive
behavioral and social deficits have been reported that are of a similar nature to
those seen in patients with schizophrenia. In line with the developmental perspec-
tive of frontal maturation, these deficits do not arise immediately following damage
in all cases but become progressively more evident as the child ages (Eslinger et al.,
1992; Marlowe, 1992). Many of the features are comparable to those described in
adult patients, such as a lack of initiative, disorganization, irritability, emotional
lability, and disinhibited behavior (Anderson et al., 1999; Eslinger and Damasio,
1985; Eslinger et al., 1992; Leduc et al., 1999). However, the severity of impairment
appears to be greater in individuals who acquired lesions in childhood, and a dis-
tinct antisocial presentation is reported in studies with adult follow-up (Price et al.,
1990). This is demonstrated as a failure to learn from experience, unresponsiveness
to punishment, increasing disruptive and demanding behavior, a lack of expressed
guilt and remorse, with some displaying verbal and physically abusive tendencies
(Eslinger and Damasio, 1985; Eslinger et al., 1992; Marlowe, 1992). The distinctive
feature that sets these individuals apart from sociopaths is their rather impulsive
and child-like performance of morally sanctioned acts, and the transparency with
which they attempt to cover up their indiscretions (Anderson et al., 1999). This
is interpreted as a reflection of the lack of insight and immature development of
79 Cognitive development in adolescence
executive skills in these individuals. It appears that damage to the anterior lobes
early in life disrupts the application of socially appropriate skills in real-life situa-
tions and compromises the establishment of adaptive behavior, the acquisition of
socially relevant knowledge, and the development of moral sensibilities (Anderson
et al., 1999; Eslinger et al., 1992).
Studies addressing the cognitive development of children with early lesions sup-
port the notion that these individuals experience more global deficits because they
fail to develop in a normal manner. In one of the first studies to look specifically
at cognitive outcomes of children with early brain insult, Mateer (1990) found
evidence of cognitive dysfunction, alongside intact or mildly depressed intellec-
tual ability, consistent with adult patterns of impairment. A number of more recent
studies have revealed a similar pattern of poor problem-solving skills, reduced plan-
ning and strategy use, and low self-monitoring in the adult survivors of early frontal
lobe damage (Anderson, 1988; Anderson and Jacobs, 2004; Anderson et al., 1999,
2000; Eslinger et al., 1992, 1997, 1999; Marlowe, 1992). Eslinger and colleagues
(1992) reported a pattern of delayed onset of impairments, with increasing diffi-
culties identified as executive skills failed to “come online” and mature at critical
stages throughout development. The progressive emergence of deficits as the child
reaches adolescence is believed to result from the combined effects of the failure to
make age-appropriate gains and the increasingly complex environmental demands
placed on the individual approaching adulthood. Interestingly, similar impairments
in both behavioral and cognitive domains have been described in many purported
neurodevelopmental and acquired pediatric conditions, including autism (Happe
and Frith, 1996), schizophrenia (Pantelis et al., 2002), attention-deficit hyperac-
tivity disorder (Barkley, 1996), head injury (Pentland et al., 1998), and epilepsy
(Anderson et al., 2001b).
Schizophrenia
Schizophrenia is a mental illness of putative neurodevelopmental origin (Murray
and Lewis, 1987; Weinberger, 1987) that represents an example of aberrant develop-
ment in the regions underpinning executive function. Because of this, we can use it as
a model to look at changes in cognitive performance over childhood/adolescence to
help to understand the neural development of brain regions mediating these skills.
One example of this is the retrospective study of Walker and colleagues (1994),
who used family home movies to study the neurodevelopment of children who
later developed schizophrenia. Although these movies demonstrated subtle abnor-
malities in motor function in early life, by the time of illness onset they were much
less prominent; instead these young people displayed disturbances of behavior and
cognitive function. This led the authors to propose a modular developmental tra-
jectory for schizophrenia, in which neuromotor dysfunction is most pronounced
80 S. J. Wood, C. R. De Luca, V. Anderson, and C. Pantelis
50
45
35
30
25
20
15
10
0
8 to 10 11 to 14 15 to 19 20 to 29 30 to 49 50 to 64
Age group
Fig. 4.1. Total number of working memory errors as a function of age group for males () and
females ( ). •
a rule that had been learned (representing failure at a simpler level of set-shifting
ability). We have now examined patients very early in the course of the illness (mean
illness duration of 95 days) and, despite obvious working memory difficulties,
set-shifting ability is less impaired (Mahony et al., 2001). Although there are no
published longitudinal studies available using this task, these findings suggest that
there is no or minimal impairment of set-shifting at first presentation, with a
possible slow decline over continuing illness.
Working memory, a late emerging aspect of executive functions, is clearly
impaired in patients with established illness (Pantelis et al., 1997) and also at the
first presentation of the disorder (Hutton et al., 1998; Proffitt, 2001), with no pro-
gression over time (Proffitt, 2001). In addition, there are deficits in young people
at ultra-high risk for psychosis prior to the onset of illness (Wood et al., 2003),
indicating that this ability has never been fully functional. Taken together with
the relatively late development of spatial working memory in normal individuals
(De Luca et al., 2003), these findings suggest that functions which normally develop
during the at-risk period for the development of psychosis manifest the most severe
deficits.
An important aspect of the maturational changes in the expressed sympto-
matology of schizophrenia is the highly contested argument of whether impair-
ments in cognitive functioning that present at the onset of psychosis represent a
82 S. J. Wood, C. R. De Luca, V. Anderson, and C. Pantelis
REFERENCES
Anderson, S. W., Bechara, A., Damasio, H., Tranel, D., Damasio, A. R. (1999). Impairment of
social and moral behavior related to early damage in human prefrontal cortex. Nat Neurosci
2: 1032–1037.
Anderson, S. W., Damasio, H., Tranel, D., Damasio, A. R. (2000). Long-term sequelae of prefrontal
cortex damage acquired in early childhood. Dev Neuropsychol 18: 281–296.
Anderson, V. (1988). Recovery of function in children: the myth of cerebral plasticity. In Brain
Impairment. Proceedings from the Thirteenth Annual Brain Impairment Conference, ed. M.
Matheson, H. Newman. Sydney: University of Sydney, pp. 223–247.
(1998). Assessing executive function in children: biological, psychological and developmental
considerations. Neuropsychol Rehab 8: 319–349.
Anderson, V., Jacobs, R. (2004). Frontal lobe damage in children: interruptions to normal devel-
opment. In Enfance and Neuropsychologie: Interface entre la Recherche et la Clinique, ed. J.-P.
Laurent. Quebec: Quebec University Press, in press.
84 S. J. Wood, C. R. De Luca, V. Anderson, and C. Pantelis
Anderson, V., Lajoie, G., Bell, R. (1995). Neuropsychological Assessment of the School-aged Child.
Department of Psychology, Melbourne: University of Melbourne.
Anderson, V. A., Anderson, P., Northam, E., Jacobs, R., Catroppa, C. (2001a). Development of
executive functions through late childhood and adolescence in an Australian sample. Dev
Neuropsychol 20: 385–406.
Anderson, V., Northam, E., Hendy, J., Wrennall, J. (2001b). Developmental Neuropsychology.
Hove, UK: Psychology Press.
Ardila, A., Rosselli, M. (1994). Development of language, memory and visuospatial abilities in
5- to 12-year-old children using a neuropsychological battery. Dev Neuropsychol 10:
97–120.
Baddeley, A. (1998). Recent developments in working memory. Curr Opin Neurol 8: 234–238.
Barkley, R. A. (1996). Linkages between attention and executive functions. In Attention, Memory
and Executive Function, ed. G. R. Lyon, A. Krasnegor. Baltimore, MD: Paul H. Brookes, pp.
307–326.
Bedwell, J. S., Keller, K., Smith, A. K., et al. (1999). Why does postpsychotic IQ decline in
childhood-onset schizophrenia? Am J Psychiatry 156: 1996–1997.
Benes, F. M. (1989). Myelination of cortical–hippocampal relays during late adolescence.
Schizophr Bull 15: 585–593.
Benes, F. M., Turtle, M., Khan, Y., Farol, P. (1994). Myelination of a key relay zone in the hippo-
campal formation occurs in the human brain during childhood, adolescence and adulthood.
Arch Gen Psychiatry 51: 477–484.
Case, R. (1992). The role of the frontal lobes in the regulation of cognitive development. Brain
Cognit 20: 51–73.
Casey, B. J., Giedd, J. N., Thomas, K. M. (2000). Structural and functional brain development
and its relation to cognitive development. Biol Psychiatry 54: 241–257.
Chelune, G. J., Baer, R. (1986). Developmental norms of the Wisconsin Card Sorting Test. J Clin
Exp Neuropsychol 8: 219–228.
Chugani, H., Phelps, M., Mazziotta, J. (1987). Positron emission tomography study of human
brain functional development. Ann Neurol 22: 487–497.
De Luca, C. R., Wood, S. J., Anderson, V., et al. (2003). Normative data from the CANTAB.
I: Development of executive function over the lifespan. J Clin Exp Neuropsychol 25:
242–254.
Dubas, J. (1991). Cognitive abilities and physical maturation. In Encyclopedia of Adolescence,
Vol. 1, ed. R. Lerner et al. New York: Garland Publishing, pp. 133–138.
Elliott, R., Sahakian, B. J. (1995). The neuropsychology of schizophrenia: relations with clinical
and neurobiological dimensions. Psychol Med 25: 581–594.
Eslinger, P. J., Damasio, A. R. (1985). Severe disturbance of higher cognition after bilateral frontal
lobe ablation: Patient EVR. Neurology 35: 1731–1741.
Eslinger, P. J., Grattan, L. M., Damasio, H., Damasio, A. R. (1992). Developmental consequences
of childhood frontal lobe damage. Arch Neurol 49: 764–769.
Eslinger, P., Biddle, K., Grattan, L. (1997). Cognitive and social development in children with
prefrontal cortex lesions. In Development of the Prefrontal Cortex: Evolution, Neurology, and
Behavior, ed. P. S. Goldman-Rakic. Baltimore, MD: Brookes, pp. 295–336.
85 Cognitive development in adolescence
Eslinger, P., Biddle, K., Pennington, B., Page, R. (1999). Cognitive and behavioral development
up to 4 years after early right frontal lobe lesion. Dev Neuropsychol 15: 157–191.
Espy, K. (1997). The shape school: assessing executive function in preschool children. Dev
Neuropsychol 13: 495–499.
Giedd, J. N., Blumenthal, J., Jeffries, N. O., et al. (1999). Brain development during childhood
and adolescence: a longitudinal MRI study. Nat Neurosci 2: 861–863.
Golden, C. (1981). Luria Nebraska Children’s Battery: theory and formulation. In Neuropsycho-
logical Assessment of the School-aged Child, ed. G. W. Hynd, J. E. Obzut. London: Grune and
Stratton, pp. 277–302.
Goldman-Rakic, P. S. (1988). Topography of cognition: parallel distributed networks in primate
association cortex. Annu Rev Neurosci 11: 137–156.
Happe, F., Frith, U. (1996). The neuropsychology of autism. Brain 119: 1377–1400.
Heilman, K., Valenstein, E., Watson, R. (1997). Disorders of attention. In Contemporary Behavioral
Neurology, ed. M. Trimble, J. Cummings. London: Harcourt, pp. 127–138.
Hitch, G. J. (2002). Developmental changes in working memory: a multicomponent view. In
Lifespan Development of Human Memory, ed. P. Graf, N. Ohta. Cambridge, MA: MIT Press,
pp. 15–38.
Hitch, G. J., Towse, J., Hutton, U. (2001). What limits children’s working memory span? Theo-
retical accounts and applications for scholastic development. J Exp Psychol Gen 130: 184–
198.
Huttenlocher, P. (1984). Synapse elimination and plasticity in developing human cerebral cortex.
Am J Ment Defic 88: 488–496.
Hutton, S. B., Puri, B. K., Duncan, L.-J., et al. (1998). Executive function in first-episode
schizophrenia. Psychol Med 28: 463–473.
Jacobs, R., Northam, E., Anderson, V. (2001). Cognitive outcome in children with myelomeningo-
cele and perinatal hydrocephalus. J Dev Phys Disabil 13: 389–405.
Klingberg, T., Vaidya, C. J., Gabrieli, J. D. E., Moseley, M. E., Hedehus, M. (1999). Myelination
and organization of the frontal white matter in children: a diffusion tensor MRI study.
Neuroreport 10: 2817–2821.
Klingberg, T., Forssberg, H., Westerberg, H. (2002). Increased brain activity in frontal and pari-
etal cortex underlies the development of visuospatial working memory capacity during
childhood. J Cogn Neurosci 14: 1–10.
Koski, L., Petrides, M. (2002). Distractibility after unilateral resections from the frontal and
anterior cingulate cortex in humans. Neuropsychologia 40: 1059–1072.
Kwon, H., Reiss, A. L., Menon, V. (2002). Neural basis of protracted developmental changes in
visuo-spatial working memory. Proc Natl Acad Sci USA 99: 13336–13341.
Leduc, M., Herron, J. E., Greenberg, D. R., Eslinger, P. J., Grattan, L. M. (1999). Impaired awareness
of social and emotional competencies following orbital frontal lobe damage. Brain Cogn 40:
174–177.
Levin, H. S., Culhane, K. A., Hartmann, J., et al. (1991). Developmental changes in performance
on tests of purported frontal lobe functioning. Dev Neuropsychol 7: 377–395.
Lieberman, J. A. (1999). Is schizophrenia a neurodegenerative disorder? A clinical and neurobi-
ological perspective. Biol Psychiatry 46: 729–739.
86 S. J. Wood, C. R. De Luca, V. Anderson, and C. Pantelis
Luciana, M., Nelson, C. A. (1998). The functional emergence of prefrontally guided working
memory systems in four- to eight-year-old children. Neuropsychologia 36: 273–293.
Luna, B., Sweeney, J. A. (2001). Studies of brain and cognitive maturation through childhood
and adolescence: a strategy for testing neurodevelopmental hypotheses. Schizophr Bull 27:
443–455.
Luna, B., Thulborn, K. R., Munoz, D. P., et al. (2001). Maturation of widely distributed brain
function subserves cognitive development. NeuroImage 13: 786–793.
Mahony, K., Pantelis, C., Proffitt, T., et al. (2001). Comparison of set-shifting ability in patients
with first-episode psychosis and chronic schizophrenia. Schizophr Res 49: 114–115.
Manly, T., Anderson, V., Nimmo-Smith, I., et al. (2001). The differential assessment of children’s
attention: The Test of Everyday Attention for Children (TEA-Ch). Normative sample and
ADHD performance. J Child Psychol Psychiatry 42: 1065–1087.
Marlowe, W. B. (1992). The impact of a right prefrontal lesion on the developing brain. Brain
Cogn 20: 205–213.
Mateer, C. A. (1990). Cognitive and behavioral sequalae of face and forehead injury in childhood.
J Clin Exp Neuropsychol 12: 95.
McKay, K., Halperin, J., Schwartz, S., Sharma, V. (1994). Developmental analysis of three aspects of
information processing: sustained attention, selective attention and response organization.
Dev Neuropsychol 10: 121–132.
Meltzoff, A., Moore, M. (1977). Imitation of facial and manual gestures by human neonates.
Science 198: 74–78.
Murray, R. M., Lewis, S. (1987). Is schizophrenia a neurodevelopmental disorder? [Editorial]
Br Med J 295: 681–682.
Nichelli, F., Bulgheroni, S., Riva, D. (2001). Developmental patterns of verbal and visuospatial
spans. Neurol Sci 22: 377–384.
Owen, A. M., Roberts, A., Hodges, J., et al. (1993). Contrasting mechanisms of impaired atten-
tional set-shifting in patients with frontal lobe damage or Parkinson’s disease. Brain 116:
1159–1175.
Pantelis, C., Barnes, T. R., Nelson, H. E., et al. (1997). Frontal-striatal cognitive deficits in patients
with chronic schizophrenia. Brain 120: 1823–1843.
Pantelis, C., Barber, F., Barnes, T. R., et al. (1999). Comparison of set-shifting ability in patients
with chronic schizophrenia and frontal lobe damage. Schizophr Res 37: 251–270.
Pantelis, C., Yücel, M., Wood, S. J., McGorry, P. D., Velakoulis, D. (2001). The timing and
functional consequences of structural brain abnormalities in schizophrenia. NeuroSci News
4: 36–46.
Pantelis, C., Wood, S. J., Maruff, P. (2002). Schizophrenia. In Cognitive Deficits in Brain Disorders,
ed. A. M. Owen, J. Harrison. London: Martin Dunitz, pp. 217–248.
Pantelis, C., Yücel, M., Wood, S. J., McGorry, P. D., Velakoulis, D. (2003). Early and late neurode-
velopmental disturbances in schizophrenia and their functional consequences. Aust NZ J
Psychiatry 37: 399–406.
Paus, T., Zijdenbos, A., Worsley, K., et al. (1999). Structural maturation of neural pathways in
children and adolescents: in vivo study. Science 283: 1908–1911.
87 Cognitive development in adolescence
Paus, T., Collins, D. L., Evans, A. C., et al. (2001). Maturation of white matter in the human brain:
a review of magnetic resonance studies. Brain Res Bull 54: 255–266.
Pentland, L., Todd, J. A., Anderson, V. (1998). The impact of head injury severity on planning
ability in adolescence: a functional analysis. Neuropsychol Rehab 8: 301–317.
Petersen, A. (1998). Adolescence. In Behavioral Medicine and Women: A Comprehensive
Handbook, ed. E. Blechman, K. Brownell. New York: Guilford Press, pp. 45–50.
Piaget, J. (1965). Moral Judgment of the Child. New York: Free Press.
Price, B. H., Daffner, K. R., Stowe, R. M., Mesulam, M. M. (1990). The compartmental learning
disabilities of early frontal lobe damage. Brain 113: 1383–1393.
Proffitt, T. (2001). The stability of spatial working memory and problem solving deficits in first-
episode psychosis and established psychotic illness. Ph.D. Thesis, University of Melbourne,
Melbourne.
Rakic, P., Bourgeois, J.-P., Goldman-Rakic, P. S. (1994). Synaptic development of the cerebral
cortex: implications for learning, memory and mental illness. In Progress in Brain Research,
The Self-organizing Brain: From Growth Cones to Functional Networks, Vol. 102, ed. J. van
Pelt, M. Corner, H. B. Uylings, F. Lopes de Silva. Amsterdam: Elsevier, pp. 227–243.
Rebok, G., Smith, C., Pascualvaca, D., et al. (1997). Developmental changes in attentional per-
formance in urban children from eight to thirteen years. Child Neuropsychol 3: 47–60.
Reiss, A. L., Abrams, M. T., Singer, H. S., Ross, J. L., Denckla, M. B. (1996). Brain development,
gender and IQ in children: a volumetric imaging study. Brain 119: 1763–1774.
Rezai, K., Andreasen, N. C., Alliger, R. J., et al. (1993). The neuropsychology of the prefrontal
cortex. Arch Neurol 50: 636–642.
Ridderinkhof, K., van der Molen, M. (1997). Mental resources, processing speed and inhibitory
control: a developmental perspective. Biol Psychol 45: 241–261.
Roberts, A., Robbins, T. W., Weiskrantz, L. (1998). Discussions and conclusions. In The Prefrontal
Cortex: Executive and Cognitive functions, ed. A. Roberts, T. W. Robbins, L. Weiskrantz.
Oxford: Oxford University Press, pp. 221–242.
Rubia, K., Overmeyer, S., Taylor, E., et al. (2000). Functional frontalisation with age: mapping
neurodevelopmental trajectories with fMRI. Neurosci Biobehav Rev 24: 13–19.
Shanab, M., Yasin, A. (1979). Intradimensional and extradimensional shifts by Jordanian students.
J Gen Psychol 100: 199–213.
Shen, J., Petersen, K. F., Behar, K. L., et al. (1999). Determination of the rate of the glutamate/
glutamine cycle in the human brain by in vivo 13 C NMR. Proc Natl Acad Sci USA 96: 8235–
8340.
Sowell, E. R., Thompson, P. M., Holmes, C., Jernigan, T. L., Toga, A. (1999). In vivo evidence for
post-adolescent brain maturation in frontal and striatal regions. Nat Neurosci 2: 859–861.
Sowell, E. R., Thompson, P. M., Tessner, K. D., Toga, A. (2001a). Mapping continued brain
growth and gray matter density reduction in dorsal frontal cortex: inverse relationships
during postadolescent brain maturation. J Neurosci 21: 8819–8829.
Sowell, E. R., Delis, D., Stiles, J., Jernigan, T. L. (2001b). Improved memory functioning and
frontal lobe maturation between childhood and adolescence: a structural MRI study. J Int
Neuropsychol Soc 7: 312–322.
88 S. J. Wood, C. R. De Luca, V. Anderson, and C. Pantelis
Sowell, E. R., Peterson, B. S., Thompson, P. M., et al. (2003). Mapping cortical change across the
human life span. Nat Neurosci 6: 309–315.
Spear, L. (2000). The adolescent brain and age-related behavioral manifestation. Neurosci
Biobehav Rev 24: 417–463.
Stuss, D. T. (1992). Biological and psychological development of executive functions. Brain Cogn
20: 8–23.
Stuss, D. T., Benson, D. (1987). The frontal lobes and control of cognition and memory. In The
Frontal Lobes Revisited, ed. E. Perecman. New York: Irbn Press, pp. 141–154.
Travis, F. (1998). Cortical and cognitive development in 4th, 8th and 12th grade students: the
contribution of speed of processing and executive function to cognitive development. Biol
Psychol 48: 37–56.
Vaidya, C., Austin, G., Kirkorian, G., et al. (1998). Selective effects of methylphenidate in attention
deficit hyperactivity disorder: a functional magnetic resonance study. Proc Natl Acad Sci 95:
14494–14499.
Velakoulis, D., Wood, S. J., McGorry, P. D., Pantelis, C. (2000). Evidence for progression of brain
structural abnormalities in schizophrenia: beyond the neurodevelopmental model. Aust NZ
J Psychiatry 34(Suppl.): S113–S126.
Walker, E., Savoie, T., Davis, D. (1994). Neuromotor precursors of schizophrenia. Schizophr Bull
20: 441–451.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
Welsh, M. C., Pennington, B. F. (1988). Assessing frontal lobe functioning in children: views from
developmental psychology. Dev Psychol 4: 199–230.
Whitney, L., White, K. (1993). Dimensional shift and the transfer of attention. Q J Exp Psychol
46B: 225–252.
Wood, S. J., Pantelis, C. (2001). Does a neurodevelopmental lesion involving the hippocampus
explain memory dysfunction in schizophrenia? Z Neuropsychol 12: 61–67.
Wood, S. J., Pantelis, C., Proffitt, T., et al. (2003). Spatial working memory ability is a marker of
risk-for-psychosis. Psychol Med 33: 1239–1247.
Yakovlev, P., Lecours, A. (1967). The myelogenetic cycles of regional maturation of the brain. In
Regional Development of the Brain in Early Life, ed. A. Minkowski. Oxford: Blackwell, pp.
3–70.
5
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
89
90 M. Allin, C. Nosarti, L. Rifkin, and R. M. Murray
initially, but their behavior became grossly disturbed once they reached maturity. In
humans, Anderson et al. (1999) studied two young adults who received focal non-
progressive prefrontal damage before 16 months of age. These individuals showed
no behavioral abnormalities up to the age of 3 years but had severely impaired
social behavior despite normal basic cognitive abilities later on in life. Therefore,
it seems that the very plasticity that enables the young brain to overcome injury
so impressively, to reorganize its structure, and spare its essential functions may
have an adverse effect on later development. The neurodevelopmental hypothe-
sis of the etiology of schizophrenia postulates that a brain which is in some way
abnormally “set up” as a result of an early insult may start to exhibit functional
impairments later in life, particularly during the period of brain maturation and
acquisition of adult cognitive capabilities in adolescence and young adulthood
(Allin and Murray, 2002; Murray and Lewis, 1987; Weinberger, 1987). To examine
these issues further, we have been studying a cohort of individuals who were sub-
jected to risk of brain injuries around the time of birth as a result of being born
preterm.
Follow-up
Prospective assessments of neurological and cognitive status of these children were
carried out at 1, 4, and 8 years. At 14–15 years, 163 (97%) individuals were traced,
including 16 who were living abroad. Of the 147 living in the UK, 118 (80%)
agreed to attend for assessment. The cohort members who were unavailable for
investigation did not differ from those studied in birth weight, gestational age
at birth, sex ratio, mode of delivery, condition at birth, the need for mechanical
ventilation, or neonatal cranial ultrasonographic findings. At 18 years, 102 of the
VPT subjects were assessed again.
Can the sequelae of preterm birth still be seen in the brains of adolescents?
The relatively crude imaging resolution provided by ultrasound had demonstrated
a significant amount of brain pathology at birth in the UCHL VPT cohort. However,
some of these abnormalities had apparently resolved at the time of discharge, and the
extent to which the brain damage suffered by these infants resulted in permanent
change in brain structure was unclear. Therefore, at follow-up at 14–15 years,
structural magnetic resonance imaging (MRI) of the brain was performed on all
the participating VPT subjects. Stewart et al. (1999) reported on 72 VPT subjects
and 21 full-term controls. The scans were “blindly” rated according to a structured
format by two neuroradiologists. The scans were classified as normal (no detected
abnormality), equivocal (negligible ventricular dilatation, or negligible thinning
of the corpus callosum, or an isolated white matter hyperintensity), or abnormal
(more than one parenchymal lesion plus definite ventricular dilatation, or definite
thinning or atrophy of the corpus callosum plus reduced white matter or cortical
volume plus multiple areas of white matter signal changes, or intraparenchymal
cysts).
95 Brain plasticity and long-term function
Only 24% of the scans of these VPT adolescents were rated as normal; 21% were
equivocal and 56% abnormal. The equivalent percentages for the controls were 71%
normal, 24% equivocal and 5% abnormal. In those preterm individuals with an MRI
scan classified as abnormal, ventricular dilatation was observed in 80%, posterior
trigonal dilatation in 73%, thinning of the corpus callosum (particularly involving
the splenium) in 65%, abnormal white matter signal in 45%, and decreased white
matter volume in 25%. Ventricular dilatation and thinning of the corpus callosum
were correlated with each other and with other brain lesions and were considered
to be markers of hypoxic–ischemic damage. The white matter signal abnormalities
were thought to reflect scattered patchy gliosis secondary to ischemic damage (Hope
et al., 1988). In addition to these qualitative assessments of MRI scans, the volumes
of the whole brain and of various brain regions and structures were determined
using the Cavalieri stereological method, supported by the MEASURE software
package (Johns Hopkins University, Baltimore, USA). The VPT subjects showed a
6.0% decrease in whole brain volume and a 11.8% decrease in total cortical gray
matter volume. They also had a 42.0% increase in the size of the lateral ventricles.
Those VPT adolescents who had experienced PVH in the neonatal period had a
mean ventricular size twice as large as those who did not, indicating that these
changes were the sequelae of the early damage. Additionally, those who had PVH
and/or ventricular enlargement on neonatal ultrasound were especially likely to
have decreased white matter volumes (Nosarti et al., 2002). This is in agreement with
Kuban et al. (1999), who found that both IVH and ventriculomegaly were powerful
predictors of white matter damage in the first few weeks of life. Nosarti et al. (2002)
also found a positive correlation between white matter volume and gestational age:
the more immature the infant, the greater the damage to the white matter. This
may be a reflection of the developmental vulnerability of white matter between
24 and 32 gestational weeks, when oligodendrocyte progenitors are immature and
vulnerable to hypoxic injury (Back et al., 2001, 2002; du Plessis and Volpe, 2002).
Damage to these cell populations may disrupt the wave of white matter myelination,
which normally rapidly increases after 29 weeks of gestation (Kinney et al., 1988),
leading to a reduction of white matter (and also possibly to an increase in ventricular
volume).
In addition, the volumes of several brain structures were measured. VPT subjects
had reduced volumes of left and right hippocampus, independent of overall brain
volume reduction (Nosarti et al., 2002). These findings are likely to be caused by
hypoxic–ischemic damage (Kuchna, 1994; Mallard et al., 1999). The volume of the
cerebellum was also significantly smaller in VPT adolescents, again independent of
total brain volume (Allin et al., 2001). This may reflect damage to cerebellar granule
cells at a sensitive period in their development, when they are actively dividing and
migrating (Johnston, 1998; Sohma et al., 1995). VPT individuals also had smaller
96 M. Allin, C. Nosarti, L. Rifkin, and R. M. Murray
corpora callosa, particularly in the posterior portion (the splenium) (Nosarti et al.,
2001a).
Neurology
At 1 year of age, 10% of the UCHL cohort had minor neurological impairments
not sufficient to cause disability, and 11% had major impairments with concomi-
tant disability (Stewart et al., 1989). The proportion of children in the cohort who
showed major neurological impairments remained relatively constant at both 4
(15%) and 8 (12%) years, in agreement with other studies (Tin et al., 1997). Ultra-
sound evidence of white matter damage, ventricular dilatation, or hydrocephalus
were powerful predictors of major disability at 1, 4 and 8 years (Costello et al., 1988;
Roth et al., 1993; Stewart et al., 1983).
In contrast to the relative stability of major abnormalities, “minor” abnormalities
increased in prevalence from 10% at age 1 to 23% at 8 years of age (Roth et al.,
1994). By the age of 14 years, 66% of the cohort had an abnormal or equivocal
neurological examination (Stewart et al., 1999). This may be because the range of
motor skills that normal individuals can perform (and which can, therefore, be
tested for) increases with age. An alternative explanation is that lesions of the brain
acquired early in development may have a more significant deleterious effect when
the affected neuronal systems reach developmental maturity, as discussed above
(Goldman, 1971; Sams-Dodd et al., 1997; see Ch. 4).
Neuropsychology
The mean full-scale intelligence quotient (IQ) of the VPT subjects was well within
the normal range at 8 years, indicating that neural plasticity had succeeded in
97 Brain plasticity and long-term function
Behavior
Behavior was assessed at 15 years, using the Rutter behavioral scale (Rutter et al.,
1981) and the social adjustment scale of Cannon-Spoor and colleagues (1982).
Behavioral abnormalities were significantly increased only in those preterm subjects
with abnormal MRI scans. Social adjustment problems were increased in subjects
with equivocal and abnormal MRI. Those with normal MRI scans showed similar
social adjustment to controls. At 18 years, there was an increased prevalence of
psychiatric illness in the VPT subjects compared with controls, but this difference
did not reach statistical significance (Rooney et al., 2001). In particular, there was a
2% rate of psychotic disorder in the VPT group, which exceeds the expected 0.2%
population prevalence for this age. This result should be interpreted with caution
in this relatively small study group but is of obvious interest in the context of the
neurodevelopmental theory of schizophrenia.
There were some areas of brain structural abnormality, however, that proved
to be more directly associated with functioning. Reduced cerebellar volume was
significantly associated with deficits in executive and visuospatial function and
language (Allin et al., 2001), findings that are broadly consistent with the “cerebellar
cognitive–affective syndrome” described by Schmahmann and Sherman (1998).
This suggests that cerebellar abnormalities may underlie some of the cognitive
deficits found in VPT individuals. There was no association between cerebellar
volume and motor neurological signs, however; perhaps suggesting that plasticity
can allow motor functions to be spared, but at the expense of subtle abnormalities
in cognitive performance.
VPT individuals have been reported to have poorer interhemispheric interac-
tion (Roth et al., 1994), and it is possible that this is related to the reduced size
of the corpus callosum, which is the major white matter tract connecting the two
hemispheres. Nosarti et al. (2001a) found that verbal IQ, reading age, and verbal
fluency scores were positively associated with reduced midsagittal area of the cor-
pus callosum. Preliminary results of a functional MRI (fMRI) study conducted by
our group showed that VPT boys with callosal thinning exhibited differential lat-
eralization of phonological processing during a verbal fluency task compared with
full-term controls (Rushe et al., 1999). Another fMRI study used visual and auditory
tasks in a similar cohort of preterm boys in adolescence (Santhouse et al., 2002)
and showed that the VPT subjects with damaged corpora callosa had significantly
different activation patterns from the control group, and from a group of VPT
adolescents without callosal damage. The findings suggest that the VPT brains
are using alternative neural networks to circumvent the damaged corpus callo-
sum. This could be a demonstration of “rerouting plasticity,” as will be discussed
below.
The left and right caudate nuclei were reduced in volume in VPT individuals,
but this was not significant after controlling for total brain volume (Nosarti et al.,
2001b). However, reduced bilateral caudate volumes were associated with higher
scores on the “hyperactivity” index of the Rutter behavioral scale (Schachar et al.,
1981). This characterizes the predominantly hyperactive/impulsive subtype of
attention-deficit hyperactivity disorder (ADHD), defined by poor inhibitory regu-
lation, limitations in perseverance, perseveration, restlessness, overactivity, and
impaired self-awareness (Landau et al., 1999). This is consistent with evidence
implicating the basal ganglia in the pathogenesis of ADHD (Semrud-Clikeman
et al., 2000). This has been investigated further (Nosarti et al., 2002, 2004) in
a group of 16-year-old VPT boys using a go–no-go fMRI paradigm. The find-
ings again suggest that VPT individuals are activating different neural networks
than controls and using alternative strategies when performing these cognitive
tasks.
99 Brain plasticity and long-term function
onto brain areas; this has been successful in many areas (a good example is Broca’s
area). This structure–function relationship, however, is altered in the case of early
brain injury because functions may be remapped onto other undamaged areas of
the brain (Moses and Stiles, 2002). Cioni et al. (2001) used fMRI to study two pairs
of monozygotic twins, where one of each pair had suffered a focal hemispheric
brain injury around the time of birth. They showed that the damaged area’s duties
had been taken over by undamaged areas in the same hemisphere adjacent to the
injury site, and also by areas in the other (intact) hemisphere. Such remapping
of functions can, therefore, occur locally and at more distant sites. The larger the
lesion, the more likely it is that the contralateral hemisphere will take over (Payne
and Lomber, 2001). This also depends on the maturational state of the system that
is lesioned. For example, language can be acquired by the right hemisphere as late
as 9 years if the left hemisphere is damaged (Vargha-Khadem et al., 1997). Bates
et al. (2001) compared language production in brain-injured children and adults.
Adults with left-hemisphere lesions showed severe language impairment and those
with right-hemisphere damage showed disinhibited and “empty” speech. Children
with similar unilateral brain damage showed no language impairment whatever
the side of the lesion (Rauschecker and Marler, 1987). However, this compensation
may not be complete: the areas of the brain that have taken over functions may not
have the right structural specialization to do the job as efficiently, and they may
be diverted from other functions, which may suffer as a result. Some evidence that
developmental compensation may not be complete comes from Weintraub and
Mesulam (1983), who found that individuals born with right-sided brain lesions
showed impairments later in life in spelling, arithmetic, prosody (the ability to vary
speech to convey emotional meanings), and social interactions. This adds to the
evidence that developmental plasticity may be a double-edged sword.
Conclusions
Plasticity is a fundamental property of vertebrate nervous systems. During devel-
opment, a high degree of plasticity allows the nervous system to be moulded to
best fit its environment, and this also enables the brain to compensate for injuries
that would cause permanent loss of function in an adult. With increasing age, this
102 M. Allin, C. Nosarti, L. Rifkin, and R. M. Murray
capacity for plasticity begins to decline, with the eventual relative “crystallization”
of brain and behavior, which allows the adult to display stable, adaptive behaviors.
The enormous resilience of the young brain to injury is demonstrated by individ-
uals born preterm, in whom functional impairments are relatively mild compared
with the abnormalities of brain structure that they show. We have drawn attention
to evidence that structural reorganization and remapping or rerouting of functions
has occurred in the brains of these individuals. Given that important events of brain
maturation and development continue into adulthood, there is the possibility that
these structural changes may adversely affect function later in life. There is some
evidence for this from animal models, and the severity of certain impairments in
the preterm population seems to increase with age. It will be important to continue
to follow up preterm individuals as they enter adulthood and to determine which
factors are associated with poor outcome. There may be ways of using the residual
plasticity of the child or even early adult brain to compensate for the late effects of
early brain lesions.
REFERENCES
Cannon, M., Jones, P. B., Murray, R. M. (2002). Obstetric complications and schizophrenia:
historical and meta-analytical review. Am J Psychiatry 159: 1080–1092.
Cannon-Spoor, H. E., Potkin, S. G., Wyatt, K. J. (1982). Measurement of premorbid adjustment
in chronic schizophrenia. Schizophr Bull 8: 470–484.
Castro-Caldas, A, Petersson, K. M., Reis, A., Stone-Elander, S., Ingvar, M. (1998). The illiterate
brain: learning to read and write during childhood influences the structural organisation of
the brain. Brain 121: 1056–1063.
Chiron, C., Raynaud, C., Maziere, B., et al. (1992). Changes in regional cerebral blood flow during
brain maturation in children and adolescents. J Nucl Med 33: 696–703.
Cioni, G., Montanaro, D., Tosetti, M., Canapicchi, R., Ghelarduci, B. (2001). Reorganisation of
the sensorimotor cortex after early focal brain lesion: a functional MRI study in monozygotic
twins. Neuroreport 12: 1335–1340.
Coltman, B. W., Earley, E. M., Shahar, A., Dudek, F. E., Ide, C. F. (1995). Factors influencing mossy
fiber collateral sprouting in organotypic slice cultures of neonatal mouse hippocampus.
J Comp Neurol 362: 209–222.
Costello, A. M. de L., Hamilton, P. A., Baudin, J., et al. (1988). Prediction of neurodevelopmental
impairment at four years from brain ultrasound appearance of very preterm infants. Dev
Med Child Neurol 30: 711–722.
du Plessis, A. J., Volpe, J. J. (2002). Perinatal brain injury in the preterm and term newborn. Curr
Opin Neurol 15: 151–157.
Elbert, T., Heim, S., Rockstroh, B. (2001). Neural plasticity and development. In Handbook of
Developmental Cognitive Neuroscience, ed. C. A. Nelson, M. Luciana. Cambridge, MA: MIT
Press, pp. 191–202.
Finney, E. M., Fine, I., Dobkins, K. R. (2001). Visual stimuli activate auditory cortex in the deaf.
Nat Neurosci 4: 1171–1173.
Frith, U. (1998). Literally changing the brain. Brain 121: 1011–1012.
Girard, N., Raybaud, C., du Lac, P. (1991). MRI study of brain myelination. J Neuroradiol 18:
291–307.
Goldman, P. S. (1971). Functional development of the prefrontal cortex in early life and the
problem of plasticity. Exp Neurol 32: 366–387.
Goyen, T-A., Lui, K., Woods, R. (1998). Visual–motor, visual–perceptual and fine motor outcomes
in very-low-birth-weight children at 5 years. Dev Med Child Neurol 40: 76–81.
Grimshaw, G. M., Adelstein, A., Bryden, M. P., MacKinnon, G. E. (1998). First-language acqui-
sition in adolescence: evidence for a critical period for verbal language development. Brain
Lang 63: 237–255.
Hadders-Algra, M., Huisjes, H. J., Touwen, B. C. L. (1988). Perinatal risk factors and minor
neurological dysfunction: significance for behaviour and school achievement at 9 years. Dev
Med Child Neurol 30: 482–491.
Hall, A., McLeod, A., Counsell, C., Thomson, L., Mutch, L. (1995). School attainment, cognitive
ability and motor function in a total Scottish very-low-birthweight population at 8 years: a
controlled study. Dev Med Child Neurol 37: 1037–1050.
Hecaen, H. (1983). Acquired aphasia in childhood: revisited. Neuropsychologia 21: 581–587.
104 M. Allin, C. Nosarti, L. Rifkin, and R. M. Murray
Linkenhoker, B. A., Knudsen, E. I. (2002). Incremental training increases the plasticity of the
auditory space map in adult barn owls. Nature 419: 293–296.
Luoma, L., Herrgard, E., Martikainen, A. (1998). Neuropsychological analysis of the visuomotor
problems in children born preterm at ≤ 32 weeks of gestation: a 5 year prospective follow-up.
Dev Med Child Neurol 40: 21–30.
Mallard, E. C., Rehn, A., Rees, S., Tolcos, M., Copolov, D. (1999). Ventriculomegaly and reduced
hippocampal volume following intrauterine growth-restriction: implications for the aetiol-
ogy of schizophrenia. Schizophr Res 40: 11–21.
Marin-Padilla, M. (1996). Developmental neuropathology and impact of perinatal brain damage.
I. Hemorrhagic lesions of neocortex. J Neuropathol Exp Neurol 55: 758–773.
(1997). Developmental neuropathology and impact of perinatal brain damage. II: white matter
lesions of the neocortex. J Neuropathol Exp Neurol 56: 219–235.
(1999). Developmental neuropathology and impact of perinatal brain damage. III: gray matter
lesions of the neocortex. J Neuropathol Exp Neurol 58: 407–429.
Moses, P., Stiles, J. (2002). The lesion methodology: contrasting views from adult and child
studies. Dev Psychobiol 40: 266–277.
Msall, M. E., Buck, G. M., Schisterman, E. F., et al. (1998). Social and biomedical risks for 8 to 10
year educational outcomes of children born with extreme prematurity and without major
disability. AACPDM Abstracts. Dev Med Child Neurol Suppl H: 27.
Murray, R. M., Lewis, S. W. (1987). Is schizophrenia a neurodevelopmental disorder? Br Med J
295: 681–682.
Neville, H. J. (1993). Neurobiology of cognitive and language processing: Effects of early experi-
ence. In Brain Development and Cognition: A Reader, ed. M. H. Johnson. Oxford: Blackwell,
pp. 424–448.
Nosarti, C., Rifkin, L., Rushe, T. M., et al. (2001a). Corpus callosum size in adolescents who were
born very preterm. Pediatr Res Special Suppl 50: 15A.
Nosarti, C., Allin, M., Al-Asady, M., et al. (2001b). Behavioural and cognitive consequences of
caudate pathology in adolescents born very preterm. Neuroimage, 13: 339.
Nosarti, C., Al-Asady, M. H. S., Frangou, S., et al. (2002). Adolescents who were born very preterm
have decreased brain volumes. Brain 125: 1616–1623.
Nosarti, C., Rubia, K., Frearson, S., Rifkin, L., Murray, R. M. (2004). Altered neuronal organisation
of the brain of adolescents born very preterm during response inhibition. Schizophr Res
60(Special Suppl.): 230.
Palfrey, J. S., Singer, J. D., Walker, D. A., Butler, J. A. (1987). Early identification of children’s
special needs: a study in five metropolitan communities. J Pediatr 111: 651–659.
Paneth, N., Rudelli, R., Kazam, E., Monte, W. (1994). Brain Damage in the Preterm Infant.
Cambridge: MacKeith Press/Cambridge University Press.
Pantev, C., Oostenveld, R., Engelien, A., et al. (1998). Increased auditory cortical representation
in musicians. Nature 392: 811–814.
Payne, B. R., Lomber, S. G. (2001). Timeline: reconstructing functional systems after lesions of
cerebral cortex. Nat Rev Neurosci 2: 911–919.
Rankin, J. M., Aram, D. M., Horwitz, S. J. (1981). Language ability in right and left hemiplegia
children. Brain Lang 14: 292–306.
106 M. Allin, C. Nosarti, L. Rifkin, and R. M. Murray
Rauschecker, J. P., Marler, P. (1987). Imprinting and Cortical Plasticity: Comparative Aspects of
Sensitive Periods. New York: Wiley.
Rooney, M., Allin, M., Rifkin, L., et al. (2001). Comparison of psychopathology in prematurely
born adults from a 1979–1981 cohort, with full term born adults. Schizophr Res 49: 41.
Rosenbloom, L., Sullivan, P. B. (1996). The nutritional and neurodevelopmental consequences
of feeding difficulties in disabled children. In Clinics in Developmental Medicine, No. 104;
Feeding the Disabled Child, ed. P. B. Sullivan, L. Rosenbloom. Cambridge, UK: MacKeith
Press/Cambridge University Press, pp. 33–39.
Roth, S. C., Baudin, J., McCormick, D. C., et al. (1993). Relation between ultrasound appearance
of the brain of very preterm infants and neurodevelopmental impairment at eight years.
Dev Med Child Neurol 35: 755–768.
Roth, S. C., Baudin, J., Pezzani-Goldsmith, M., et al. (1994). Relation between neurodevelop-
mental status of very preterm infants at one and eight years. Dev Med Child Neurol 36:
1049–1062.
Rushe, T. M., Woodruff, P. W. R., Bullmore, E. B., et al. (1999). Lateralisation of language function
in adults born very preterm. Magn Reson Mater Phys, Biol Med 8(Suppl 1): 82.
Rushe, T. M., Rifkin, L., Stewart, A. L., et al. (2001). Neuropsychological outcome at adolescence
of very preterm birth and its relation to brain structure. Dev Med Child Neurol 43: 226–33.
Rutter, M., Tizard, J., Whitmore, K. (1981). Education, Health and Behaviour. Melbourne: Kreiger.
Sams-Dodd, F., Lipska, B. K., Weinberger, D. R. (1997). Neonatal lesions of the rat ventral
hippocampus result in hyperlocomotion and deficits in social behaviour in adulthood.
Psychopharmacology 132: 303–310.
Santhouse, A. M., Ffytche, D. H., Howard, R. J., et al. (2002). The functional significance of
perinatal corpus callosum damage: an fMRI study in young adults. Brain 125: 1782–1792.
Schachar, R., Rutter, M., Smith, A. (1981). The characteristics of situationally and pervasively
hyperactive children: implications for syndrome definition. J Child Psychol Psychiatry 22:
375–392.
Schmahmann, J. D., Sherman, J. C. (1998). The cerebellar cognitive–affective syndrome. Brain
121: 561–579.
Semrud-Clikeman, M., Steingard, R. J., Filipek, P., et al. (2000). Using MRI to examine brain–
behavior relationships in males with attention deficit disorder with hyperactivity. J Am Acad
Child Adolesc Psychiatry 39: 477–484.
Shewmon, D. A., Holmes, G. L., Byrne, P. A. (1999). Consciousness in congenitally decorticate
children: developmental vegetative state as a self-fulfilling prophecy. Dev Med Child Neurol
41: 364–374.
Snider, L. M. (1998). Preschool performance skills of extremely low birth weight children.
AACPDM Abstracts. Dev Med Child Neurol Suppl H: 27.
Sohma, O., Mito, T., Mizuguchi, M., Takashima, S. (1995). The prenatal age critical for the
development of the pontosubicular necrosis. Acta Neuropathol 90: 7–10.
Sowell, E. R., Thompson, P. M., Holmes, C. J., Jernigan, T. L., Toga, A. W. (1999). In vivo evidence
for post-adolescent brain maturation in frontal and striatal regions. Nat Neurosci 2: 859–
861.
Spear, P. D. (1996). Neural plasticity after brain damage. Prog Brain Res 108: 391–408.
107 Brain plasticity and long-term function
Stewart, A. L., Thorburn, R. J., Hope, P. L., et al. (1983). Ultrasound appearance of the brain in
very preterm infants and neurodevelopmental outcome at 18 months of age. Arch Dis Child
58: 598–604.
Stewart, A. L., Costello, A. M., Hamilton, P. A., et al. (1989). Relationship between neuro-
developmental status of very preterm infants at 1 and 4 years. Dev Med Child Neurol 31:
756–765.
Stewart, A. L., Rifkin, L., Amess, P. N., et al. (1999). Brain structure and neurocognitive and
behavioural function in adolescents who were born very preterm. Lancet 353: 1653–1657.
Tin, W., Wariyar, U., Hey, E. (1997). Changing prognosis for babies of less than 28 weeks’ gestation
in the north of England between 1983 and 1994. Br Med J 314: 107–111.
Vargha-Khadem, F., Carr, L. J., Isaacs, E., et al. (1997). Onset of speech after left hemispherectomy
in a nine-year-old boy. Brain 120: 159–182.
Volpe, J. (1998). Neurologic outcome of prematurity. Arch Neurol 55: 297–300.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
Weintraub, S., Mesulam, M. M. (1983). Developmental learning disabilities of the right hemi-
sphere: emotional, interpersonal, and cognitive components. Arch Neurol 40: 463–468.
Wiesel, T. N., Hubel, D. H. (1963). Single-cell responses in striate cortex of kittens deprived of
vision in one eye. J Neurophysiol 26: 1003–1017.
Part II
Etiological factors
6
111
112 C. M. Pariante and D. Cotter
The microscopic basis of these cortical volume reductions are not known, but
clues are provided by recent neuropathological studies that have shown reductions
in cortical neuronal size, dendritic complexity, and synaptic proteins in schizophre-
nia (Harrison, 1999). Smaller neurons with fewer and less-elaborate branches would
result in a diminution in the amount of neuropil, more compacted cells, and
thus increased neuronal density. It is certainly feasible to suggest that these alter-
ations underlie the regional cortical volume reductions observed in schizophrenia.
The current challenge, however, is to understand the mechanism underlying these
changes. The term atrophic is preferred over degenerative because evidence of cell
death and of a gliotic response so characteristic of degenerative diseases is absent.
However, regardless of what term is used, the findings necessitate a re-evaluation of
the neurodevelopmental hypothesis of schizophrenia as it is currently understood.
schizophrenia could cause changes in keeping with the known cellular changes
described above? Glucocorticoid-related neurotoxicity is one such candidate.
(Purba et al., 1995; Raadsheer et al., 1995). Finally, a number of studies have provided
evidence that CRF may play a role not only in the endocrinopathy of depression but
also in the behavioral signs and symptoms of the disorder. Indeed, the behavioral
effects of CRF after direct central nervous system administration are remarkably
similar to the signs and symptoms of major depression (decreased libido, decreased
appetite, psychomotor alterations, and disturbed sleep).
These increased levels of CRF in the hypothalamus are related, at least in part, to
altered feedback inhibition by endogenous glucocorticoids (de Kloet et al., 1998;
Holsboer, 2000; McQuade and Young, 2000; Pariante and Miller, 2001; Pariante
et al., 2002). In fact, glucocorticoids interact with their receptors (the mineralocor-
ticoid receptor [MR] and the glucocorticoid receptor [GR]) in HPA axis tissues,
where they are responsible for feedback inhibition of the secretion of adrenocor-
ticotropic hormone (ACTH) from the pituitary and CRF from the hypothalamus
(de Kloet et al., 1998; McEwen, 2000; Nemeroff, 1996; Young et al., 1998). The MR
has a high affinity for endogenous glucocorticoids and is believed to play a role
in the regulation of circadian fluctuations of these hormones. In contrast to the
MR, the GR has a lower affinity for endogenous glucocorticoids and is believed to
be more important in the regulation of the response to stress, when endogenous
levels of glucocorticoids are high (de Kloet et al., 1998). Consistent with the fact
that patients with major depression exhibit impaired HPA negative feedback in
the context of elevated circulating levels of cortisol, when the negative feedback is
largely mediated by the GR, many studies have demonstrated that GR-mediated
feedback inhibition is impaired in major depression. Depressed patients have non-
suppression of cortisol secretion following dexamethasone in the dexamethasone
suppression test (DST), non-suppression of ACTH secretion following hydrocorti-
sone (fast-feedback test), and lack of inhibition of ACTH responses to CRF following
dexamethasone pretreatment (DEX/CRF test) (Heuser et al., 1994, 1996; Holsboer,
2000; Nemeroff, 1996; Ribeiro et al., 1993; Young et al., 1991). The only study that
specifically looked at MR-mediated negative feedback in depression found that this
pathway is intact (or possibly oversensitive) in these patients (Young et al., 2003).
In further support of the suggestion that patients with major depression exhibit
impaired GR-mediated HPA negative feedback, a number of studies have demon-
strated that GR function is also reduced in other tissues of depressed patients, as
shown by a decreased sensitivity to the effects of glucocorticoids on immune and
metabolic functions (Pariante and Miller, 2001). While there is no consistent evi-
dence of GR expression changes in blood mononuclear cells or fibroblasts from
depressed patients (Pariante and Miller, 2001), three studies that have examined
postmortem brains have found reduced corticosteroid receptor expression (Lopez
et al., 1998; Webster et al., 2002; Xing et al., 2004).
115 Glucocorticoids in major psychiatric disorders
Conclusions
Evidence is accumulating that there are brain changes occurring during and pos-
sibly after the period of the first acute psychosis: that is, changes that are not
developmental in the traditional sense. Furthermore, these changes are not specific
to schizophrenia, either in terms of macroscopic or microscopic brain structure,
for they are also present, to a generally milder degree, in subjects with mood disor-
der, and they are in keeping with glucocorticoid-related brain changes. These brain
changes may be epiphenomena secondary to stress-related changes in glucocorti-
coid hormones and not primary pathogenetic pathways. Nevertheless, they could
have crucial clinical effects through diminishing neuronal and cortical function
and so complicate recovery from the primary illness. These changes may possibly
be reversed by therapies that protect from glucocorticoid-related neurotoxicity or
which act to promote neuroprotective cell signaling pathways. What is clear, how-
ever, is that the developmental theory of schizophrenia as it was first presented
(Lewis and Murray, 1987; Weinberger, 1987) is challenged, as it alone is insufficient
to explain the neuroanatomy of the disorder. Consequently, it is now important
to understand the illness in terms of both early and late events, which will involve
both developmental and atrophic processes.
REFERENCES
Altshuler, L. L., Bartzokis, G., Grieder, T., Curran, J., Mintz, J. (1998). Amygdala enlargement in
bipolar disorder and hippocampal reduction in schizophrenia: an MRI study demonstrating
neuroanatomic specificity. Arch Gen Psychiatry 55: 663–664.
Avitsur, R., Stark, J. L., Sheridan, J. F. (2001). Social stress induces glucocorticoid resistance in
subordinate animals. Horm Behav 39: 247–257.
Axelson, D. A., Doraiswamy, P. M., Boyko, O. B., et al. (1992). In vivo assessment of pituitary
volume with magnetic resonance imaging and systematic stereology: relationship to dexa-
methasone suppression test results in patients. Psychiatry Res 44: 63–70.
Banki, C. M., Bissette, G., Arato, M., O’Connor, L., Nemeroff, C. B. (1987). CSF corticotropin-
releasing factor-like immunoreactivity in depression and schizophrenia. Am J Psychiatry
144: 873–877.
Bauer, M. E., Vedhara, K., Perks, P., et al. (2000). Chronic stress in caregivers of dementia patients
is associated with reduced lymphocyte sensitivity to glucocorticoids. J Neuroimmunol 103:
84–92.
Bouwer, C., Claassen, J., Dinan, T. G., Nemeroff, C. B. (2000). Prednisone augmentation in
treatment-resistant depression with fatigue and hypocortisolaemia: a case series. Depress
Anxiety 12: 44–50.
Coryell, W., Tsuang, D. (1992). Hypothalamic–pituitary–adrenal axis hyperactivity and psychosis:
recovery during an 8-year follow-up. Am J Psychiatry 149: 1033–1039.
Cotter, D., Pariante, C. M. (2002). Stress and the progression of the developmental hypothesis of
schizophrenia. Br J Psychiatry 181: 363–365.
118 C. M. Pariante and D. Cotter
Cotter, D., Mackay, D., Landau, S., Kerwin, R., and Everall, I. (2001). Glial cell loss and reduced
neuronal size in the anterior cingulate cortex in major depressive disorder. Arch Gen Psychi-
atry 58: 545–553.
Crossin, K. L., Tai, M. H., Krushel, L. A., Mauro, V. P., Edelman, G. M. (1997). Glucocorticoid
receptor pathways are involved in the inhibition of astrocyte proliferation. Proc Natl Acad
Sci USA 94: 2687–2692.
de Kloet, E. R., Vreugdenhil, E., Oitzl, M. S., Joels, M. (1998). Brain corticosteroid receptor
balance in health and disease. Endocr Rev 19: 269–301.
DeBattista, C., Posener, J. A., Kalehzan, B. M., Schatzberg, A. F. (2000). Acute antidepressant effects
of intravenous hydrocortisone and CRH in depressed patients: a double-blind, placebo-
controlled study. Am J Psychiatry 157: 1334–1337.
Dinan, T. G. (1996). Noradrenergic and serotonergic abnormalities in depression: stress-induced
dysfunction? J Clin Psychiatry 57(Suppl 4): 14–18.
Dinan, T. G., Lavelle, E., Cooney, J., et al. (1997). Dexamethasone augmentation in treatment-
resistant depression. Acta Psychiatr Scand 95: 58–61.
Eastwood, S. L., Harrison, P. J. (2001). Synaptic pathology in the anterior cingulate cortex in
schizophrenia and mood disorders. A review and a western blot study of synaptophysin,
GAP-43, and the complexins. Brain Res Bull 55: 519–578.
Harrison, P. J. (1999). The neuropathology of schizophrenia: a critical review of the data and
their interpretation. Brain 122: 593–624.
Herz, M. I., Fava, G. A., Molnar, G., Edwards, L. (1985). The dexamethasone suppression test in
newly hospitalized schizophrenic patients. Am J Psychiatry 142: 127–129.
Heuser I. J., Yassouridis, A., Holsboer, F. (1994). The combined dexamethasone/CRH test: a
refined laboratory test for psychiatric disorders. J Psychiatr Res 28: 341–356.
Heuser, I. J., Schweiger, U., Gotthardt, U., et al. (1996). Pituitary–adrenal-system regulation and
psychopathology during amitriptyline treatment in elderly depressed patients and normal
comparison subjects. Am J Psychiatry 153: 93–99.
Heuser, I. J., Bissette, G., Dettling, M., et al. (1998). Cerebrospinal fluid concentrations of
corticotropin-releasing hormone, vasopressin, and somatostatin in depressed patients and
healthy controls: response to amitriptyline treatment. Depress Anxiety 8: 71–79.
Holsboer, F. (2000). The corticosteroid receptor hypothesis of depression. Neuropsychopharma-
cology 23: 477–501.
Ismail, K., Murray, R. M., Wheeler, M. J., O’Keane, V. (1998). The dexamethasone suppression
test in schizophrenia. Psychol Med 28: 311–317.
Keshavan, M. S., Haas, G. L., Kahn, C. E., et al. (1998). Superior temporal gyrus and the course
of early schizophrenia: progressive static or reversible? J Psychiatr Res 32: 161–167.
Lammers, C. H., Garcia-Borreguero, D., Schmider, J., et al. (1995). Combined dexamethasone/
corticotropin-releasing hormone test in patients with schizophrenia and in normal controls:
II. Biol Psychiatry 38: 803–807.
Lawrie, S. M., Whalley, H., Byrne, M., et al. (2000). Brain structure change and psychopathology
in subjects at high risk of schizophrenia. Schizophr Res 41: 11.
Lewis, S. W., Murray, R. M. (1987). Obstetric complications, neurodevelopmental deviance, and
risk of schizophrenia. J Psychiatr Res 21: 413–421.
119 Glucocorticoids in major psychiatric disorders
Lopez, J. F., Chalmers, D. T., Little, K. Y., Watson, S. J. (1998). A. E. Bennett Research Award.
Regulation of serotonin1A , glucocorticoid, and mineralocorticoid receptor in rat and human
hippocampus: implications for the neurobiology of depression. Biol Psychiatry 43: 547–573.
McAllister-Williams, R. H., Ferrier, I. N., Young, A. H. (1998). Mood and neuropsychological
function in depression: the role of corticosteroids and serotonin. Psychol Med 28: 573–584.
McCarley, R. W., Wible, C. G., Frumin, M., et al. (1999). MRI anatomy of schizophrenia. Biol
Psychiatry 45: 1099–1119.
McEwen, B. S. (2000). The neurobiology of stress: from serendipity to clinical relevance. Brain
Res 886: 172–189.
McQuade, R., Young, A. H. (2000). Future therapeutic targets in mood disorders: the glucocor-
ticoid receptor. Br J Psychiatry 177: 390–395.
Nemeroff, C. B. (1996). The corticotropin-releasing factor (CRF) hypothesis of depression: new
findings and new directions. Mol Psychiatry 1: 336–342.
Nemeroff, C. B., Widerlov, E., Bissette, G., et al. (1984). Elevated concentrations of CSF
corticotropin-releasing factor-like immunoreactivity in depressed patients. Science 226:
1342–1344.
Owens, M. J., Nemeroff, C. B. (1993). The role of corticotropin-releasing factor in the pathophysi-
ology of affective and anxiety disorders: laboratory and clinical studies. Ciba Found Symp 172:
296–308.
Pantellis, C., Velakoulis, D., Suckling, J., et al. (2000). Left medial temporal lobe volume reduction
occurs during the transition from high risk to first episode psychosis. Schizophr Res 41: 35.
Pariante, C. M. (2003). Depression, stress and the adrenal axis. J Neuroendocrinol 15: 811–812.
Pariante, C. M., Miller, A. H. (2001). Glucocorticoid receptors in major depression: relevance to
pathophysiology and treatment. Biol Psychiatry 49: 391–404.
Pariante, C. M., Pearce, B. D., Pisell, T. L., Owens, M. J., Miller, A. H. (1997). Steroid-independent
translocation of the glucocorticoid receptor by the antidepressant desipramine. Mol Phar-
macol 52: 571–581.
Pariante, C. M., Makoff, A., Lovestone, S., et al., (2001). Antidepressants enhance glucocorticoid
receptor function in vitro by modulating the membrane steroid transporters. Br J Pharmacol
134: 1335–1343.
Pariante, C. M., Papadopoulos, A. S., Poon, L., et al. (2002). A novel prednisolone suppression
test for the hypothalamic–pituitary–adrenal axis. Biol Psychiatry 51: 922–930.
Pariante, C. M., Hye, A., Williamson, R., et al. (2003a). The antidepressant clomipramine regulates
cortisol intracellular concentrations and glucocorticoid receptor expression in fibroblasts
and rat primary neurones. Neuropsychopharmacology 28: 1553–1561.
Pariante, C. M., Kim, R. B., Makoff, A., Kerwin, R. W. (2003b). The antidepressant fluoxetine
enhances glucocorticoid receptor function in vitro by modulating membrane steroid trans-
porters. Br J Pharmacol 139: 1111–1118.
Pariante, C. M., Vassilopoulou, K., Velakoulis, D., et al. (2004a). Pituitary volume in psychosis.
Br J Psychiatry 185: 5–10.
Pariante, C. M., Thomas, S. A., Lovestone, S., Makoff, A., Kerwin, R. W. (2004b). Do antidepres-
sants regulate how cortisol affects the brain? 2003 Curt Richter Award Paper. Psychoneuro-
endocrinology 29: 423–447.
120 C. M. Pariante and D. Cotter
Purba, J. S., Raadsheer, F. C., Hofman, M. A., et al. (1995). Increased number of corticotropin-
releasing hormone expressing neurons in the hypothalamic paraventricular nucleus of
patients with multiple sclerosis. Neuroendocrinology 62: 62–70.
Raadsheer, F. C., van Heerikhuize, J. J., Lucassen, P. J., et al. (1995). Corticotropin-releasing
hormone mRNA levels in the paraventricular nucleus of patients with Alzheimer’s disease
and depression. Am J Psychiatry 152: 1372–1376.
Rajkowska, G., Miguel-Hidalgo, J. J., Wei. J. (1999). Morphometric evidence for neuronal and
glial prefrontal cell pathology in major depression. Biol Psychiatry 45: 1085–1098.
Rappaport, J. L., Giedd, J. N., Blumenthal, J., et al. (1999). Progressive cortical change during
adolescence in childhood-onset schizophrenia. A longitudinal magnetic resonance imaging
study. Arch Gen Psychiatry 56: 649–654.
Ribeiro, S. C., Tandon, R., Grunhaus, L., Greden, J. F. (1993). The DST as a predictor of outcome
in depression: a meta-analysis. Am J Psychiatry 150: 1618–1629.
Rosoklija, G., Toomayan, G., Ellis, S. P., et al. (2000). Structural abnormalities of subicular
dendrites in subjects with schizophrenia and mood disorders. Arch Gen Psychiatry 57: 349–
356.
Sachar, E. J., Kanter, S. S., Buie, D., Engle, R., Mehlman, R. (1970). Psychoendocrinology of ego
disintegration. Am J Psychiatry 126: 1067–1078.
Sapolsky, R. (2000). The possibility of neurotoxicity in the hippocampus in major depression:
a primer on neuron death. Biol Psychiatry 48: 755–65.
Sharma, R. P., Pandey, G. N., Janicak, P. G., et al. (1988). The effect of diagnosis and age on the
DST: a metaanalytic approach. Biol Psychiatry 24: 555–568.
Tandon, R., Mazzara, C., DeQuardo, J., et al. (1991). Dexamethasone suppression test in
schizophrenia: relationship to symptomatology, ventricular enlargement, and outcome. Biol
Psychiatry 29: 953–964.
Walder, D. J., Walker, E. F., Lewine, R. J. (2000). Cognitive functioning, cortisol release, and
symptom severity in patients with schizophrenia. Biol Psychiatry 48: 1121–1132.
Walker, E. F., Walder, D. J., Reynolds, F. (2001). Developmental changes in cortisol secretion in
normal and at-risk youth. Dev Psychopathol 13: 721–732.
Webster, M. J., Knable, M. B., O’Grady, J., Orthmann, J., Weickert, C. S. (2002). Regional specificity
of brain glucocorticoid receptor mRNA alterations in subjects with schizophrenia and mood
disorders. Mol Psychiatry 7: 985–994, 924.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
Xing, G. Q., Russell, S., Webster, M. J., Post, R. M. (2004). Decreased expression of mineralo-
corticoid receptor mRNA in the prefrontal cortex in schizophrenia and bipolar disorder. Int
J Neuropsychopharmacol 7: 143–153.
Young, E. A., Haskett, R. F., Murphy-Weinberg, V., Watson, S. J., Akil, H. (1991). Loss of gluco-
corticoid fast feedback in depression. Arch Gen Psychiatry 48: 693–699.
Young, E. A., Lopez, J. F., Murphy-Weinberg, V., Watson, S. J., Akil, H. (1998). The role of
mineralocorticoid receptors in hypothalamic–pituitary–adrenal axis regulation in humans.
J Clin Endocrinol Metab 83: 3339–3345.
Young, E. A., Lopez, J. F., Murphy-Weinberg, V., Watson, S. J., Akil, H. (2003). Mineralocorticoid
receptor function in major depression. Arch Gen Psychiatry 60: 24–28.
7
Since the pioneering reports by Barbara Fish in 1977 and of Daniel Weinberger
in 1987, there has been increasing awareness that schizophrenia is most likely a
neurodevelopmental disorder (Fish, 1977; Weinberger, 1987). According to this
model of pathogenesis, there are underlying deviations already present in the early
brain development of individuals who develop schizophrenia in adulthood. These
neurodevelopmental abnormalities do not present as classical schizophrenia but as
more subtle neurocognitive deficits. To test this theoretical model, several prospec-
tive longitudinal studies as well as other approaches that link earlier childhood
clinical data to onset of schizophrenia in adulthood have been utilized in the search
for early life indicators of vulnerability to this tragic disorder. Recent findings from
these studies consistently indicate that children at high genetic risk for schizophre-
nia do, in fact, have significantly elevated rates of neurocognitive and behavioral
problems (Cannon et al., 2002; Erlenmeyer-Kimling et al., 2000; Fish et al., 1992;
Hans et al., 1999; Marcus et al., 1993). In addition, closely related research has
demonstrated that, prior to the onset of schizophrenia in cohorts of adults, clear
indicators of neurocognitive impairments were present earlier in life (Reichenberg
et al., 2002). In a prospective longitudinal study of a population-based (non-high-
risk) cohort, significant neuromotor, cognitive, and language impairments were
present only among children who were later diagnosed with schizophreniform dis-
order or schizophrenia (Cannon et al., 2002). Most recently, subtle but similar
neurocognitive impairments have been found in both non-schizophrenia children
and adults with elevated familial risk for schizophrenia (Asarnow et al., 2002a,b).
In summary, there is now a very impressive body of data supporting the neurode-
velopmental hypothesis of schizophrenia.
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
121
122 S. Eliez and C. Feinstein
Just prior to the research findings summarized above, in 1992, Shprintzen and col-
leagues reported that children with velo-cardio-facial syndrome (VCFS), a genetic
disability syndrome caused, in most cases, by a de novo 3 MB microdeletion at chro-
mosome region 22q11.2, were at remarkably high risk for developing schizophrenia
in adulthood. Of children with this medically complex chromosome 22 microdele-
tion syndrome, known mostly to pediatric specialists (and certainly obscure to
most schizophrenia researchers), 30% developed schizophrenia or schizoaffective
disorder by young adulthood. This finding has since been confirmed and extended
by successive studies, which now indicate that approximately 2% of all individuals
with adult-onset schizophrenia, and a possibly higher percentage of youngsters with
childhood-onset schizophrenia, have VCFS, as confirmed by reliable laboratory-
based genetic diagnostic methods.
There was initial reluctance among many mainstream researchers in schizophre-
nia to acknowledge this finding. Instead, it was argued that VCFS is a childhood
developmental disability that, in adults, merely mimics the phenotype of “true”
schizophrenia. Nevertheless, the accumulation of data confirming prior childhood
neurodevelopmental impairments in adults with schizophrenia suggests that a more
heuristic approach would be to view VCFS as a model type of disorder for studying
the developmental pathogenesis of schizophrenia. Unlike the clusters of subtle neu-
rocognitive deficits that are gradually emerging from other lines of research, VCFS
has a known molecular genetic cause that can be biologically validated in earliest
infancy. This makes possible detailed study of the interaction of known genetic
factors with brain development from infancy through adulthood, as assayed by
neuroimaging, neurophysiological, neuropsychological, and psychiatric diagnostic
techniques. Through such studies, at least one biologically validated developmen-
tal pathway to the development of schizophrenia could be elucidated. This, in
turn, would create opportunities to design early interventions for the prevention
or amelioration of the schizophrenia before its most destructive symptoms appear.
In this chapter, we summarize the available clinical, neuroimaging, and genetic
information regarding VCFS, so that it may be available for clinical research in
schizophrenia.
Medical aspects
VCFS was first described by Shprintzen et al. (1978) as a distinctive congenital pat-
tern presenting in early childhood as hypernasal speech (associated either with
cleft palate or submucosal cleft), cardiac defects, distinctive facial appearance,
learning disabilities, and behavioral disturbances. Several other contemporane-
ous authors had also described children with conotruncal heart defects, palate
123 Velo-cardio-facial (22q11.2 deletion) syndrome
is very common, and also described high rates of separation-anxiety disorder and
obsessive–compulsive disorder. Dimensional behavior ratings utilizing the Child
Behavioral Checklist noted abnormally high scores in the subscales for social prob-
lems, attention problems, thought problems, withdrawn behaviors, and anxiety/
depression (Swillen et al., 1997, 1999). Children with VCFS are at an increased
risk for childhood-onset schizophrenia. Usiskin et al. (1999) recently reported that
6.4% of a cohort of youngsters with childhood-onset schizophrenia had VCFS.
However, it is also well known that rates of psychiatric disorder are elevated in
children with borderline and mild mental retardation, developmental language dis-
orders, and learning disabilities (Feinstein and Reiss, 1996; Feinstein and Weiner,
1997; Fraser and Nolan, 1994; Toppelberg and Shapiro, 2000). Consequently, it was
not clear that the high rates of psychopathology in VCFS children were distinc-
tively different from those of other children with similar degrees of developmental
impairment. Feinstein et al. (2002) compared psychiatric findings in a group of
VCFS children with a control group matched for reduced IQ and severe learning
disabilities. They found very high rates of specific phobias, generalized anxiety disor-
der, major depressive disorder, oppositional defiant disorder, higher than expected
rates of obsessive–compulsive disorder, and evidence of delusions and hallucina-
tions in a few subjects. However, VCFS patients did not have significantly higher
rates of any these disorders than the cognitively matched controls.
Despite the difficulties to date in finding specific cognitive or behavioral features
in children with VCFS that are specific indicators of later-onset psychosis, it may
be that a subset of symptoms within the total array are, indeed, specific markers.
Further longitudinal study of children with VCFS into adulthood offers the best
hope of identifying any specific markers present in childhood.
(or other significant congenital anomalies) and assaying for the VCFS chromosomal
deletion. This approach identified three VCFS patients with schizophrenia from a
pool of 20. Bassett and colleagues (Bassett and Chow, 1999; Bassett et al., 1998) have
reported association between VCFS and schizophrenia, using a similar approach
to that of Gothelf, identifying a group of 15 adult schizophrenia patients with at
least two of five common physical or developmental phenotypic features of VCFS.
Ten (67%) were found to have VCFS. The IQ scores of this group were within the
borderline to mildly retarded range. In addition to delusions, hallucinations, social
withdrawal, and adaptive impairment, most schizophrenia patients with VCFS had
a cluster of associated behavioral features, including impulsivity, frequent aggressive
or temper outbursts, mood lability, anxiety symptoms, and compulsive features.
Murphy et al. (1999) provided a decisive replication for previous findings of a
high rate of schizophrenia in adults with VCFS employing a structured psychiatric
research diagnostic algorithm for 50 VCFS patients: 30% were psychotic (24%
schizophrenia, 2% schizoaffective, 2% psychosis [not otherwise specified], and
2% rapid cycling bipolar disorder) and 12% also had a lifetime history of major
depressive disorder. The range of intelligence in the VCFS subjects was similar to
other studies (mean IQ scores in the borderline retarded range). There was no
statistical association between IQ, cardiac defects, or cleft palate problems and the
presence or absence of psychosis. No association was found between psychosis and
the allele for catechol-O-methyltransferase (COMT; see below).
Genetics
In 1992, Scambler et al. identified a microdeletion at chromosome 22q11.2 in a
majority of patients presenting with VCFS, as well as the other similar named
syndromes. Individuals with this deletion are hemizygous for all genes encompassed
by the deletion site. The 22q11.2 deletion is estimated to occur in at least 1 per 2000
to 4500 live births (Tezenas Du Montcel et al., 1996). In most affected individuals,
a de novo 3 Mb deletion at chromosome 22q11.2 is responsible for the syndrome;
however, in 10–25%, VCFS is inherited from a parent with VCFS (Carlson et al.,
1997a,b; Driscoll et al., 1993; Lindsay et al., 1995; Scambler et al., 1992; Shaikh
et al., 2000). There is some evidence that patients with inherited VCFS may have
more severe clinical manifestations (Ryan et al., 1997; Swillen et al., 1997). There
also appears to be a preponderance of maternally transmitted inheritance, perhaps
because of greater likelihood for having offspring in woman with VCFS (Murphy
et al., 1999; Ryan et al., 1997).
In fact, several microdeletions of different sizes can occur (presumably involving
different genes or different overlapping or non-overlapping regions in the deletion
127 Velo-cardio-facial (22q11.2 deletion) syndrome
site) in VCFS, all sited within the larger and by far the most commonly occurring
3 Mb 22q11.2 deletion region (Shaikh et al., 2000). The relationship between the
size and genetic content of the deletion and the resulting phenotoype is not well
understood. Moreover, not all patients presenting with features of VCFS have the
22q11.2 microdeletion. Clinic surveys have found the deletion in 76% of VCFS, 88%
of those with DiGeorge syndrome, and 84% of those with conotruncal anomaly
face syndrome (Driscoll et al., 1993; Fokstuen et al., 1998; Lindsay et al., 1995).
As diagnostic certainty will increase among clinicians, “non-deleted” individuals
with VCFS might appear as exceptions. Nevertheless, the ability to test for the
22q11.2 microdeletion greatly facilitates identifying and classifying the somatic,
cognitive, and psychiatric sequelae of hemizygosity for the chromosome 22q11.2
site.
At least 30 genes are encoded in the commonly deleted segment (Dunham
et al., 1999), a few of which are highly expressed in brain tissue and are likely
to be essential for normal brain development (Funke et al., 1997; Gottlieb et al.,
1997; Lachman et al., 1996a; Roberts et al., 1997; Yamagishi et al., 1999). Certain
specific genes within the 22q11.2 site have attracted intense interest because of
their known relationship to central nervous system structure or functioning. These
include the gene for the goosecoid-like protein (GSCL), the genes, Es2, UFDIL, the
gene for proline dehydrogenase (PDH), PIK4CA, and the gene for COMT.
Both the GSCL and ES2 genes play a role in mouse brain embryogenesis and thus
could be related to structural brain abnormalities observed in VCFS (Funke et al.,
1997; Gottlieb et al., 1997, 1998; Lindsay et al., 1998; Saint-Jore et al., 1998). Yamag-
ishi et al. (1998) measured the expression of a newly identified gene, UFDIL, which
has been localized to the commonly deleted region. Of 182 subjects with VCFS, all
had that specific gene deleted. Findings from this study indicate that UFDIL plays
a key role in the embryonic development of the heart, palate, frontonasal regions,
and brain. Specific polymorphisms of PIK4CA (Saito et al., 2003) and the gene
for PDH (Jacquet et al., 2002; Liu et al., 2002a) have been found to be potentially
more common among populations of individuals with schizophrenia or bipolar
disorder. Therefore, polymorphisms of PIK4CA and the gene for PDH have been
hypothesized to be also more frequent in individuals with VCFS and psychosis. So
far, no reports have clearly established, within the VCFS population, a link between
psychiatric symptoms and allelic variants.
More recently, three independent groups have targeted the T-box gene Tbx1
for gene inactivation in mouse models (Jerome and Papaioannou, 2001; Lindsay
et al., 2001; Merscher et al., 2001). Tbx1 homozygosity was perinatally lethal and
associated with several usual features of VCFS such as thymus and parathyroid
gland aplasia, major ear malformations, cleft palate, and conotruncal heart defects
128 S. Eliez and C. Feinstein
haplotype within the segment, in which PRODH2 and DGCR6 are the only known
genes, where the schizophrenia susceptibility variant(s) most likely resides.
Neuroimaging
Since the early 1990s, magnetic resonance imaging (MRI) methodologies have been
utilized to investigate the effect of the 22q11.2 deletion on brain structure. Mitnick
et al. (1994) reported brain abnormalities, most commonly small cerebellar ver-
mises, among 9 of 11 children with VCFS. Reduced volume of the posterior fossa
was found in four, and cysts adjacent to the anterior horns of the ventricles were
found in three. Chow et al. (1999) studied 11 adults with VCFS and schizophrenia
and found that 90% exhibited bilateral white matter hyperintensities, distributed
mainly within the frontal lobes; 45% had either cavum septum pellucidum or
cavum vergae, and 36% had cerebellar hypoplasia. More recently, several cases of
cortical dysgenesis including uni/bilateral frontoparietal polymicrogyria have been
reported in children with 22q11 deletion syndrome (Bingham et al., 1998; Bird,
2001; Ghariani et al., 2002; Kawame et al., 2000; Worthington et al., 2000), possibly
resulting in hemiplegia and seizures in the most severely affected patient. The under-
lying pathogenic mechanism for the 22q11 deletion syndrome-associated cortical
dysgenesis is still unclear. Abnormal neuronal migration is one proposed explan-
ation, but vascular disruption has also been suggested as a potential mechanism
(Bird, 2001).
There are only a limited number of publications presenting MRI quantitative data
in VCFS (Chow et al., 2002; Eliez et al., 2000; Kates et al., 2001; van Amelsvoort
et al., 2001). Eliez et al. (2000) first reported significant alterations in cerebral
morphology among children and adolescents with VCFS; an 11% reduction in total
brain volume was detected related to both gray and white matter reduction. Frontal
lobe brain volume was relatively enlarged in subjects with VCFS. Most notable
was a significant reduction of parietal lobe gray matter in the left hemisphere and
alteration of cerebellar volumes and vermis (Eliez et al., 2001a). The dissociation
between the alteration of frontal and non-frontal regions in children with VCFS
was confirmed by Kates et al. (2001) with white matter tissue affected to a greater
extent than white matter. Interestingly, these authors also reported a reduction of
white matter in the left temporal cortex.
Investigating differences between 10 adults with VCFS and 13 age- and IQ-
matched controls (among which two met criteria for schizophrenia and four for
mood disorder), van Amelsvoort et al. (2001) described reduction of gray matter in
the cerebellum and left temporal lobe, and reduction of white matter in the superior
medial regions of the frontal lobe and in the frontotemporal regions. Another study,
comparing 14 adults with VCFS and schizophrenia with 14 normal gender- and
130 S. Eliez and C. Feinstein
Fig. 7.1. As suggested by several authors (McGlashan and Hoffman, 2000), the reduction of gray
matter volumes in schizophrenia (SZ) is principally a result of abnormal dendritic arboriz-
ation, giving rise to excessive pruning rather than decreased number of neurons. One might
speculate that identical neurohistological characteristics are responsible for the gray matter
volume decrease observed in individuals with the 22q11 deletion. To date, no detailed
postmortem neuroanatomical and neurohistological studies have been published on human
or animal models with velo-cardio-facial syndrome. These studies will be most valuable in
order to understand the relation between genetic alteration and changes in white and gray
matter tissue compartments.
age-matched controls (Chow et al., 2002), reported an overall decrease of gray but
not white matter. Lobar gray matter reduction was observed in the frontal and
temporal lobes bilaterally and in the left parietal and occipital lobes. Ventricular
cerebrospinal fluid was increased in individuals with VCFS and schizophrenia com-
pared with controls. Most of the brain alterations described in VCFS are common to
those reported in neuroimaging literature on schizophrenia (Shenton et al., 2001).
A review of most of the available neuroimaging studies of children with VCFS
would suggest that there is an early alteration of parietal lobe and cerebellum, and
that the decrease of temporal lobe gray matter and hippocampus can be observed
only in adults. However, in a study investigating volumetric changes with age in
VCFS, Eliez et al. (2001b) found that accelerated decrease in volumes of gray matter
in the temporal lobes and hippocampus already occurred during childhood and
adolescence. The accelerated decrease of gray matter in temporal and mesiotem-
poral structures is likely to lead to significant volumetric differences by the time
individuals with VCFS reach young adulthood. In another publication by the same
group (Eliez et al., 2001c), it has been suggested that individuals with a maternally
derived 22q11.2 microdeletion have an accelerated decrease of gray matter, leading
131 Velo-cardio-facial (22q11.2 deletion) syndrome
Conclusions
In summary, we have described the medical, genetic, cognitive, and behavioral phe-
notype of VCFS from infancy to adulthood, with particular emphasis on the vari-
ability of the phenotype and the strong association between the 22q11.2 chromoso-
mal microdeletion and schizophrenia. VCFS represents a developmental model of a
genetically homogeneous subtype of schizophrenia and a unique window to under-
stand the interaction in children and adolescents between genes, brain, behavior,
and cognition to lead to adult psychiatric disorders.
REFERENCES
Asarnow, R. F., Nuechterlein, K. H., Asamen, J., et al. (2002a). Neurocognitive functioning and
schizophrenia spectrum disorders can be independent expressions of familial liability for
schizophrenia in community control children: the UCLA family study. Schizophr Res 54:
111–120.
132 S. Eliez and C. Feinstein
Asarnow, R. F., Nuechterlein, K. H., Subotnik, K. L., et al. (2002b). Neurocognitive impairments
in nonpsychotic parents of children with schizophrenia and attention-deficit/hyperactivity
disorder: the University of California, Los Angeles Family Study. Arch Gen Psychiatry 59:
1053–1060.
Bassett, A. S., Chow, E. W. (1999). 22q11 deletion syndrome: a genetic subtype of schizophrenia.
Biol Psychiatry 46: 882–891.
Bassett, A. S., Hodgkinson, K., Chow, E. W., et al. (1998). 22q11 deletion syndrome in adults
with schizophrenia. Am J Med Genet 81: 328–337.
Bilder, R., Volavka, J., Czobor, P., et al. (2002). Neurocognitive correlates of the COMT
Val(158)Met polymorphism in chronic schizophrenia. Biol Psychiatry 52: 701–707.
Bingham, P. M., Lynch, D., McDonald-McGinn, D., Zackai, E. (1998). Polymicrogyria in chro-
mosome 22 delection syndrome. Neurology 51: 1500–1502.
Bird, L. M. (2001). Cortical dysgenesis and 22q11 deletion. Clin Dysmorphol 10: 77.
Cannon, M., Caspi, A., Moffitt, T. E., et al. (2002). Evidence for early-childhood, pan-
developmental impairment specific to schizophreniform disorder: results from a longi-
tudinal birth cohort. Arch Gen Psychiatry 59: 449–456.
Carlson, C., Papolos, D., Pandita, R. K., et al. (1997a). Molecular analysis of velo-cardio-facial
syndrome patients with psychiatric disorders. Am J Hum Genet 60: 851–859.
Carlson, C., Sirotkin, H., Pandita, R., et al. (1997b). Molecular definition of 22q11 deletions in
151 velo-cardio-facial syndrome patients. Am J Hum Genet 61: 620–629.
Chakravarti, A. (2002). A compelling genetic hypothesis for a complex disease, PRODHH2/
DGCR6 variation leads to schizophrenia susceptibility. Proc Natl Acad Sci USA 99: 4755–
4756.
Chow, E., Zipursky, R. B., Mikulis, D., et al. (1999). MRI findings in adults with 22q11 deletion
syndrome (22qDS) and schizophrenia. Schizophr Res 36: 89.
Chow, E. W., Zipursky, R. B., Mikulis, D. J., Bassett, A. S. (2002). Structural brain abnormalities
in patients with schizophrenia and 22q11 deletion syndrome. Biol Psychiatry 51: 208–215.
Cohen, E., Chow, E., Weksberg, R., Bassett, A. S. (1999). Phenotype of adults with the 22q11
deletion syndrome. Am J Med Genet 86: 359–365.
Cooper, J., Bloom, F., Roth, R. (1996): The Biochemical Basis of Neuropharmacology, 7th edn.
New York: Oxford University Press.
Driscoll, D. A., Salvin, J., Sellinger, B., et al. (1993). Prevalence of 22q11 microdeletions in
DiGeorge and velocardiofacial syndromes: implications for genetic counselling and prenatal
diagnosis. J Med Genet 30: 813–817.
Dunham, I., Shimizu, N., Roe, B. A., et al. (1999). The DNA sequence of human chromosome
22. Nature 402: 489–495.
Eliez, S., Schmitt, J. E., White, C. D., Reiss, A. L. (2000). Children and adolescents with velocar-
diofacial syndrome: a volumetric MRI study. Am J Psychiatry 157: 409–415.
Eliez, S., Schmitt, J. E., White, C. D., Wellis, V. G., Reiss, A. L. (2001a). A quantitative MRI study
of posterior fossa development in velocardiofacial syndrome. Biol Psychiatry 49: 540–546.
Eliez, S., Blasey, C. M., Schmitt, E. J., et al. (2001b). Velocardiofacial syndrome: are structural
changes in the temporal and mesial temporal regions related to schizophrenia? Am J Psychi-
atry 158: 447–453.
133 Velo-cardio-facial (22q11.2 deletion) syndrome
Eliez, S., Antonarakis, S. E., Morris, M. A., Dahoun, S. P., Reiss,, A. L. (2001c). Parental origin
of the deletion 22q11.2 and brain development in velocardiofacial syndrome: a preliminary
study. Arch Gen Psychiatry 58: 64–68.
Erlenmeyer-Kimling, L., Rock, D., Roberts, S., et al. (2000). Attention, memory, and motor
skills as childhood predictors of schizophrenia-related psychosis. Am J Psychiatry 157:
1416–1422.
Feinstein, C., Reiss, A. L. (1996). Psychiatric disorders in mentally retarded children and adoles-
cents. Child Adolesc Psychiatr Clin N Am 5: 827–852.
Feinstein, C., Weiner, J. (1997). Developmental disorders of learning, motor skills, and commu-
nication. In Textbook of Child and Adolescent Psychiatary, ed. J. Weiner. Washington, DC:
American Psychiatry Press, pp. 281–300.
Feinstein, C., Eliez, S., Blasey, C., Reiss, A. L. (2002). Psychiatric disorders and behavioral prob-
lems in children with velocardiofacial syndrome: usefulness as phenotypic indicators of
schizophrenia risk. Biol Psychiatry 51: 312–318.
Fish, B. (1977). Neurobiologic antecedents of schizophrenia in children. Evidence for an inherited,
congenital neurointegrative defect. Arch Gen Psychiatry 34: 1297–1313.
Fish, B., Marcus, J., Hans, S. L., Auerbach, J. G., Perdue, S. (1992). Infants at risk for schizophrenia:
sequelae of a genetic neurointegrative defect. A review and replication analysis of pandsy-
maturation in the Jerusalem Infant Development Study. Arch Gen Psychiatry 49: 221–235.
Fokstuen, S., Arbenz, U., Artan, S., et al. (1998). 22q11.2 deletions in a series of patients with
non-selective congenital heart defects: incidence, type of defects and parental origin. Clin
Genet 53: 63–69.
Fraser, W., Nolan, M. (1994). Psychiatric disorders in mental retardation. In Mental Health in
Mental Retardation, ed. N. Bouras. Cambridge: Cambridge University Press, pp. 79–92.
Fuller, R., Nopoulos, P., Arndt, S., et al. (2002). Longitudinal assessment of premorbid cognitive
functioning in patients with schizophrenia through examination of standardized scholastic
test performance. Am J Psychiatry 159: 1183–1189.
Funke, B., Saint-Jore, B., Puech, A., et al. (1997). Characterization and mutation analysis of
goosecoid-like (GSCL), a homeodomain-containing gene that maps to the critical region
for VCFS/DGS on 22q11. Genomics 46: 364–372.
Gerdes, M., Solot, C., Wang, P. P., et al. (1999). Cognitive and behavior profile of preschool
children with chromosome 22q11.2 deletion. Am J Med Genet 85: 127–133.
Ghariani, S., Dahan, K., Saint-Martin, C., et al. (2002). Polymicrogyria in chromosome 22q11
deletion syndrome. Eur J Paediatr Neurol 6: 73–77.
Glaser, B., Mumme, D. L., Blasey, C., et al. (2002). Language skills in children with velocardiofacial
syndrome (deletion 22q11.2). J Pediatr 140: 753–758.
Gold, S., Arndt, S., Nopoulos, P., O’Leary, D. S., Andreasen, N. C. (1999). Longitudinal study
of cognitive function in first-episode and recent-onset schizophrenia. Am J Psychiatry 156:
1342–1348.
Goldberg, R., Motzkin, B., Marion, R., Scambler, P. J., Shprintzen, R. J. (1993). Velo-cardio-facial
syndrome: a review of 120 patients. Am J Med Genet 45: 313–319.
Golding-Kushner, K. J., Weller, G., Shprintzen, R. J. (1985). Velo-cardio-facial syndrome: lan-
guage and psychological profiles. J Craniofac Genet Dev Biol 5: 259–266.
134 S. Eliez and C. Feinstein
Gong, W., Emanuel, B. B., Collins, J. E., et al. (1996). A transcription map of the DiGeorge and
velocardiofacial syndrome minimal critical region. Hum Mol Genet 5: 789–800.
Gothelf, D., Frisch, A., Munitz, H., et al. (1997). Velocardiofacial manifestations and microdele-
tions in schizophrenic inpatients. Am J Med Genet 72: 455–461.
Gottlieb, S., Emanuel, B. S., Driscoll, D. A., et al. (1997). The DiGeorge syndrome minimal critical
region contains a goosecoid-like (GSCL) homeobox gene that is expressed early in human
development. Am J Hum Genet 60: 1194–1201.
Gottlieb, S., Hanes, S. D., Golden, J. A., Oakey, R. J., Budarf, M. L. (1998). Goosecoid-like, a gene
deleted in DiGeorge and velocardiofacial syndromes, recognizes DNA with a bicoid-like
specificity and is expressed in the developing mouse brain. Hum Mol Genet 7: 1497–1505.
Gur, R. E., Turetsky, B. I., Bilker, W. B., Gur, R. C. (1999). Reduced gray matter volume in
schizophrenia. Arch Gen Psychiatry 56: 905–911.
Hans, S. L., Marcus, J., Nuechterlein, K. H., et al. (1999). Neurobehavioral deficits at adolescence
in children at risk for schizophrenia: the Jersualem Infant Development Study. Arch Gen
Psychiatry 56: 741–748.
Jacquet, H., Raux, G., Thibaut, F., et al. (2002). PRODH mutations and hyperprolinemia in a
subset of schizophrenic patients. Hum Mol Genet 11: 2243–2249.
Jerome, L. A., Papaioannou, V. E. (2001). DiGeorge syndrome phenotype in mice mutant for the
T-box gene, Tbx1. Nat Genet 27: 286–291.
Karayiorgou, M., Morris, M. A., Morrow, B., et al. (1995). Schizophrenia susceptibility associated
with interstitial deletions of chromosome 22q11. Proc Natl Acad Sci USA 92: 7612–7616.
Kates, W. R., Burnette, C. P., Jabs, E. W., et al. (2001). Regional cortical white matter reductions
in velocardiofacial syndrome: a volumetric MRI analysis. Biol Psychiatry 49: 677–684.
Kawame, H., Kurosawa, K., Akatsuka, A., Ochiai, Y., Mizuno, K. (2000). Polymicrogyria is an
uncommon manifestation in 22q11.2 deletion syndrome. Am J Med Genet 94: 77–78.
Kirov, G., Murphy, K. C., Arranz, M., et al. (1998). Low activity allele of catechol-O-
methyltransferase gene associated with rapid cylcing bipolar disorder. Mol Psychiatry 3:
342–345.
Kunugi, H., Vallada, H., Sham, P., et al. (1997). Catechol-O-methyltransferase polymorphisms
and schizophrenia: a transmission disequilibrium study in multiply affected families.
Psychiatr Genet 7: 97–101.
Lachman, H. M., Morrow, B., Shprintzen, R., et al. (1996a). Association of codon 108/158
catechol-O-methyltransferase gene polymorphism with the psychiatric manifestations of
velo-cardio-facial syndrome. Am J Med Genet 67: 468–472.
Lachman, H. M., Papolos, D. F., Saito, T., et al. (1996b). Human catechol-O-methyltransferase
pharmacogenetics: description of a functional polymorphism and its potential application
to neuropsychiatric disorders. Pharmacogenetics 6: 243–250.
Lachman, H., Kelsoe, J., Moreno, L., Katz, S., Papolos, D. (1997). Lack of association of catechol-O-
methyltransferase (COMT) functional polymorphism in bipolar affective disorder. Psychiatr
Genet 7: 13–17.
Lindsay, E. A., Greenberg, F., Shaffer, L. G., et al. (1995). Submicroscopic deletions at 22q11.2:
variability of the clinical picture and delineation of a commonly deleted region. Am J Med
Genet 56: 191–197.
135 Velo-cardio-facial (22q11.2 deletion) syndrome
Lindsay, E. A., Harvey, E., Scambler, P., Baldini, A. (1998). ES2, a gene deleted in DiGeorge
syndrome, encodes a nuclear protein and is expressed during early mouse development,
where it shares an expression domain with a goosecoid-like gene. Hum Mol Genet 7: 629–
635.
Lindsay, E. A., Vitelli, F., Su, H., et al. (2001). Tbx1 haploinsufficiency in the DiGeorge syndrome
region causes aortic arch defects in mice. Nature 410: 97–101.
Liu, H., Heath, S. C., Sobin, C., et al. (2002a). Genetic variation at the 22q11 PRODH 2/DGCR6
locus presents an unusual pattern and increases susceptibility to schizophrenia. Proc Natl
Acad Sci USA 99: 3717–3722.
Liu, H., Abecasis, G. R., Heath, S. C., et al. (2002b). Genetic variation in the 22q11 locus and
susceptibility to schizophrenia. Proc Natl Acad Sci USA 99: 16859–16864.
Liu, H., Heath, S. C., Sobin, C., et al. (2002c). Genetic variation at the 22q11 PRODH2/DGCR6
locus presents an unusual pattern and increases susceptibility to schizophrenia. Proc Natl
Acad Sci USA 99: 3717–3722.
Marcus, J., Hans, S. L., Auerbach, J. G., Auerbach, A. G. (1993). Children at risk for schizophrenia:
the Jerusalem Infant Development Study II. Neurobehavioral deficits at school age. Arch Gen
Psychiatry 50: 797–809.
McDermid, H. E., Morrow, B. E. (2002). Genomic disorders on 22q11. Am J Hum Genet 70:
1077–1088.
McDonald-McGinn, D. M., Kirschner, R., Goldmuntz, E., et al. (1999). The Philadelphia story:
the 22q11.2 deletion. Report on 250 patients. Genet Couns 10: 11–24.
McGlashan, T. H., Hoffman, R. E. (2000). Schizophrenia as a disorder of developmentally reduced
synaptic connectivity. Arch Gen Psychiatry 57: 637–648.
Merscher, S., Funke, B., Epstein, J. A., et al. (2001). TBX1 is responsible for cardiovascular defects
in velo-cardio-facial/DiGeorge syndrome. Cell 104: 619–629.
Mitnick, R. J., Bello, J. A., Shprintzen, R. J. (1994). Brain anomalies in velo-cardio-facial syndrome.
Am J Med Genet 54: 100–106.
Mohamed, S., Paulsen, J. S., O’Leary, D., Arndt, S., Andreasen, N. (1999). Generalized cognitive
deficits in schizophrenia: a study of first-episode patients. Arch Gen Psychiatry 56: 749–754.
Moss, E. M., Batshaw, M. L., Solot, C. B., et al. (1999). Psychoeducational profile of the 22q11.2
microdeletion: a complex pattern. J Pediatr 134: 193–198.
Murphy, K. C., Jones, L. A., Owen, M. J. (1999). High rates of schizophrenia in adults with
velo-cardio-facial syndrome. Arch Gen Psychiatry 56: 940–945.
Palmatier, M. A., Kang, A. M., Kidd, K. K. (1999). Global variation in the frequencies of func-
tionally different catechol-O-methyltransferase alleles. Biol Psychiatry 46: 557–567.
Papolos, D. F., Faedda, G. L., Veit, S., et al. (1996). Bipolar spectrum disorders in patients diagnosed
with velo-cardio-facial syndrome: does a hemizygous deletion of chromosome 22q11 result
in bipolar affective disorder? Am J Psychiatry 153: 1541–1547.
Papolos, D. F., Veit, S., Faedda, G. L., Saito, T., Lachman, H. M. (1998). Ultra-ultra rapid cycling
bipolar disorder is associated with the low activity catecholamine-O-methyltransferase allele.
Mol Psychiatry 3: 346–349.
Pulver, A. E., Nestadt, G., Goldberg, R., et al. (1994). Psychotic illness in patients diagnosed with
velo-cardio-facial syndrome and their relatives. J Nerv Ment Dis 182: 476–478.
136 S. Eliez and C. Feinstein
Reichenberg, A., Weiser, M., Rabinowitz, J., et al. (2002). A population-based cohort study of
premorbid intellectual, language, and behavioral functioning in patients with schizophrenia,
schizoaffective disorder, and nonpsychotic bipolar disorder. Am J Psychiatry 159: 2027–
2035.
Roberts, C., Daw, S. C., Halford, S., Scambler, P. J. (1997). Cloning and developmental expression
analysis of chick Hira (Chira), a candidate gene for DiGeorge syndrome. Hum Mol Genet 6:
237–245.
Ryan, A. K., Goodship, J. A., Wilson, D. I., et al. (1997). Spectrum of clinical features associated
with interstitial chromosome 22q11 deletions: a European collaborative study. J Med Genet
34: 798–804.
Saint-Jore, B., Puech, A., Heyer, J., et al. (1998). Goosecoid-like (Gscl ), a candidate gene for
velocardiofacial syndrome, is not essential for normal mouse development. Hum Mol Genet
7: 1841–1849.
Saito, T., Stopkova, P., Diaz, L., et al. (2003). Polymorphism screening of PIK4CA: possible
candidate gene for chromosome 22q11-linked psychiatric disorders. Am J Med Genet 116:
77–83.
Scambler, P. J., Kelly, D., Lindsay, E., et al. (1992). Velo-cardio-facial syndrome associated with
chromosome 22 deletions encompassing the DiGeorge locus. Lancet 339: 1138–1139.
Scherer, N. J., D’Antonio, L. L., Kalbfleisch, J. H. (1999). Early speech and language development
in children with velocardiofacial syndrome. Am J Med Genet 88: 714–723.
Shaikh, T. H., Kurahashi, H., Saitta, S. C., et al. (2000). Chromosome 22-specific low copy repeats
and the 22q11.2 deletion syndrome: genomic organization and deletion endpoint analysis.
Hum Mol Genet 9: 489–501.
Shenton, M. E., Dickey, C. C., Frumin, M., McCarley, R. W. (2001). A review of MRI findings in
schizophrenia. Schizophr Res 49: 1–52.
Shifman, S., Bronstein, M., Sternfeld, M., et al. (2002). A highly significant association between
a COMT haplotype and schizophrenia. Am J Hum Genet 71: 1296–1302.
Shprintzen, R. J. (1999). Historic overview of VCFS. Genet Couns 10: 1–2.
Shprintzen, R. J., Goldberg, R. B., Lewin, M. L., et al. (1978). A new syndrome involving cleft palate,
cardiac anomalies, typical facies, and learning disabilities: velo-cardio-facial syndrome. Cleft
Palate J 15: 56–62.
Shprintzen, R. J., Goldberg, R., Golding-Kushner, K. J., Marion, R. W. (1992). Late-onset psychosis
in the velo-cardio-facial syndrome. Am J Med Genet 42: 141–142.
Strous, R., Bark, N., Parsia, S., Vovlavka, J., Lachman, H. (1997). Analysis of a functional catechol-
O-methyltransferase gene polymorphism in schizophrenia: evidence for association with
aggressive and antisocial behavior. Psychiatr Res 69: 71–77.
Swillen, A., Devriendt, K., Legius, E., et al. (1997). Intelligence and psychosocial adjustment in
velocardiofacial syndrome: a study of 37 children and adolescents with VCFS. J Med Genet
34: 453–458.
(1999). The behavioural phenotype in velo-cardio-facial syndrome (VCFS): from infancy to
adolescence. Genet Couns 10: 79–88.
Tezenas Du Montcel, S., Mendizabai, H., et al. (1996). Prevalence of 22q11 microdeletion [letter].
J Med Genet 33: 719.
137 Velo-cardio-facial (22q11.2 deletion) syndrome
Thompson, P. M., Vidal, C., Giedd, J. N., et al. (2001). Mapping adolescent brain change reveals
dynamic wave of accelerated gray matter loss in very early-onset schizophrenia. Proc Natl
Acad Sci USA 98: 11650–11655.
Toppelberg, C., Shapiro, T. (2000). Language disorders: a 10-year research update review. J Am
Acad Child Adolesc Psychiatry 39: 143–152.
Usiskin, S. I., Nicolson, R., Krasnewich, D. M., et al. (1999). Velocardiofacial syndrome in
childhood-onset schizophrenia. J Am Acad Child Adolesc Psychiatry 38: 1536–1543.
van Amelsvoort, T., Daly, E., Robertson, D., et al. (2001). Structural brain abnormalities associated
with deletion at chromosome 22q11: quantitative neuroimaging study of adults with velo-
cardio-facial syndrome. Br J Psychiatry 178: 412–419.
Vantrappen, G., Devriendt, K., Swillen, A., et al. (1999). Presenting symptoms and clinical features
in 130 patients with the velo-cardio-facial syndrome. The Leuven experience. Genet Couns
10: 3–9.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
Weinberger, D. R., Egan, M. F., Bertolino, A., et al. (2001). Prefrontal neurons and the genetics
of schizophrenia. Biol Psychiatry 50: 825–844.
Weinshilboum, R. M. (1978). Human biochemical genetics of plasma dopamine-beta-
hydroxylase and erythrocyte catechol-O-methyltransferase. Hum Genet Suppl 1: 101–112.
Worthington, S., Turner, A., Elber, J., Andrews, P. I. (2000). 22q11 deletion and polymicrogyria:
cause or coincidence? Clin Dysmorphol 9: 193–197.
Yamagishi, H., Garg, V., Matsuoka, R., Thomas, T., Srivastava, D. (1999). A molecular pathway
revealing a genetic basis for human cardiac and craniofacial defects. Science 283: 1158–1161.
8
There is overwhelming evidence from family, adoption, and twin studies that
schizophrenia has a genetic component to its etiology. Estimates of the heritability
of schizophrenia from modern twin studies are in the region of 80% (Cardno et al.,
1999), with the remaining variance attributed to non-shared environmental factors.
Despite two decades of molecular genetics research, however, there has been slow
progress towards identifying susceptibility genes for the illness, although there have
been linkage findings replicated at some sites on the genome and potential suscep-
tibility genes within these sites have recently been reported (Stefansson et al., 2002;
Straub et al., 2002). The relative lack of success in identifying genes of major effect
for the illness, combined with the pattern of inheritance of schizophrenia, has led
many researchers to favor a multifactorial model for susceptibility, whereby liability
is continuous within the population and multiple genes interact with environmental
stressors to propel the individual over a threshold for illness expression (Gottesman,
1991). Unlike other multifactorial diseases, such as diabetes mellitus and coronary
heart disease, genetic research into schizophrenia suffers from the added disad-
vantages of having an uncertain phenotype with as yet no validating biological
test, even at postmortem, and environmental risk factors that are unknown or of
minor effect. There is growing interest, therefore, in utilizing other approaches in
the search for susceptibility genes. Given a model of continuous liability within the
population, the use of the qualitative phenotype (affected/unaffected) is often likely
to be underpowered, but it may be possible to enhance the traditional categorical
approach to phenotypic definition by identifying genetically valid traits that can
be measured quantitatively. Candidates for such traits include symptom clusters or
schizotypal traits as well as biological markers of neurobiological dysfunction or
“endophenotypes.”
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
138
139 Magnetic resonance imaging and phenotypes
ventricular volume in 85 elderly twin pairs, and White et al. (2002) reported a
correlation of 0.85 for ventricular volume among a sample of MZ pairs (although
this did not significantly differ from the correlation in unrelated matched controls,
0.52, which was relatively high presumably because of the influence of age and
gender over ventricular volume). Ventricular volume is known to be affected by
environmental insults such as low birth weight (Nosarti et al., 2002) and the true
extent of genetic control over ventricular volume will require further studies.
Bartley and colleagues (1997) reported higher correlations among MZ than
DZ twins for measures of cerebral volume, and subsequent statistical modeling
demonstrated that this was most likely a result of additive genetic effects rather
than shared environmental effects, with heritability estimated at 94%. This high
degree of genetic control over brain volume has now been confirmed beyond doubt
by several other MRI studies of twins (Baare et al., 2001a; Carmelli et al., 1998;
Pennington et al., 2000; Pfefferbaum et al., 2000; Tramo et al., 1998; White et al.,
2002). In one of the largest such studies to date, which included siblings of the
twins in an attempt to dissect further the contribution of genetic as distinct from
common environmental effects (since even DZ twins share the same prenatal envi-
ronment), very high heritabilities were also found for global gray matter (82%) and
white matter (88%) (Baare et al., 2001a). In another large twin study, Pennington
and colleagues (2000) demonstrated high genetic control over factors representing
volumes of cortical and especially subcortical brain regions. In a sample of elderly
male twins, there was evidence for high genetic control over lobar brain volumes,
especially right frontotemporal regions, volume of white matter intensities, and
the size of the corpus callosum (Carmelli et al., 1998, 2002; Pfefferbaum et al.,
2000), but for lower genetic control over hippocampal volume compared with the
other brain regions, the heritability of which was estimated at 40% (Sullivan et al.,
2001). As with ventricular volume, hippocampal volume is known to be suscep-
tible to environmental insults such as early hypoxia (Rutherford et al., 1995). A
recent study of MZ twins found high correlations between twins for measures of
gray matter, white matter, lobar volumes, cortical depth, cerebellum, thalamus, cau-
date, and putamen (White et al., 2002). Other twin studies reported a high degree of
genetic control over the size of the corpus callosum (Oppenheim et al., 1989; Tramo
et al., 1998). By comparinson, there appears to be a poorer correlation between
MZ twins for measures of cortical gyrification (Bartley et al., 1997; Steinmetz
et al., 1994), gyral and sulcal curvature (White et al., 2002), and asymmetry of
the planum temporale (Steinmetz et al., 1995), indicating less genetic control over
these measures of brain structure.
Wright et al. (2002), in a reanalysis of the twins previously analyzed by Bartley
et al. (1997) with whole brain voxel-based morphometry and more novel statistical
analysis, found evidence for large genetic effects shared by several bilateral brain
141 Magnetic resonance imaging and phenotypes
regions, especially paralimbic structures and the temporoparietal cortex. Also, using
automated morphometry of the cortical surface, Thompson et al. (2001) reported
a high degree of genetic control over large areas of gray matter encompassing
the frontal lobes and the sensorimotor and linguistic neocortical regions. Conse-
quently, there is considerable evidence for a high degree of genetic control over
cerebral volume and many subregions within the adult human brain as assessed
by volumetric measurement of MRI scans, which supports the value of exploring
brain structure in vivo for potential endophenotypes of complex genetic illnesses
like schizophrenia. Future twin studies will continue to dissect the genetic and
environmental contributions towards other structures of interest to schizophrenia
researchers.
Goldstein et al. (1999), in a volumetric analysis of the entire neocortex divided into
parcellation units, identified several regions of volume deficit, most prominently
in the middle frontal gyrus, supramarginal gyrus, and paralimbic cortices, includ-
ing the anterior cingulate, paracingulate gyri, and the insula. Other recent studies
have used computational morphometry, a range of developing methodologies that
involves automated whole brain analysis of MRI images at the voxel level. These
studies have identified volumetric reduction in a range of distributed networks
usually most prominent in fronto-temporolimbic areas and including the dorso-
lateral prefrontal cortex, frontomedial cortex, superior temporal gyrus, thalamus,
cerebellum, insula, and limbic/paralimbic regions, as well as associated white mat-
ter tracts (Ananth et al., 2002; Sigmundsson et al., 2001; Suzuki et al., 2002; Wilke
et al., 2001; Wright et al., 1999).
fronto-occipital petalias also develop during fetal life, and some studies have
reported loss or reversal of such asymmetries in schizophrenia (Bilder et al., 1994;
Falkai et al., 1995). Similarly, gyrification of the prefrontal cortex achieves stability
soon after birth and right frontal cortical hypergyrification has been reported in
schizophrenia (Vogeley et al., 2001).
Third, a series of correlations have been reported between early environmental
risk factors for schizophrenia and structural deviations in the adult brain. These
include early reports (Murray et al., 1985) of an association between increased
ventricle-to-brain ratio on CT scans and exposure to obstetric complications,
although other studies did not find a relationship between obstetric complications
and lateral ventricular enlargement (McGrath and Murray, 1995). Other struc-
tural abnormalities, such as reduced hippocampal volume, have also been linked
to obstetric complications (McNeil et al., 2000; Stefanis et al., 1999).
Fourth, some of the brain abnormalities have also been reported in the unaf-
fected relatives of patients, which would indicate that such abnormalities are not
restricted to the pathological process of psychosis but are a manifestation of familial
risk factors, the most likely candidates being genes influencing neurodevelopment
(see below). Therefore, several of the brain deviations linked to schizophrenia are
probably related to aberrant neurodevelopment. Since at least some of the suscepti-
bility genes for schizophrenia are likely to influence neurodevelopmental processes
(Jones and Murray, 1991), such brain deviations could present suitable alternative
phenotypes to utilize in the search for such genes.
Ventricles
Unaffected first-degree relatives of schizophrenia patients were first included in
CT studies as a comparison group for patients in order to reduce the considerable
genetic variation of ventricular size that exists in the population. These studies
generally demonstrated greater ventricular enlargement in patients than in their
unaffected relatives (DeLisi et al., 1986; Reveley et al., 1982; Silverman et al., 1998;
Weinberger et al., 1981; Zorrilla et al., 1997). Most did not include an independ-
ent control group and thus were not able to determine whether unaffected rela-
tives themselves displayed greater ventricular enlargement than controls. Of those
sibling CT studies that did include an independent control group, Weinberger
et al. (1981) reported significantly increased ventricle-to-brain ratio in unaffected
siblings compared with controls, whereas no significant differences were found in
studies by DeLisi et al. (1986) and Silverman et al. (1998) (the latter study also failing
to detect a significant difference in this ratio between the schizophrenia subjects and
controls). Reveley et al. (1982) reported a non-significant trend for the unaffected
twins from a small group of MZ pairs discordant for schizophrenia to have larger
ventricle-to-brain ratio than healthy twins. In a Danish cohort study of 97 adult
offspring of mothers with schizophrenia (who had actually largely lived through the
risk period for schizophrenia, with a mean age of 42 years) and 60 controls (Cannon
et al., 1993), the offspring had ventricular enlargement that increased in accordance
with likely genetic risk: from having no parent with schizophrenia, to one parent
affected, to both parents affected with a schizophrenia spectrum disorder.
This finding that ventricular enlargement is most prominent in those unaffected
relatives more likely to carry susceptibility genes for schizophrenia is supported
by the Maudsley Family Study, which used high-resolution MRI and volumet-
ric assessments of ventricular size (McDonald et al. 2002; Sharma et al., 1998).
McDonald et al. (2002) analysed MRI measurements that were performed on
66 subjects with schizophrenia, 96 of their unaffected first-degree relatives, and
68 controls. A gradient of likely genetic risk was provided by these families since the
patients were divided into those from multiply affected families and those with no
family history of psychosis. The relatives of patients from multiply affected families
included a group of parents who were considered especially likely to be unaffected
gene carriers (“presumed obligate carriers”), since they appeared to transmit the
liability for schizophrenia to their affected children by virtue of also having a sibling
and/or parent affected. The patients had significantly enlarged lateral ventricles, and
so did the presumed obligate carriers, whereas those relatives from singly affected
families had similar ventricular volume to the controls. Other MRI studies do
not report significant lateral ventricular enlargement in relatives of schizophrenia
patients (Cannon et al., 1998; Lawrie et al., 1999; Seidman et al., 1999; Staal et al.,
2000), although mild ventricular enlargement that falls short of significance is often
145 Magnetic resonance imaging and phenotypes
noted. Since these studies did not focus on relatives from multiply affected families,
the lack of significant results is unsurprising. The finding that ventricular volume
enlargement is present only in those relatives most likely to carry susceptibility genes
is consistent with data from another MRI study on offspring, which reported no
overall significant difference in ventricular volume between relatives and controls,
but increasing ventricular enlargement in unaffected relatives with a stronger fam-
ily history of schizophrenia (Lawrie et al., 1999). There is also evidence from twin
studies that non-genetic factors such as obstetric complications contribute to larger
lateral ventricles in schizophrenia (Baare et al., 2001b; McNeil et al., 2000; Reveley
et al., 1984b; Suddath et al., 1990). Recent family studies point to an interaction
between genetic risk for schizophrenia and hypoxic birth events upon ventricular
volume, since subjects most likely to be gene carriers demonstrate the greatest ven-
tricular enlargement in association with obstetric complications (Cannon et al.,
1993, 2002a; McDonald et al., 2002).
Third ventricular volume tends to correlate with lateral ventricular volume, and
several studies on relatives have found that third ventricular volume is increased in
size (Keshavan et al., 1997; Lawrie et al., 2001; Seidman et al., 1999; Staal et al., 2000).
As with lateral ventricular volume, there is evidence that third ventricular volume
increases with likelihood of carrying susceptibility genes in unaffected relatives
(Lawrie et al., 1999; McDonald et al., 2002).
Subcortical structures
Studies of unaffected siblings and offspring have found the thalamus to be reduced
in volume compared with controls (Lawrie et al., 2001; Seidman et al., 1999; Staal
et al., 1998), consistent with the increased volume of the third ventricle identified
in the same studies. There was little evidence for abnormalities of the caudate or
lentiform nuclei from these studies (Lawrie et al., 2001; Seidman et al., 1999; Staal
et al., 2000).
Temporal lobe
No evidence has been reported in unaffected relatives for abnormality of the total
temporal lobe volume (Lawrie et al., 2001; McDonald et al., 2002; Schreiber et al.,
1999) nor for the superior temporal gyrus (Frangou et al., 1997b). However, several
groups have reported that the amygdala-hippocampal formation or the hippocam-
pus is reduced in volume in unaffected relatives or offspring (Baare et al., 2001b;
O’Driscoll et al., 2001; Schreiber et al., 1999; Steel et al., 2002; van Erp et al., 2002),
especially on the left (Lawrie et al., 2001, Keshavan et al., 2002; Seidman et al., 2002).
There have also been negative studies (Staal et al., 2000) and one study reporting
an enlarged hippocampus in those relatives likely to be transmitting genetic risk
(Harris et al., 2002). Schulze et al. (2003) also failed to detect reduced volume of the
hippocampus in unaffected relatives who took part in the Maudsley Family Study
but found that a history of obstetric complications was associated with reduced
volume of the left hippocampus. Evidence for a gene – environment interaction
for birth complications upon hippocampal volume, similar to that found for ven-
tricular volume in the same sample, was provided by van Erp et al. (2002), who
found that fetal hypoxia was associated with reduced volume of the hippocampus
in probands only and not in their siblings or controls.
147 Magnetic resonance imaging and phenotypes
Frontal lobe
No evidence has been reported for abnormality of the prefrontal lobe comparing
groups of unaffected siblings or offspring of schizophrenia subjects with controls
(Keshavan et al., 2002; Lawrie et al., 2001; Schreiber et al., 1999; Staal et al., 2000).
However Baare et al. (2001b) reported that both patients with schizophrenia from
discordant MZ twin pairs and their co-twins display decrements in frontal lobe
volume compared with normal twins whereas DZ discordant twin pairs do not,
strongly suggestive of a genetic etiology. Furthermore, although no group differ-
ences were found, Lawrie et al. (2001) reported that the prefrontal lobe volume
reduces as likelihood of carrying susceptibility genes increases among high risk
adolescents. Cannon et al. (2002b), in a computational morphometry study of
three-dimensional cortical maps from the MRI scans of 10 MZ and 10 DZ twin
pairs discordant for schizophrenia and 20 matched control twin pairs from a Finnish
cohort, reported evidence for reduced volume predominantly of the anterior and
superior prefrontal cortex in accordance with likely genetic risk among unaffected
twins.
Other structures
Sharma et al. (1999) reported that unaffected relatives from multiply affected fami-
lies had loss of normal fronto-occipital brain asymmetries compared with controls,
to a lesser degree than that found in affected probands, although a subsequent
study from the same Maudsley group, using a similar design, failed to replicate this
finding (Chapple et al., 2004). There is no evidence for abnormal volume of the
cerebellum in unaffected relatives compared with controls (McDonald et al., 2002;
Seidman et al., 1999; Staal et al., 2000) nor for midsagittal area reduction of the
corpus callosum (Chua et al., 2000; Narr et al., 2002). In the Finnish twin study,
significant vertical displacement of the corpus callosum (which was associated with
lateral ventricular enlargement) was found in the unaffected co-twins as well as in
the co-twins affected with schizophrenia in MZ twins but was not found in unaf-
fected DZ twins, suggesting that this structural deviation also represents a genetic
marker for schizophrenia (Narr et al., 2002).
The true nature of the morphological phenotype of schizophrenia is likely to
involve more widespread networks than those chosen for analysis by the region-
of-interest studies performed to date on unaffected relatives, and future studies
using whole brain analysis are likely to be helpful in more comprehensively inves-
tigating these. In a recent paper, Faraone et al. (2003) studied volumetric measure-
ments of the whole brain divided into parcellation units in relatives of patients
with schizophrenia and in controls. They identified three factors loaded by several
parcellation units throughout the neocortical ribbon as well as in subcortical and
limbic regions that could be used to describe an MRI phenotype which succeeded
148 C. McDonald and R. M. Murray
in discriminating relatives from controls and was most deviant in relatives from
more densely affected families.
The varying morphometric deviations reported in unaffected relatives most likely
reflect the effect of differing study designs, samples, and methodologies, as well as
the limited statistical power associated with many of the studies: since deviations in
relatives tend to be even more subtle than those found in probands, larger numbers
of subjects are required to demonstrate a statistically significant result in studies
of relatives than when comparing only probands with controls. Despite these dis-
agreements, work to date in this field does demonstrate that unaffected relatives
frequently display abnormalities of brain structure compared with controls that are
similar in nature to, but less severe than, those associated with schizophrenia. The
greatest evidence to date is for increased ventricular volume; reduced gray matter
volume, especially in frontotemporal regions; reduced volume of the thalamus; and
reduced volume of the amygdala–hippocampal complex. These “familial” abnor-
malities identified in probands and unaffected relatives are presumably genetic in
etiology but could also represent the impact of shared environmental factors. Fur-
ther evidence for the likely genetic origin of such traits can be inferred if they are
found most commonly in those relatives who are more likely to be gene carriers.
Studies that have examined likely genetic loading of unaffected relatives, based on
number of affected relatives or genetic relatedness to the proband, have generally
found the structural abnormalities in unaffected relatives to be more prominent
in those with a higher genetic loading (Cannon et al., 1993, 2002b; Faraone et al.,
2003; Lawrie et al., 1999, 2001; McDonald et al., 2002; Seidman et al., 2002). This
finding of increased impairment in relatives in accordance with their genetic load-
ing is in keeping with the predictions of the multifactorial model for transmis-
sion of the illness. Given the impact that the known environmental risk factor of
obstetric complications may have on ventricular and hippocampal volume, which
may act interactively with susceptibility genes, future studies should collect such
information where feasible.
Molecular genetics
Few published studies to date have attempted to link measures of brain morphology
in schizophrenia with specific genetic factors. Shihabuddin et al. (1996) found that
those family members with a marker allele on chromosome 5p14.1–13.1, which
had previously been linked to schizophrenia-related disorders in a single pedigree,
had larger ventricle-to-brain ratios and greater fronto-parietal sulcal atrophy than
those who lacked the marker allele. Other studies have examined relationships
between different allele frequencies of polymorphisms in genes likely to have a
role in neurodevelopment and MRI morphometric measures. In a small sample of
patients with psychosis, Kunugi et al. (1999) reported that hippocampal volume
149 Magnetic resonance imaging and phenotypes
was reduced in patients with schizophrenia who were carriers, compared with
non-carriers, of the A3 allele of a dinucleotide repeat polymorphism in the neu-
rotrophin 3 gene. Wassink and colleagues (1999, 2000) examined the relationship
between brain morphometry and alleles of polymorphisms in the genes for brain-
derived neurotrophic factor and tumor necrosis factor receptor II in patients with
schizophrenia, their parents, and controls. The polymorphisms were not related to
the clinical schizophrenia phenotype, but subjects with at least one copy of “allele
1” of the brain-derived neurotrophic factor polymorphism had larger parietal lobes
than those who did not (Wassink et al., 1999). Subjects with schizophrenia who were
homozygous for “allele 1” of the tumor neurosis factor receptor II polymorphism
had larger ventricles and smaller frontal lobes than subjects with at least one copy
of “allele 2” (Wassink et al., 2000). Meisenzahl et al. (2001) examined the associa-
tion of MRI measurements from a sample of male patients with schizophrenia and
controls with a polymorphism in the promoter region of the gene for interleukin-
1β. The polymorphism had no influence on brain morphology in controls, but
schizophrenia patients who were “allele 2” carriers of the polymorphism had gray
matter volume deficits in frontotemporal regions and generalized white matter
deficits compared with non-carriers. In a similar sample, Rujescu et al. (2002)
examined a common biallelic polymorphism, Met129Val, in the gene coding the
prion protein (PRNP). Allele frequencies did not differ between patients and con-
trols, but homozygosity for methionine was significantly associated with decreased
white matter volume and enlarged CSF volume in the subjects, independent of
diagnosis.
The results of these molecular genetic studies must be considered very tentative
in the absence of replication. However, they demonstrate the value of using brain
morphometry as an alternative phenotype in genetic studies of schizophrenia and
suggest that a richer understanding of the actions of potential susceptibility genes
could be achieved through such a design.
Conclusions
The study of the relationship between genetics and neuroimaging represents an
emerging field of research in complex neuropsychiatric disorders. Further studies
using more advanced methodology are likely to help to clarify the brain struc-
tural manifestations of schizophrenia susceptibility genes. Research to date indi-
cates that there is a high degree of genetic control over brain morphometry, that
schizophrenia is associated with several subtle deviations of brain morphology,
which are frequently neurodevelopmental in origin, and that unaffected relatives
also demonstrate some of these deviations. The brain morphological phenotype of
schizophrenia is likely to be increasingly used as a valuable alternative to the clinical
150 C. McDonald and R. M. Murray
phenotype in molecular genetic studies and should help to simplify the genetic and
biological complexity of the illness, and thus facilitate the search for susceptibility
genes.
REFERENCES
Ananth, H., Popescu, I., Critchley, H. D., et al. (2002). Cortical and subcortical gray matter
abnormalities in schizophrenia determined through structural magnetic resonance imaging
with optimized volumetric voxel-based morphometry. Am J Psychiatry 159: 1497–1505.
Baare, W. F., Hulshoff Pol, H. E., Boomsma, D. I., et al. (2001a). Quantitative genetic modeling
of variation in human brain morphology. Cereb Cortex 11: 816–824.
Baare, W. F., van Oel, C. J., Hulshoff Pol, H. E., et al. (2001b). Volumes of brain structures in
twins discordant for schizophrenia. Arch Gen Psychiatry 58: 33–40.
Bartley, A. J., Jones, D. W., Weinberger, D. R. (1997). Genetic variability of human brain size and
cortical gyral patterns. Brain 120: 257–269.
Bilder, R. M., Wu, H., Bogerts, B., et al. (1994). Absence of regional hemispheric volume asym-
metries in first-episode schizophrenia. Am J Psychiatry 151: 1437–1447.
Cannon, T. D., Mednick, S. A., Parnas, J., et al. (1993). Developmental brain abnormalities in the
offspring of schizophrenic mothers. I. Contributions of genetic and perinatal factors. Arch
Gen Psychiatry 50: 551–564.
Cannon, T. D., Zorrilla, L. E., Shtasel, D., et al. (1994). Neuropsychological functioning in siblings
discordant for schizophrenia and healthy volunteers. Arch Gen Psychiatry 51: 651–661.
Cannon, T. D., van Erp, T. G. M., Huttunen, M., et al. (1998). Regional gray matter, white matter,
and cerebrospinal fluid distributions in schizophrenic patients, their siblings, and controls.
Arch Gen Psychiatry 55: 1084–1091.
Cannon, T. D., van Erp, T. G., Rosso, I. M., et al. (2002a). Fetal hypoxia and structural brain
abnormalities in schizophrenic patients, their siblings, and controls. Arch Gen Psychiatry 59:
35–41.
Cannon, T. D., Thompson, P. M., van Erp, T. G., et al. (2002b). Cortex mapping reveals regionally
specific patterns of genetic and disease-specific gray-matter deficits in twins discordant for
schizophrenia. Proc Natl Acad Sci USA 99: 3228–3233.
Cardno, A. G., Marshall, E. J., Coid, B., et al. (1999). Heritability estimates for psychotic disorders:
the Maudsley twin psychosis series. Arch Gen Psychiatry 56: 162–168.
Carmelli, D., DeCarli, C., Swan, G. E., et al. (1998). Evidence for genetic variance in white matter
hyperintensity volume in normal elderly male twins. Stroke 29: 1177–1181.
Carmelli, D., Swan, G. E., DeCarli, C., et al. (2002). Quantitative genetic modeling of regional
brain volumes and cognitive performance in older male twins. Biol Psychol 61: 139–155.
Chakos, M. H., Lieberman, J. A., Alvir, J., et al. (1995). Caudate nuclei volumes in schizophrenic
patients treated with typical antipsychotics or clozapine. Lancet 345: 456–457.
Chapple, B., Grech A., Sham, P., et al. (2004). Normal cerebral asymmetry in familial and non-
familial schizophrenic probands and their unaffected relatives. Schizophr Res 67: 33–40.
151 Magnetic resonance imaging and phenotypes
Chua, S. E., Sharma, T., Takei, N., et al. (2000). A magnetic resonance imaging study of cor-
pus callosum size in familial schizophrenic subjects, their relatives, and normal controls.
Schizophr Res 41: 397–403.
Crawford, T. J., Sharma, T., Puri, B. K., et al. (1998). Saccadic eye movements in families multiply
affected with schizophrenia: the Maudsley Family Study. Am J Psychiatry 155: 1703–1710.
DeLisi, L. E., Goldin, L. R., Hamovit, J. R., et al. (1986). A family study of the association of
increased ventricular size with schizophrenia. Arch Gen Psychiatry 43: 148–153.
DeLisi, L. E., Hoff, A. L., Schwartz, J. E., et al. (1991). Brain morphology in first-episode
schizophrenic-like psychotic patients: a quantitative magnetic resonance imaging study.
Biol Psychiatry 29: 159–175.
Falkai, P., Bogerts, B., Schneider, T., et al. (1995). Disturbed planum temporale asymmetry in
schizophrenia. A quantitative post-mortem study. Schizophr Res 14: 161–176.
Fannon, D., Chitnis, X., Doku, V., et al. (2000). Features of structural brain abnormality detected
in first-episode psychosis. Am J Psychiatry 157: 1829–1834.
Faraone, S. V., Seidman, L. J., Kremen, W. S., et al. (2003). Structural brain abnormalities among
relatives of patients with schizophrenia: implications for linkage studies. Schizophr Res 60:
125–140.
Frangou, S., Sharma, T., Alarcon, G., et al. (1997a). The Maudsley Family Study, II: endogenous
event-related potentials in familial schizophrenia. Schizophr Res 23: 45–53.
Frangou, S., Sharma, T., Sigmudsson, T., et al. (1997b). The Maudsley Family Study IV. Normal
planum temporale asymmetry in familial schizophrenia – A volumetric MRI study. Br J
Psychiatry 170: 328–333.
Gershon, E. S. Goldin, L. R. (1986). Clinical methods in psychiatric genetics. I. Robustness of
genetic marker investigative strategies. Acta Psychiatr Scand 74: 113–118.
Goldstein, J. M., Goodman, J. M., Seidman, L. J., et al. (1999). Cortical abnormalities in
schizophrenia identified by structural magnetic resonance imaging. Arch Gen Psychiatry
56: 537–547.
Gottesman, I. I. (1991). Schizophrenia Genesis: The Origins of Madness. New York: H Freeman.
Gur, R. E., Cowell, P., Turetsky, B. I., et al. (1998). A follow-up magnetic resonance imaging study
of schizophrenia. Relationship of neuroanatomical changes to clinical and neurobehavioral
measures. Arch Gen Psychiatry 55: 145–152.
Harris, J. G., Young, D. A., Rojas, D. C., et al. (2002). Increased hippocampal volume in
schizophrenics’ parents with ancestral history of schizophrenia. Schizophr Res 55: 11–17.
Harrison, P. J. (1999). The neuropathology of schizophrenia: a critical review of the data and
their interpretation. Brain 122: 593–624.
Hirayasu, Y., Shenton, M. E., Salisbury, D. F., et al. (1998). Lower left temporal lobe MRI volumes
in patients with first-episode schizophrenia compared with psychotic patients with first-
episode affective disorder and normal subjects. Am J Psychiatry 155: 1384–1391.
Holzman, P. S., Kringlen, E., Matthysse, S., et al. (1988). A single dominant gene can account
for eye tracking dysfunctions and schizophrenia in offspring of discordant twins. Arch Gen
Psychiatry 45: 641–647.
Jones, P., Murray, R. M. (1991). The genetics of schizophrenia is the genetics of neurodevelopment.
Br J Psychiatry 158: 615–623.
152 C. McDonald and R. M. Murray
Keshavan, M. S., Montrose, D. M., Pierri, J. N., et al. (1997). Magnetic resonance imaging and
spectroscopy in offspring at risk for schizophrenia: Preliminary studies. Prog Neuropsy-
chopharmacol Biol Psychiatry 21: 1285–1295.
Keshavan, M. S., Dick, E., Mankowski, I., et al. (2002). Decreased left amygdala and hippocampal
volumes in young offspring at risk for schizophrenia. Schizophr Res, 58: 173–183.
Kunugi, H., Hattori, M., Nanko, S., et al. (1999). Dinucleotide repeat polymorphism in the
neurotrophin-3 gene and hippocampal volume in psychoses. Schizophr Res 37: 271–273.
Lawrie, S. M., Whalley, H., Kestelman, J. N., et al. (1999). Magnetic resonance imaging of brain
in people at high risk of developing schizophrenia. Lancet 353: 30–33.
Lawrie, S. M., Whalley, H. C., Abukmeil, S. S., et al. (2001). Brain structure, genetic liability, and
psychotic symptoms in subjects at high risk of developing schizophrenia. Biol Psychiatry 49:
811–823.
Leboyer, M., Bellivier, F., Nosten-Bertrand, M., et al. (1998). Psychiatric genetics: search for
phenotypes. Trends Neurosci 21: 102–105.
Lewis, S. W. (1990). Computerised tomography in schizophrenia 15 years on. Br J Psychiatry
9(Suppl.): 16–24.
Lieberman, J., Chakos, M., Wu, H., et al. (2001). Longitudinal study of brain morphology in first
episode schizophrenia. Biol Psychiatry 49: 487–499.
McCarley, R. W., Wible, C. G., Frumin, M., et al. (1999). MRI anatomy of schizophrenia. Biol
Psychiatry 45: 1099–1119.
McDonald, C., Grech, A., Toulopoulou, T., et al. (2002). Brain volumes in familial and non-familial
schizophrenic probands and their unaffected relatives. Am J Med Genet 114: 616–625.
McGrath, J., Murray, R. (1995). Risk factors for schizophrenia: from conception to birth. In
Schizophrenia, ed. S. Hirsch, D. R., Weinberger. Oxford: Blackwell Scientific, pp. 187–205.
McNeil, T. F., Cantor-Graae, E., Weinberger, D. R. (2000). Relationship of obstetric complica-
tions and differences in size of brain structures in monozygotic twin pairs discordant for
schizophrenia. Am J Psychiatry 157: 203–212.
Meisenzahl, E. M., Rujescu, D., Kirner, A., et al. (2001). Association of an interleukin-1beta
genetic polymorphism with altered brain structure in patients with schizophrenia. Am J
Psychiatry 158: 1316–1319.
Murray, R. M., Lewis, S. W., Reveley, A. M. (1985). Towards an aetiological classification of
schizophrenia. Lancet i: 1023–1026.
Narr, K. L., Cannon, T. D., Woods, R. P., et al. (2002). Genetic contributions to altered callosal
morphology in schizophrenia. J Neurosci 22: 3720–3729.
Noga, J. T., Bartley, A. J., Jones, D. W., et al. (1996). Cortical gyral anatomy and gross brain
dimensions in monozygotic twins discordant for schizophrenia. Schizophr Res 22: 27–40.
Nosarti, C., Al-Asady, M. H., Frangou, S., et al. (2002). Adolescents who were born very preterm
have decreased brain volumes. Brain 125: 1616–1623.
O’Driscoll, G. A., Florencio, P. S., Gagnon, D., et al. (2001). Amygdala-hippocampal volume and
verbal memory in first-degree relatives of schizophrenic patients. Psychiatry Res 107: 75–85.
Oppenheim, J. S., Skerry, J. E., Tramo, M. J., et al. (1989). Magnetic resonance imaging morphol-
ogy of the corpus callosum in monozygotic twins. Ann Neurol 26: 100–104.
153 Magnetic resonance imaging and phenotypes
Pennington, B. F., Filipek, P. A., Lefly, D., et al. (2000). A twin MRI study of size variations in
human brain. J Cogn Neurosci 12: 223–232.
Pfefferbaum, A., Sullivan, E. V., Swan, G. E., et al. (2000). Brain structure in men remains highly
heritable in the seventh and eighth decades of life. Neurobiol Aging 21: 63–74.
Reveley, A. M., Reveley, M. A., Clifford, C. A., et al. (1982). Cerebral ventricular size in twins
discordant for schizophrenia. Lancet i: 540–541.
Reveley, A. M., Reveley, M. A., Chitkara, B., et al. (1984a). The genetic basis of cerebral ventricular
volume. Psychiatr Res 13: 261–266.
Reveley, A. M., Reveley, M. A., Murray, R. M. (1984b). Cerebral ventricular enlargement in
non-genetic schizophrenia: a controlled twin study. Br J Psychiatry 144: 89–93.
Rujescu, D., Meisenzahl, E. M., Giegling, I., et al. (2002). Methionine homozygosity at codon
129 in the prion protein is associated with white matter reduction and enlargement of CSF
compartments in healthy volunteers and schizophrenic patients. Neuroimage 15: 200–206.
Rutherford, M. C., Pennock, J. M., Schwieso, J. E., et al. (1995). Hypoxic ischaemic encephalopa-
thy: early magnetic resonance findings and their evolution. Neuropediatrics 26: 183–191.
Schreiber, H., Baur-Seack, K., Kornhuber, H. H., et al. (1999). Brain morphology in adolescents
at genetic risk for schizophrenia assessed by qualitative and quantitative magnetic resonance
imaging. Schizophr Res 40: 81–84.
Schulze, K., McDonald, C., Frangou, S., et al. (2003). Hippocampal volume in familial and
nonfamilial schizophrenic probands and their unaffected relatives. Biol Psychiatry 53: 562–
570.
Seidman, L. J., Faraone, S. V., Goldstein, J. M., et al. (1999). Thalamic and amygdala–hippocampal
volume reductions in first-degree relatives of patients with schizophrenia: an MRI-based
morphometric analysis. Biol Psychiatry 46: 941–954.
Seidman, L. J., Faraone, S. V., Goldstein, J. M., et al. (2002). Left hippocampal volume as a
vulnerability indicator for schizophrenia: a magnetic resonance imaging morphometric
study of nonpsychotic first- degree relatives. Arch Gen Psychiatry 59: 839–849.
Sharma, T., Lancaster, E., Lee, D., et al. (1998). Brain changes in schizophrenia – volumetric MRI
study of families multiply affected with schizophrenia: The Maudsley Family Study 5. Br J
Psychiatry 173: 132–138.
Sharma, T., Lancaster, E., Sigmundsson, T., et al. (1999). Lack of normal pattern of cerebral
asymmetry in familial schizophrenic patients and their relatives: the Maudsley Family Study.
Schizophr Res 40: 111–120.
Shihabuddin, L., Silverman, J. M., Buchsbaum, M. S., et al. (1996). Ventricular enlargement
associated with linkage marker for schizophrenia-related disorders in one pedigree. Mol
Psychiatry 1: 215–222.
Siegel, C., Waldo, M., Mizner, G., et al. (1984). Deficits in sensory gating in schizophrenic patients
and their relatives. Evidence obtained with auditory evoked responses. Arch Gen Psychiatry
41: 607–612.
Sigmundsson, T., Suckling, J., Maier, M., et al. (2001). Structural abnormalities in frontal, tem-
poral, and limbic regions and interconnecting white matter tracts in schizophrenic patients
with prominent negative symptoms. Am J Psychiatry 158: 234–243.
154 C. McDonald and R. M. Murray
Silverman, J. M., Smith, C. J., Guo, S. L., et al. (1998). Lateral ventricular enlargement
in schizophrenic probands and their siblings with schizophrenia-related disorders. Biol
Psychiatry 43: 97–106.
Staal, W. G., Hulshoff, H. E., Schnack, H., et al. (1998). Partial volume decrease of the thalamus
in relatives of patients with schizophrenia. Am J Psychiatry 155: 1784–1786.
Staal, W. G., Pol, H. E. H., Schnack, H. G., et al. (2000). Structural brain abnormalities in patients
with schizophrenia and their healthy siblings. Am J Psychiatry 157: 416–421.
Steel, R. M., Whalley, H. C., Miller, P., et al. (2002). Structural MRI of the brain in presumed
carriers of genes for schizophrenia, their affected and unaffected siblings. J Neurol Neurosurg
Psychiatry 72: 455–458.
Stefanis, N., Frangou, S., Yakeley, J., et al. (1999). Hippocampal volume reduction in schizophre-
nia: effects of genetic risk and pregnancy and birth complications. Biol Psychiatry 46: 697–
702.
Stefansson, H., Sigurdsson, E., Steinthorsdottir, V., et al. (2002). Neuregulin 1 and susceptibility
to schizophrenia. Am J Hum Genet 71: 877–892.
Steinmetz, H., Herzog, A., Huang, Y., et al. (1994). Discordant brain-surface anatomy in monozy-
gotic twins. N Engl J Med 331: 951–952.
Steinmetz, H., Herzog, A., Schlaug, G., et al. (1995). Brain asymmetry in monozygotic twins.
Cereb Cortex 5: 296–300.
Straub, R. E., Jiang, Y., MacLean, C. J., et al. (2002). Genetic variation in the 6p22.3 gene DTNBP1,
the human ortholog of the mouse dysbindin gene, is associated with schizophrenia. Am J
Hum Genet 71: 337–348.
Suddath, R. L., Christison, G. W., Torrey, E. F., et al. (1990). Anatomical abnormalities in the
brains of monozygotic twins discordant for schizophrenia. N Engl J Med 322: 789–794.
Sullivan, E. V., Pfefferbaum, A., Swan, G. E., et al. (2001). Heritability of hippocampal size
in elderly twin men: equivalent influence from genes and environment. Hippocampus 11:
754–762.
Suzuki, M., Nohara, S., Hagino, H., et al. (2002). Regional changes in brain gray and white matter
in patients with schizophrenia demonstrated with voxel-based analysis of MRI. Schizophr
Res 55: 41–54.
Thompson, P. M., Cannon, T. D., Narr, K. L., et al. (2001). Genetic influences on brain structure.
Nat Neurosci 4: 1253–1258.
Toulopoulou, T., Morris, R. G., Rabe-Hesketh, S., et al. (2003). Selectivity of verbal memory
deficit in schizophrenic patients and their relatives. Am J Med Genet 116: 1–7.
Tramo, M. J., Loftus, W. C., Stukel, T. A., et al. (1998). Brain size, head size, and intelligence
quotient in monozygotic twins. Neurology 50: 1246–1252.
van Erp, T. G., Saleh, P. A., Rosso, I. M., et al. (2002). Contributions of genetic risk and fetal
hypoxia to hippocampal volume in patients with schizophrenia or schizoaffective disorder,
their unaffected siblings, and healthy unrelated volunteers. Am J Psychiatry 159: 1514–1520.
Vogeley, K., Tepest, R., Pfeiffer, U., et al. (2001). Right frontal hypergyria differentiation in affected
and unaffected siblings from families multiply affected with schizophrenia: a morphometric
MRI study. Am J Psychiatry 158: 494–496.
155 Magnetic resonance imaging and phenotypes
Wassink, T. H., Nelson, J. J., Crowe, R. R., et al. (1999). Heritability of BDNF alleles and their
effect on brain morphology in schizophrenia. Am J Med Genet 88: 724–728.
Wassink, T. H., Crowe, R. R., Andreasen, N. C. (2000). Tumor necrosis factor receptor-II: heri-
tability and effect on brain morphology in schizophrenia. Mol Psychiatry 5: 678–682.
Weinberger, D. R., McClure, R. K. (2002). Neurotoxicity, neuroplasticity, and magnetic resonance
imaging morphometry: what is happening in the schizophrenic brain? Arch Gen Psychiatry
59: 553–558.
Weinberger, D. R., DeLisi, L. E., Neophytides, A. N., et al. (1981). Familial aspects of CT scan
abnormalities in chronic schizophrenic patients. Psychiatry Res 4: 65–71.
White, T., Andreasen, N. C., Nopoulos, P. (2002). Brain volumes and surface morphology in
monozygotic twins. Cereb Cortex 12: 486–493.
Wickham, H. Murray, R. M. (1997). Can biological markers identify endophenotypes predispos-
ing to schizophrenia? Int Rev Psychiatry 9: 355–364.
Wilke, M., Kaufmann, C., Grabner, A., et al. (2001). Gray matter-changes and correlates of disease
severity in schizophrenia: a statistical parametric mapping study. Neuroimage 13: 814–824.
Wright, I. C., Ellison, Z. R., Sharma, T., et al. (1999). Mapping of grey matter changes in
schizophrenia. Schizophr Res 35: 1–14.
Wright, I. C., Rabe-Hesketh, S., Woodruff, P. W. R., et al. (2000). Meta-analysis of regional brain
volumes in schizophrenia. Am J Psychiatry 157: 16–25.
Wright, I. C., Sham, P., Murray, R. M., et al. (2002). Genetic contributions to regional variability
in human brain structure: methods and preliminary results. Neuroimage 17: 256–271.
Zorrilla, L. T., Cannon, T. D., Kronenberg, S., et al. (1997). Structural brain abnormalities in
schizophrenia: a family study. Biol Psychiatry 42: 1080–1086.
9
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
156
157 Nutritional factors and schizophrenia
to serious behavioral disturbances, which cause the patient major suffering and
disability.
At this stage, the role of nutrition in the etiology of schizophrenia is circumstan-
tial and speculative; however, nutrition may play a critical role in its variable course
and outcome. Large nutritional differences, resulting from the quality and quan-
tity of food available, exist between cultures and socioeconomic classes (Mahadik
et al., 1999a). Cultures evolve by selection of food that supports the healthy body
and mind. An editorial by Murphy (1984) suggested that diseases such as general
paresis, peptic ulcer, and schizophrenia could be caused as well as cured by civiliza-
tion. Civilization, which is conceived through a process rather than established as a
state, combines single human individuals into one great unity (Freud, 1961). Torrey
(1980) has suggested a close correlation between the prevalence of schizophrenia
and the degree to which a civilization has advanced; he favored a viral theory
of the etiology of schizophrenia, since specific types of virus can be propagated,
modified, and transmitted from generation to generation. Others have blamed
the diet of a civilization for the diseases it suffers. The pathogenic factors that cause
these diseases may be incidental, rather than an essential aspect of civilization. The
nutrition, lifestyle, and viruses are unique and essential aspects of each culture
and socioeconomic class. However, since the incidence of schizophrenia is similar
in all cultures, while the course and outcome vary widely (see below), nutritional
factors may influence the underlying pathophysiology of the disease to varying
degrees.
Multinational studies coordinated by the World Health Organization (WHO)
have found that the incidence, prevalence, and manifestation of schizophrenia are
similar across different countries and cultures, but the course and outcome of the
disease vary widely from mild in developing countries to chronic in developed
countries (Jablensky et al., 1991; Sartorius et al., 1986; WHO, 1973). These findings
have led to the notion of a “universality” of schizophrenia prevalence but variability
in outcome. Several studies also have shown that the outcome of schizophrenia is
related to the socioeconomic class of the patient within a culture (Eaton, 1985;
Nandi et al., 1980). Several variables, such as social stigma, family support, type of
treatment, and family history, examined in the WHO studies could not satisfac-
torily account for the cross-national/cross-cultural variability in outcome (Craig
et al., 1997; Edgerton and Cohen, 1994; Karno and Jenkins, 1993). Recently, the
influence of culture and socioeconomic classes on various facets (i.e. epidemiol-
ogy, etiology, phenomenology, course, and outcome) of schizophrenia has been
discussed extensively (Kulhara and Chakrabarti, 2001). In addition to these factors,
viral infections, famines, and season of birth, which have all been found to be asso-
ciated with schizophrenia and its course and outcome, can significantly affect the
availability and metabolism of essential nutrients during the early stages of human
158 S. P. Mahadik
development (Horrobin et al., 1994; Mahadik et al., 1999a). Therefore, this chapter
will focus primarily on the role of essential nutritional factors such as amino acids,
lipids, vitamins, and minerals on the course and outcome of schizophrenia.
Furthermore, studies indicate that the need for these nutrients has increased because
of significant changes in lifestyle during this period. Therefore, dietary availability
of essential amino acids and most vitamins and minerals may not be relevant to
schizophrenia. Nevertheless, a low prenatal vitamin D hypothesis for schizophrenia
has been proposed (McGrath, 1999). However, since this hypothesis is primarily
based on the higher prevalence of schizophrenia among individuals born in winter
months or at higher latitudes, this may still be related to some other dietary factors
(e.g. EFAs and antioxidants) that are reduced under these circumstances (Mahadik
et al., 1999a). Furthermore malnutrition is probably not related to defective utiliza-
tion (absorption, transport, and incorporation into biologically active molecules)
of amino acids, vitamins, and minerals, since such defects are often fatal. This brings
us to the primary issue of dietary availability of essential lipids, specifically essential
polyunsaturated fatty acids (EPUFAs) and antioxidants. The former are needed
to synthesize membrane phospholipids, particularly neural membrane phospho-
lipids, which are highly enriched in EPUFAs (Horrocks et al., 1982). Antioxidants
are needed to protect these EPUFAs from alteration by free radicals (Mahadik and
Gowda, 1996; Mahadik et al., 2001).
Since the 1950s, schizophrenia has been treated with a wide range of antipsy-
chotic drugs, and these are going to be the drugs of choice for the treatment of
schizophrenia for a long time. However, Khan et al. (2001) recently analyzed the
full FDA database on clinical trials of antipsychotic drugs and reported that the aver-
age reduction in symptoms was 17.3% when a conventional antipsychotic drug was
administered, and 16.6% when a newer antipsychotic drug was administered. This,
and the earlier finding by Hegarty et al. (1994) that long-term favorable outcomes
in terms of employment and reintegration into the community do not significantly
differ from 100 years ago, suggests that current therapeutic strategies have limita-
tions in correcting the neuropathophysiology of schizophrenia and in improving
the psychopathology associated with the disease. In addition, antipsychotic treat-
ments, in general, are associated with numerous morbidities, including weight gain,
insulin resistance, cardiovascular problems, and abnormal lipid metabolism, which
result in a diminished quality of life and even in increased mortality (McIntyre
et al., 2001). Increasing evidence suggests that dietary supplementation with selec-
tive essential nutritional factors, such as, essential fatty acids and antioxidants, may
significantly improve the course and outcome of schizophrenia (Fenton et al., 2000;
Horrobin, 1998; Mahadik and Evans, 1997; Mahadik and Gowda, 1996; Mahadik
and Scheffer, 1996; Mahadik et al., 2001; Reddy and Yao, 1996).
and carbon-3 chemical double bonds on each molecule, respectively. Algae and
plants exclusively make these EPUFAs. All animals actually consume the precursors
of EPUFAs (linolenic acid [LA; C18:2n6] and α linolenic acid [ALA; C18:3n3] or
their predominant functional analogs (arachidonic acid [AA] and docosahexaenoic
acid [DHA], respectively). AA and DHA make 50% of the fatty acids that are
attached to brain phospholipids, which comprise almost 60% of brain mass (Suzuki,
1981).
acid) (Axelrod, 1990) and growth factors (Virdee et al., 1994) are mediated by these
mechanisms.
metabolism of EPUFAs (Mahadik et al., 2001). However, since the 1970s with
increased migration of population and very significant changes in food production
and delivery, some of the influences of season, culture, and family-related factors
are being attenuated.
Adjunctive medication
Typical antipsychotic drugs such as haloperidol have pro-oxidant properties
(Jeding et al., 1995). These medications can also affect EPUFA metabolism
(Horrobin et al., 1994). The newer atypical antipsychotic drugs have antioxidant
effects, particularly clozapine, which has been found to improve membrane EPUFA
levels (Arvindakshan et al., 2003a; Horrobin, 1999; Khan et al., 2002). Recently, we
have found that haloperidol reduced the antioxidant enzymes and increased the
lipid peroxides in the rat brain; however, none of the atypical drugs such as cloza-
pine, olanzapine, and risperidone did this (Parikh et al., 2002).
Duration of treatment
Earlier studies have suggested that supplementation for at least 4 months is required
to restore the level of EPUFA in red blood cells and brain membranes to a steady state
(Mahadik and Evans, 1997). However, one study indicated that supplementation
with EPA had therapeutic effects within a few weeks, indicating a possible indirect
role for EPA and/or its metabolites (Peet and Horrobin, 2002).
Placebo control
Since psychiatric disorders show a significant placebo effect on symptom reduction,
well-designed, placebo-controlled, dose-ranging replication studies would be crit-
ical to establish the therapeutic benefits of omega-3 EPUFAs (Peet and Horrobin,
2002).
Conclusions
Since proper nutrition regulates the normal development of body and mind, and
maintains their health throughout a person’s life, it can play a critical role in influ-
encing the pathophysiology, and thereby the psychopathology, of schizophrenia.
However, based on global epidemiological data, nutrition can primarily be con-
sidered as a means of improving the course and outcome of the disease. Proper
nutrition is a complex issue, particularly for schizophrenia patients, whose dietary
intake can be affected by lifestyle, illness, medications, support services, and other
yet unknown metabolic genetic defects. Reports on diet in developed countries such
as the USA indicate that there is malnutrition (lack or reduced intake) with respect to
dietary intake of omega-3 EPUFAs and antioxidants. This and the increased oxida-
tive stress and reduced levels of membrane phospholipid omega-3 EFAs from very
early stages of schizophrenia illness suggest that early nutritional supplementation
of omega-3 EFAs and antioxidants could significantly improve the clinical course
and outcome of patients; such supplementation may also potentially diminish the
167 Nutritional factors and schizophrenia
REFERENCES
Adler, L. A., Rotrosen, J., Edson, R., et al. (1999). Vitamin E treatment for tardive dyskinesia. Arch
J Psychiatry 56: 836–841.
Akbar, M., Kim, H. Y. (2002). Protective effects of docosahexaenoic acid in staurosporine-induced
apoptosis: involvement of phosphatidylinositol-3 kinase pathway. J Neurochem 82: 655–665.
Arnold, L. E., Kleykamp, D., Votolato, N., Gibson, R. A., Horrocks, L. (1994). Potential link
between dietary intake of fatty acids and behavior: pilot exploration of serum lipids in
attention-deficit hyperactivity disorder. J Child Adoles Psychopharmacol 4: 171–182.
Arvindakshan, M. (2003). The role of membrane essential fatty acids in schizophrenia outcome.
Ph.D. Thesis, Pune University India.
Arvindakshan, M., Sitasawad, S., Debsikdar, V., et al. (2003a). Membrane essential polyunsatu-
rated fatty acids (EPUFA) and schizophrenia outcome: EPUFA and lipid peroxide levels in
never-medicated and medicated schizophrenics. Biol Psychiatry 53: 56–64.
Arvindakshan, M., Ghate, M., Ranjekar, P. K., Evans, D.R., Mahadik, S. P. (2003b). Supplemen-
tation with a combination of omega-3 fatty acids and antioxidants (vitamins E and C)
improves the outcome of schizophrenia. Schizophr Res 62: 195–204.
Axelrod, J. (1990). Receptor-mediated activation of phospholipase A2 and arachidonic acid
release in signal transduction. Biochem Soc Trans 18: 503–507.
Berridge, M. J. (1981). Phosphatidylinositol hydrolysis: a multifunctional transducting mecha-
nism. Mol Cell Endocrinol 24: 115–140.
Bloom, F. (1993). Advancing neurodevelopmental origin for schizophrenia. Arch Gen Psychiatry
50: 224–227.
Brand, A., Gil, S., Yavin, E. (2000). N-Methyl bases of ethanolamine prevent apoptotic cell death
induced by oxidative stress in cells of oligodendroglia origin. J Neurochem 74: 1596–1604.
Brown, S., Birtwistle, J., Roe, L., Thompson, C. (1999). The unhealthy lifestyle of people with
schizophrenia. Psychol Med 29: 697–701.
Burr, G. O., Burr, M. M. (1929). A new deficiency disease produced by the rigid exclusion of fat
from the diet. J Biol Chem 82: 345–367.
Carlson, S. E., Rhodes, P. G., Ferguson, M. G. (1986). Docosahexaenoic acid status of preterm
infants at birth and following feeding with human milk or formula 1–3. Am J Clin Nutr 44:
798–804.
Chafetz, M. D. (1990). Nutrition and Neurotransmitters: The Nutrient Bases of Behavior. Engle-
wood Cliffs, NJ: Prentice Hall.
Christinsen, O., Christinsen, E. (1988). Fat consumption and schizophrenia. Acta Psychiatr Scand
78: 587–591.
Chua, S. E., McKenna, P. J. (1995). Schizophrenia: a brain disease? A critical review of structural
and cerebral abnormalities in the disorder. Br J Psychiatry 166: 563–582.
168 S. P. Mahadik
Coleman, M., Gillberg, C. (1996). The Schizophrenias. A Biological Approach to the Schizophrenia
Spectrum Disorders. New York: Springer.
Craig, T. J., Siegel, C., Hopper, K., Lin, S., Sartorius, N. (1997). Outcome in schizophrenia and
related disorders compared between developing and developed countries. Br J Psychiatry
170: 229–233.
Crawford, M. A. (1992). Essential fatty acids and neurodevelopmental disorder. In Neurobiology
of Essential Fatty Acids, ed. N. G. Bazan. New York: Plenum Press, pp. 307–314.
Dhopeshwarkar, G. A. (1983). Nutrition and Brain Development. New York: Plenum Press.
Eaton, W. W. (1985). Epidemiology of schizophrenia. Epidemiol Cal Rev 7: 105–126.
Edelson, E. (1988). Nutrition and the Brain. New York: Chelsea House.
Edgerton, R. B., Cohen, A. (1994). Culture and schizophrenia: the DOSMD challenge. Br J
Psychiatry 164: 222–231.
Essali, M. A., Das, R., de Belleroche, J., Hirsch, S. R. (1990). The platelets polyphosphoinosi-
tide system in schizophrenia: the effects of neuroleptic treatment. Biol Psychiatry 28: 478–
487.
Feinberg, I. (1990). Cortical pruning and the development of schizophrenia. Schizophr Bull 16:
567–568.
Fenton, W. S., Hibbeln, J., Knable, M. (2000). Essential fatty acids, lipid membrane abnormalities,
and the diagnosis and treatment of schizophrenia. Biol Psychiatry 47: 8–21.
Forster, M. J., Dubey, A., Dawson, K. M., et al. (1996). Age-related losses of cognitive and motor
skills in mice are associated with oxidative protein damage in the brain. Proc Natl Acad Sci,
USA 93: 4765–4769.
Freud, S. (1961). Civilization and its discontents. In The Standard Edition of the Com-
plete Psychological Work of Sigmund Freud, Vol. 21, ed. J. Strachey. New York: Norton,
pp. 59–145.
Fukuzako, H. Fukuzako, T., Takeuchi, K., et al. (1996). Phosphorus magnetic resonance spec-
troscopy in schizophrenia: correlation between membrane PL metabolism in the temporal
lobe and positive symptoms. Progr Neuropsychopharmacol Biol Psychiatry 20: 629–640.
Glantz, L. A., Lewis, D. A. (2000). Decreased dendritic spine density on prefrontal cortical pyra-
midal neurons in schizophrenia. Arch Gen Psychiatry 57: 65–73.
Glen, A. I. M., Glen, E. M. T., Horrobin, D. F., et al. (1994). A red cell membrane abnormality in
a subgroup of schizophrenic patients: evidence for two diseases. Schizophr Res 12: 53–61.
Heckers, S. (1997). Neuropathology of schizophrenia: cortex, thalamus, basal ganglia, and
neurotransmitter-specific projection system. Schizophr Bull 23: 403–421.
Hegarty, J. D., Baldessarini, R. J., Tohen, M., Waternaux, C., Oepen, G. (1994). One hundred years
of schizophrenia: a meta-analysis of the outcome literature. Am J Psychiatry 151: 1409–1416.
Hoek, H. W., Brown, A. S., Susser, E. (1998). The Dutch famine and schizophrenia spectrum
disorders. Soc Psychiatry Psychiatr Epidemiol 33: 373–379.
Horrobin, D. F. (1996). Schizophrenia as a membrane lipid disorder which is expressed through-
out the body. Prostaglandins, Leukot Essent Fatty Acids 55: 3–8.
(1998). The membrane phospholipid hypothesis as a biochemical basis for the neurodevelop-
mental concept of schizophrenia. Schizophr Res 30: 193–208
169 Nutritional factors and schizophrenia
(1999). The effects of antipsychotic drugs on membrane phospholipids: a possible novel mech-
anism of action of clozapine. In Phospholipid Spectrum Disorders in Psychiatric, ed. M. Peet,
A. L. Glen, D. Horrobin. Carnforth, UK: Marius Press, pp. 113–117.
Horrobin, D. F., Glen, A. I. M., Vaddadi, K. S. (1994). The membrane hypothesis of schizophrenia.
Schizophr Res 13: 195–208.
Horrocks, L. A., Ansell, G. B., Porcellati, G. (ed.) (1982). Phospholipids in the Nervous System,
Vol. 1, Metabolism. New York: Raven Press.
Hudson, C. J., Young, L. T., Li, P. P., Warsh, J. J. (1993). CNS signal transduction in the patho-
physiology and pharmacology of affective disorders and schizophrenia. Synapse 13: 278–
293.
Hulshoff Pol, H. E., Hoek, H. W., Susser, E., et al. (2000). Prenatal exposure to famine and brain
morphology in schizophrenia. Am J Psychiatry 157: 1170–1172.
Innis, S. M. (1991). Essential fatty acids in growth and development. Progr Lipid Res 30: 39–
103.
Jablensky, A., Sartorius, N., Ernberg, G., et al. (1991). Schizophrenia: manifestations, incidence
and course in different cultures. Psychol Med Monogr Suppl 20: 1–97.
Jeding, I., Evans, P. J., Akanmu, D., et al. (1995). Characterization of the potential antioxidant
and pro-oxidant actions of some neuroleptic drugs. Biochem Pharmacol 49: 359–365.
Kaiya, H., Nishida, A., Imai, A., Nakashima, S., Nozawa, Y. (1989). Accumulation of diacylglyc-
erol in platelet phosphoinositides turnover in schizophrenia: a biological marker of good
prognosis? Biol Psychiatry 26: 669–676.
Karno, M., Jenkins, J. H. (1993). Cross-cultural issues in the course and treatment of schizophre-
nia. Psych Clin North Am 16: 339–350.
Keshavan, M. S., Murray, R. M. (1997). Neurodevelopment and Adult Psychopathology. Cambridge,
UK: Cambridge University Press.
Keshavan, M. S., Mallinger, A. G., Pettegrew, J. W., Dippold, C. (1993). Erythrocyte membrane
phospholipids in psychotic patients. Psychiatr Res 49: 9–95.
Keshavan, M. S., Anderson, S., Pettegrew, J. W. (1994). Is schizophrenia due to excessive synaptic
pruning in the prefrontal cortex? The Feinberg hypothesis revisited. J Psychiatr Res 28:
239–265.
Keshavan, M. S., Sujata, A., Mehra, M., Montrose, D. M., Sweeney, J. A. (2003a). Psychosis prone-
ness and ADHD in young relatives of schizophrenia patients. Schizophr Res 59: 85–92.
Keshavan, M. S., Stanley, J. A., Montrose, D. M., Minshew, N. J., Pettegrew, J. W. (2003b). Pre-
frontal membrane phospholipid metabolism of child and adolescent offspring at risk for
schizophrenia or schizoaffective disorder: an in vivo 31 P MRS study. Mol Psychiatry 8: 316–
323.
Khan, A., Khan, S. R., Leventhal, R. M., Brown, W. A. (2001). Symptom reduction and suicide
risk among patients treated with placebo in antipsychotic clinical trials: an analysis of the
Food and Drug Administration database. Am J Psychiatry 158: 1449–1454.
Khan, M. M., Evans, D. R., Gunna, V., et al. (2002). Reduced erythrocyte membrane essential
fatty acids increased lipid peroxides in schizophrenia at the never-medicated first-episode
of psychosis and after years of treatment with antipsychotics. Schizophr Res 58: 1–10.
170 S. P. Mahadik
Kulhara, P., Chakrabarti, S. (2001). Culture and schizophrenia and other psychotic disorders.
Psychiatr Clin North Am 24: 449–464.
Lawrie, S. M., Abukmeil, S. S. (1998). Brain abnormality in schizophrenia: a systematic and
quantitative review of volumetric magnetic resonance imaging studies. Br J Psychiatry 172:
119–120.
Mahadik, S. P., Evans, D. (1997). Essential fatty acids in the treatment of schizophrenia. Drugs
Today 33: 5–17.
Mahadik, S. P., Gowda, S. (1996). Antioxidants in the treatment of schizophrenia. Drugs Today
32: 1–13.
Mahadik, S. P., Mukherjee, S. (1996). Free radical pathology and the antioxidant defense in
schizophrenia. Schizophr Res 19: 1–18.
Mahadik, S. P., Scheffer, R. E. (1996). Oxidative injury and potential use of antioxidants in
schizophrenia. Prostaglandins, Leukot Essent Fatty Acids 55: 45–54.
Mahadik, S. P., Mukherjee, S., Correnti, E., et al. (1994). Distribution of plasma membrane
phospholipids and cholesterol in skin fibroblasts from drug-naive patients at the onset of
psychosis. Schizophr Res 13: 239–247.
Mahadik, S. P., Mukherjee, S., Horrobin, D., et al. (1996a). Plasma membrane phospholipid fatty
acid composition of cultured skin fibroblasts from schizophrenic patients: comparison with
bipolar and normal controls. Psychiatr Res 63: 133–142.
Mahadik, S. P., Shendarkar, N. S., Scheffer, R., Mukherjee, S., Correnti, E. E. (1996b). Utilization
of precursor essential fatty acids in culture by skin fibroblasts from schizophrenic patients
and normal controls. Prostaglandins, Leukot Essent Fatty Acids 55: 65–70.
Mahadik, S. P., Mulchandani, M., Hegde, M. V., Ranjekar, P. K. (1999a). Cultural and socioeco-
nomic differences in dietary intake of essential fatty acids and antioxidants-effects on the
outcome. In Phospholipid Spectrum Disorders in Psychiatric, ed. D., Horrobin, A. L. Glen,
M. Peet. Carnforth, UK: Marius Press, pp. 167–179.
Mahadik, S. P., Sitasawad, V., Mulchandani, M. (1999b). Membrane peroxidation and the neu-
ropathology of schizophrenia. In Phospholipid Spectrum Disorders in Psychiatric, ed. D.
Horrobin, A. L. Glen, M. Peet. Carnforth, UK: Marius Press, pp. 99–111.
Mahadik, S. P., Evans, D., Lal, H. (2001). Oxidative stress and the role of antioxidant and ω-
3 essential fatty acid supplementation in schizophrenia. Prog Neuropsychopharmacol Biol
Psychiatry 25: 463–493.
Margolis, R. L., Chung, D.-M., Post, R. M. (1994). Programmed cell death: Implications for
neuropsychiatric disorders. Biol Psychiatry 35: 946–956.
McCreadie, R. G. (1997). The Nithsdale Schizophrenia Surveys 16: breast-feeding and schizophre-
nia: preliminary results and hypothesis. Br J Psychiatry 170: 334–337.
McGrath, J. (1999). Hypothesis: is low vitamin D a risk-modifying factor for schizophrenia?
Schizophr Res 40: 173–177.
McIntyre, R. S., McCann, S. M., Kennedy, S. H. (2001). Antipsychotic metabolic effects: weight
gain, diabetes mellitus and lipid abnormalities. Can J Psychiatr 46: 272–281.
Mednick, S. A., Machon, R. A., Huttunen, M. O., Bonett, D. (1988). Adult schizophrenia following
prenatal exposure to an influenza epidemic. Arch Gen Psychiatry 45: 189–192.
Murphy, H. B. M. (1984). Editorial: diseases of civilization? Psychol Med 14: 487–490.
171 Nutritional factors and schizophrenia
Yao, J. K. (1999). Red blood cell and platelet fatty acid metabolism in schizophrenia. In Phospho-
lipid Spectrum in Psychiatric Disorders, ed. D. Horrobin, A. L. Glen, M. Peet. Carnforth, UK:
Marius Press, pp. 57–71.
Yao, J. K., Yasaei, P., van Kammen, D. P. (1992). Increased turnover of platelet phosphatidylinositol
in schizophrenia. Prostaglandins, Leukotr Essent Fatty Acids 46: 39–46.
Yao, J. K., van Kammen, D. P., Welker, J. A. (1994). Red cell membrane dynamics in schizophrenia:
II fatty acid composition. Schizophr Res 13: 217–226.
10
Schizophrenia, neurodevelopment,
and epigenetics
Arturas Petronis
Centre for Addiction and Mental Health, Toronto, Canada
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
174
175 Schizophrenia,neurodevelopment, and epigenetics
the ectoderm during embryogenesis, which serves as indirect support for aberrant
neurodevelopment.
What could be the cause(s) of the above brain development and schizophrenia-
related aberrations? A large group of putative etiological factors has been sug-
gested, investigated, and categorized into environmental and genetic groups. The
first group includes various obstetric complications such birth traumas, maternal
viral infection during pregnancy, pre-eclampsia, and deficiencies in nutrition. The
second group puts the emphasis on DNA sequence variation in the genes that may
play a role in neurodevelopment. So far, none of the above factors has been proven
to be in a cause–effect relationship with schizophrenia. In this chapter, the idea that
developmental changes in schizophrenia can be caused and/or mediated by epigen-
etic factors is suggested. It is argued that shifting the emphasis from the traditional
“gene–environment” dichotomy to epigenetics may provide a cohesive theoretical
framework for the myriad of fragmented phenomenological and molecular find-
ings in schizophrenia and lead to a series of new molecular strategies, designs, and
approaches.
A A AA A
AAAAAA
AAAA
A A AA A
A A AA A
A A AA A
Fig. 10.1. DNA and histone modifications in regulation of gene expression. DNA is wrapped around
histone complexes (gray cylinders) to form nucleosomes. Depending on DNA and histone
modifications (small circles and squares) chromatin can be transcriptionally competent
(A) or not (B). PolyA sequences represent mRNA molecules.
and Ginder, 1999; Stancheva and Meehan, 2000; Yeivin and Razin, 1993) although
this is not universal (Walsh and Bestor, 1999).
One of the mechanisms of epigenetic regulation of genes is related to methy-
lation of the binding sites for transcription factors, serving to change the affinity
of these factors for the regulatory sequences of these specific genes (Riggs et al.,
1998). In addition to the “critical site” effects of met C, the overall ratio of met Cs
over unmethylated cytosines in a gene-regulatory region also contributes to gene
activity. This type of regulation seems to be linked to another mechanism of epi-
genetic regulation, namely various types of histone modification. Histones are
key components of nucleosomes represented by four highly conservative proteins:
H2A, H2B, H3, and H4. The role of mediators between DNA modification and
chromatin modification is performed by a group of met C-binding proteins such
as MECP2, MBD1, and MBD2, among others (reviewed in Li, 2002). The met C-
binding proteins recruit different chromatin remodeling proteins and transcription
complexes to the loci of methylated DNA. For example, MECP2 binds to methy-
lated DNA and attracts histone deacetylases that hypoacetylate histones. Tran-
scriptionally competent chromatin is normally enriched with acetylated histones,
while transcriptionally silent chromatin is deacetylated (reviewed in Robertson
177 Schizophrenia,neurodevelopment, and epigenetics
and Wolffe, 2000). The interaction of DNA methylation and histone acetylation
demonstrates that the two types of epigenetic regulation act in concert. In addition
to acetylation, nucleosomal histones can be modified by methylation, phosphory-
lation, ubiquitylation, and possibly other, yet undiscovered, ways. It is important to
note that histone modification targets are numerous residues of lysine, arginine, and
serine. For example, Lys9, Lys14, Lys18, and Lys23 of H3 and Lys5, Lys8, Lys12, and
Lys16 of H4, together with lysines on H2A and H2B, can be acetylated; whereas Lys4,
Lys9, Lys27, Arg2, Arg17, and Arg26 of H3 and Lys20 and Arg3 of H4 can be methy-
lated (Li, 2002). It is evident that a large number of combinations of various types of
histone modification are possible even over a short stretch of chromatin. Further-
more, the same DNA sequence may be in a wide variety of functional states. This
provided the basis for the concept of the “histone code” (Jenuwein and Allis, 2001).
In addition to their putative role in regulation of autosomal biallelically expressed
genes, epigenetic mechanisms of transcriptional repression are of primary impor-
tance for X chromosome inactivation and genomic imprinting, as well as in the tran-
scriptional inactivation of endogenous retroviruses and other “parasitic” sequences
(reviewed in Wolffe and Matzke, 1999). Furthermore, epigenetic factors play a role
in DNA mutagenesis and repair as well as in DNA recombination and possibly
replication.
Epigenetic patterns are transmitted similarly to DNA sequences, from maternal
chromatids to daughter chromatids during mitotic divisions, and transmission of
the epigenetic status is called the epigenetic inheritance system (Maynard Smith,
1990). Unlike DNA sequences, which exhibit nearly complete interclonal fidelity,
epigenetic status usually exhibits only partial stability; over time, this can result in
substantial changes across identical sequences. DNA and chromatin modifications
do react to the extracellular environment. Stochastic factors in the cell also con-
tribute to epigenetic differences in the cells of the same line. After mitotic division,
the daughter chromosomes do not necessarily carry identical epigenetic patterns in
comparison with the parental chromosomes, and, although belonging to the same
group of cells (myocytes, lymphocytes, and epithelial cells), these cells exhibit small
quantitative differences in terms of their phenotypes and functions. Over time, sub-
stantial epigenetic differences may be accumulated across the cells of the same cell
line or the same tissue (Riggs et al., 1998). This represents a fundamental difference
between epigenetic systems and DNA sequence-based hereditary factors. The par-
tial stability of epigenetic modification is called epigenetic metastability. Epigenetic
metastability also applies to the germline. Although it has been generally accepted
that the parental epigenetic profile is replaced by a new one during the maturation
of gametes, there is increasing evidence demonstrating that some epigenetic signals
escape erasure and can be transmitted from one generation to another (Rakyan
et al., 2002).
178 A. Petronis
The idea of a putative relationship between gene activity and development was
entertained by numerous biologists during the twentieth century. For example, T.
H. Morgan hypothesized that “genes . . . are changing in some way as development
proceeds in response to that part of the protoplasm in which they lie, and that these
changes have reciprocal influence on the protoplasm” (Morgan, 1934). In the 1950s,
C. H. Waddington coined the term “epigenetics,” which referred to the develop-
mental processes that “connect” the genotype to phenotype, or the processes by
which genotype gives a rise to phenotype (reviewed in Slack, 2002). These ideas
originated long before the advent of molecular biology and represented predomi-
nantly a theoretical, although inspiring, framework that influenced various other
fields of scientific activity including psychiatric research (Gottesman and Shields,
1982; Woolf, 1997). Waddington is best known for the concept of “epigenetic land-
scape.” The uneven surface of the landscape with “hills” and “valleys” represents
a simplified version of multidimensional and intricate non-linear interactions of
various molecules within the cell during the embryonic development (Fig. 10.2A).
The rolling ball symbolizes a phenotype of a differentiating cell, which at each
developmental step acquires new features. The end result of this process is a huge
variety of highly specialized cells (Fig. 10.2B).
The idea of the primary role of epigenetic factors in development was raised
again in two seminal papers published in 1975 (Holliday and Pugh, 1975; Riggs,
1975). At that time, the presence of epigenetic DNA modification (methylation)
was already known and there was experimental evidence suggesting that epige-
netic modification may have an impact on the regulation of gene activity. The two
papers represented the first systematic attempt to synthesize the available evidence
regarding linkage of epigenetic regulation to both differentiation of cells and main-
tenance of the differentiated cell states. There are two key questions. First, how do
the pluripotent undifferentiated embryonic cells become highly specialized.
Second, how is the specific phenotype of a specialized cell maintained over numer-
ous mitotic divisions. For example, ectoderm cells differentiate into two very dif-
ferent phenotypes, neurons and keratocytes, and the latter would never convert
into the former, and vice versa. What keeps the cell “locked” in a particular regu-
lated state? Understanding the molecular principles of the developmental programs
remains one of the main challenges for developmental biologists. Although there
are no final answers, epigenetics seems to be one of the best candidates for a molec-
ular mechanism that would be consistent with a wide variety of developmental
events in a cell.
Maintenance of the epigenetic profile is consistent with the phenotypic stability of
somatic cells. In mammals, methylation occurs predominantly at the symmetrical
dinucleotide CpG positions, which after DNA replication become hemimethylated.
This hemimethylated DNA is the favored substrate for DNMT1, which efficiently
181 Schizophrenia,neurodevelopment, and epigenetics
Fig. 10.2. Epigenetics and cell differentiation. (A) Waddington’s epigenetic landscape represents the
imaginary path of an undifferentiated pluripotent embryonic cell during development.
(B) The end result of cell differentiation is a wide variety of highly specialized cells that look
different and perform different functions, although they contain the same DNA sequences.
(See plate section for color version.)
recognizes unmethylated CpGs in the newly synthesized strands that are comple-
mentary to the methylated CpG and proceeds to methylate them (Bestor, 2000).
In tissue culture, fidelity of maintenance methylation in mammalian cells was 97–
99.9% and de novo methylation activity was 3–5% per mitosis (Riggs et al., 1998).
Fidelity of maintenance methylation in different types of cell in vivo is unknown
but it is very possible that it is sufficient to assure clonal continuity of myocytes,
hepatocytes, and the many other kinds of specialized cell. The ways by which
182 A. Petronis
Fig. 10.3. (A) Methylation reprogramming in the germline. Primordial germ cells (PGCs) in the mouse
become demethylated early in development. Remethylation begins in pro-spermatogonia
in male germ cells and after birth in growing oocytes. Some stages of germ cell develop-
ment are shown. (B) Methylation reprogramming in preimplantation embryos. The paternal
genome is demethylated by an active mechanism immediately after fertilization. The mater-
nal genome is demethylated by a passive mechanism that depends on DNA replication. Both
are remethylated around the time of implantation to different extents in embryonic (EM)
and extraembryonic (EX) lineages. Methylated imprinted genes and some repeat sequences
(upper dashed line) do not become demethylated. Unmethylated imprinted genes (lower
dashed line) do not become methylated. (Reproduced with permission from Reik, W., Dean,
W., and Walter, J. (2001). Epigenetic reprogramming in mammalian development. Science,
293: 1089–1093. Copyright 2001 American Association for the Advancement of Science.)
Fig. 10.4. Waddington’s epigenetic landscape: a view from “behind.” The peaks and troughs are formed
by the epigenetic status of various genes (A, B, C, etc.) in the cell, and this determines cellular
phenotype and functions.
adrenal hormone homeostasis during puberty) is the key factor that changes from
a predisposing epimutation to a disease-causing one. Environmental factors may
contribute to the progression of pre-epimutation, but it is more likely that develop-
mental changes and stochastic events play a more important role (Petronis, 2001).
Consistent with the latter, some first episodes of schizophrenia come without any
herald of the disease and in the absence of hazardous environmental factors. Sever-
ity of epigenetic misregulation may fluctuate over time, which in clinical terms
will be treated as disease remissions and relapses. The age-dependent epigenetic
changes may reach epigenetic reversion to a near normal state, which is seen as
a decline of psychotic symptoms and partial recovery from the disease (Petronis,
2001).
There is one key issue that the epigenetic theory treats differently to the neu-
rodevelopmental approach. From the epigenetic point of view, age- and hormone-
dependent neurochemical changes rather than structural changes in the brain are
the main disease mechanism. Brain morphological aberrations are more likely to
be just the reporters of mild deviation in the developmental program rather than
factors causing or predisposing to schizophrenia. There is strong reason to believe
that those early and minor developmental aberrations can be fully compensated
by the developing brain, which exhibits surprising plasticity and compensatory
potential (Kolb, 1995; Woods, 1998). The shift from the brain structural peculiarities
to the functional ones is consistent with the rare but very informative cases of
monozygotic twins discordant for schizophrenia, where brain anomalies are more
severe in the unaffected twin (Torrey et al., 1999, Fig. 6.4 on p. 114). The subtle
localized non-progressive prenatal brain developmental aberrations are just like
superficial scars, which do not do any harm but only remind of an old minor
injury. The problem of latency of the neurodevelopmental changes – one of the
main unclear issues in the neurodevelopmental theory – can now be reformulated
into the latency of epigenetic misregulation of some critical genes.
The heuristic value of the epigenetic model of schizophrenia lies in the possibil-
ity of integrating a variety of unrelated data into a new theoretical framework that
provides the basis for new experimental approaches in the study of this disease.
As it was attempted to show above, epigenetics is likely to be intimately related
to development and, therefore, represents an important aspect in the interpreta-
tion of the neurodevelopmental findings in schizophrenia. In one of her seminal
papers on “dynamic genome,” Barbara McClintock (1951) wrote: “If the ordered
processes of development are deranged, then genes which usually become active at
very specific times may instead be activated spasmodically or in random fashion
during development.” Fifty years later we raise a reverse question: can abnormally
expressed genes derange development? The answer to this question should provide
new insights into the mystery of schizophrenia.
188 A. Petronis
Acknowledgements
I thank Dr. A. Wong and Mr. Z. A. Kaminsky (CAMH, Toronto, Canada) for their
valuable comments and advice. This work was supported by grants from the Ontario
Mental Health Foundation, the National Alliance for Research on Schizophrenia
and Depression, and the Stanley Foundation.
REFERENCES
Bestor, T. H. (2000). The DNA methyltransferases of mammals. Hum Mol Genet 9: 2395–
2402.
Cardno, A. G., Gottesman, I. I. (2000). Twin studies of schizophrenia: from bow-and-arrow
concordances to Star Wars Mx and functional genomics. Am J Med Genet 97: 12–17.
Chen, Y., Sharma, R. P., Costa, R. H., Costa, E., Grayson, D. R. (2002). On the epigenetic regulation
of the human reelin promoter. Nucl Acids Res 30: 2930–2939.
El-Osta, A. (2002). FMR1 silencing and the signals to chromatin: a unified model of transcrip-
tional regulation. Biochem Biophys Res Commun 295: 575–581.
El-Osta, A., Baker, E. K., Wolffe, A. P. (2001). Profiling methyl-CpG specific determinants on
transcriptionally silent chromatin. Mol Biol Rep 28: 209–215.
Gottesman, I., Shields, J. (1982). Schizophrenia: The Epigenetic Puzzle. Cambridge, UK: Cambridge
University Press.
Gottlicher, M., Minucci, S., Zhu, P., et al. (2001). Valproic acid defines a novel class of HDAC
inhibitors inducing differentiation of transformed cells. EMBO J 20: 6969–6978.
Henikoff, S., Matzke, M. A. (1997). Exploring and explaining epigenetic effects. Trends Genet 13:
293–295.
Holliday, R. (1994). Epigenetics: an overview. Dev Genet 15: 453–457.
(1996). DNA methylation in eukaryotes: 20 years on. In Epigenetic Mechanisms of Gene Regu-
lation, ed. V. Russo, R. Martienssen, A. Riggs. Cold Spring Harbor, NY: Cold Spring Harbor
Laboratory Press, pp. 5–27.
Holliday, R., Pugh, J. E. (1975). DNA modification mechanisms and gene activity during devel-
opment. Science 187: 226–232.
Jablonka, E., Lamb, M. (1995). Epigenetic Inheritance and Evolution. Oxford: Oxford University
Press.
Jackson-Grusby, L., Beard, C., Possemato, R., et al. (2001). Loss of genomic methylation causes
p53-dependent apoptosis and epigenetic deregulation. Nat Genet 27: 31–39.
Jenuwein, T., Allis, C. D. (2001). Translating the histone code. Science 293: 1074–1080.
Kolb, B. (1995). Brain Plasticity and Behavior. Mahwah, NJ: Lawrence Erlbaum Associates.
Kraepelin, E. (1919). Dementia Praecox and Paraphrenia. Edinburgh: Livingstone.
Lewis, D. A., Levitt, P. (2002). Schizophrenia as a disorder of neurodevelopment. Annu Rev
Neurosci 25: 409–432.
Li, E. (2002). Chromatin modification and epigenetic reprogramming in mammalian develop-
ment. Nat Rev Genet 3: 662–673.
189 Schizophrenia,neurodevelopment, and epigenetics
Litt, M. D., Simpson, M., Gaszner, M., Allis, C. D., Felsenfeld, G. (2001). Correlation between
histone lysine methylation and developmental changes at the chicken beta-globin locus.
Science 293: 2453–2455.
Maynard Smith, J. (1990). Models of a dual inheritance system. J Theor Biol 143: 41–53.
McClintock, B. (1951). Chromosome organization and genic expression. Genes and Mutations.
Cold Spring Harb Symp Quant Biol XVI: 13–47.
Mertineit, C., Yoder, J. A., Taketo, T., et al. (1998). Sex-specific exons control DNA methyltrans-
ferase in mammalian germ cells. Development 125: 889–897.
Morgan, T. H. (1934). Embryology and Genetics. New York: Columbia University Press.
Petronis, A. (2000). The genes for major psychosis: aberrant sequence or regulation? Neuropsy-
chopharmacology 23: 1–12.
(2001). Human morbid genetics revisited: relevance of epigenetics. Trends Genet 17: 142–146.
Petronis, A., Paterson, A. D., Kennedy, J. L. (1999). Schizophrenia: an epigenetic puzzle? Schizophr
Bull 25: 639–655.
Petronis, A., Popendikyte, V., Kan, P. X., Sasaki, T. (2002). Major psychosis and chromosome 22:
genetics meets epigenetics. CNS Spectrums 7: 209–214.
Petronis, A., Gottesman, I. I., Kan, P. X., et al. (2003). Monozygotic twins exhibit numerous
epigenetic differences: clues to twin discordance? Schizophr Bull 29: 169–178.
Phiel, C. J., Zhang, F., Huang, E. Y., et al. (2001). Histone deacetylase is a direct target of valproic
acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem 276: 36734–
36741.
Rakyan, V. K., Blewitt, M. E., Druker, R., Preis, J. I., Whitelaw, E. (2002). Metastable epialleles in
mammals. Trends Genet 18: 348–351.
Razin, A., Shemer, R. (1999). Epigenetic control of gene expression. Results Probl Cell Differ 25:
189–204.
Reik, W., Dean, W., Walter, J. (2001). Epigenetic reprogramming in mammalian development.
Science 293: 1089–1093.
Rideout, III, W. M., Eggan, K., Jaenisch, R. (2001). Nuclear cloning and epigenetic reprogramming
of the genome. Science 293: 1093–1098.
Riggs, A., Xiong, Z., Wang, L., LeBon, J. M. (1998). Methylation dynamics, epigenetic fidelity
and X chromosome structure. In Epigenetics, Vol. Novartis Foundation Symposium 214:
Epigenetics, ed. A. Wolffe. Chichester, UK: Wiley, pp. 214–227.
Riggs, A. D. (1975). X inactivation, differentiation, and DNA methylation. Cytogenet Cell Genet
14: 9–25.
Robertson, K. D., Wolffe, A. P. (2000). DNA methylation in health and disease. Nat Rev Genet 1:
11–19.
Schumacher, A. (2001). Mechanisms and brain specific consequences of genomic imprinting in
Prader–Willi and Angelman syndromes. Gene Funct Dis 1: 7–25.
Seeman, M. V. (1997). Psychopathology in women and men: focus on female hormones. Am J
Psychiatry 154: 1641–1647.
Siegfried, Z., Eden, S., Mendelsohn, M., et al. (1999). DNA methylation represses transcription
in vivo. Nat Genet 22: 203–206.
Singal, R., Ginder, G. D. (1999). DNA methylation. Blood 93: 4059–4070.
190 A. Petronis
Slack, J. M. (2002). Conrad Hal Waddington: the last Renaissance biologist? Nat Rev Genet 3:
889–895.
Stancheva, I., Meehan, R. R. (2000). Transient depletion of xDnmt1 leads to premature gene
activation in Xenopus embryos. Genes Dev 14: 313–327.
Torrey, E. F., Bowler, A. E., Taylor, E. H., Gottesman, I. I. (1999). Schizophrenia and Manic
Depressive Disorder. The Biological Roots of Mental Illness as Revealed by the Landmark Study
of Identical Twins. New York: Basic Books.
Tremolizzo, L., Carboni, G., Ruzicka, W. B., et al. (2002). An epigenetic mouse model for molecular
and behavioral neuropathologies related to schizophrenia vulnerability. Proc Natl Acad Sci
USA 99: 17095–17100.
van den Veyver, I. B., Zoghbi, H. Y. (2001). Mutations in the gene encoding methyl-CpG-binding
protein 2 cause Rett syndrome. Brain Dev 23(Suppl. 1): S147–S151.
Walsh, C. P., Bestor, T. H. (1999). Cytosine methylation and mammalian development. Genes
Dev 13: 26–34.
Warnecke, P. M., Clark, S. J. (1999). DNA methylation profile of the mouse skeletal alpha-actin
promoter during development and differentiation. Mol Cell Biol 19: 164–172.
Wolffe, A. P. (1994). Inheritance of chromatin states. Dev Genet 15: 463–470.
Wolffe, A. P., Barton, M. C. (2000). Developmental regulation of chromatin function and gene
expression. In Chromatin Structure and Regulation, ed. S. C. R. Elgin, J. L. Workman. Oxford:
Oxford University Press, pp. 182–202.
Wolffe, A. P., Matzke, M. A. (1999). Epigenetics: regulation through repression. Science 286:
481–486.
Woods, B. T. (1998). Is schizophrenia a progressive neurodevelopmental disorder? Toward a
unitary pathogenetic mechanism. Am J Psychiatry 155: 1661–1670.
Woolf, C. M. (1997). Does the genotype for schizophrenia often remain unexpressed because of
canalization and stochastic events during development? Psychol Med 27: 659–668.
Yeivin, A., Razin, A. (1993). Gene methylation patterns and expression. In DNA Methylation:
Molecular Biology and Biological Significance, ed. J. Jost, H. Saluz. Basel: Birkhauser Verlag,
pp. 523–568.
11
The existence of early environmental risk factors for schizophrenia is central to the
notion of schizophrenia as a neurodevelopmental disorder (Marenco and Wein-
berger, 2000; McDonald et al., 2000) and these risk factors represent some of the
most challenging and interesting targets of schizophrenia epidemiology. This chap-
ter discusses prenatal and perinatal risk factors for schizophrenia. Childhood devel-
opmental impairment and later environmental and psychosocial risk factors are
dealt with in Chs. 22 and 13, respectively.
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
191
192 M. Cannon, K. Dean, and P. B. Jones
number of possible risk factors, including prenatal and childhood infections, the
prevalence of which tends to increase with increasing population density and in
winter.
Prenatal influenza
The early evidence regarding the role of prenatal infections has come from ecolog-
ical, or population-association studies. In 1988, Mednick and colleagues demon-
strated that the offspring of women who were in the second trimester of pregnancy
during the 1957–8 influenza A2 pandemic in Helsinki were about twice as likely
to be hospitalized with a diagnosis of schizophrenia as those not exposed dur-
ing pregnancy or exposed earlier or later in pregnancy. There have been many
attempts to replicate this finding for the 1957 epidemic using an ecological design
(Erlenmeyer-Kimling et al., 1994; Izumoto et al., 1999; Kendell and Adams, 1991;
Kendell and Kemp, 1989; Kunugi et al., 1995; McGrath et al., 1994; Mednick et al.,
1990; O’Callaghan et al., 1991; Torrey et al., 1992). The effect sizes demon-
strated in these studies are small: somewhere between 1.5 and 2.0 (Cannon and
Jones, 1996), and not all ecological studies replicated the association (Kendell and
Kemp, 1989; Selten and Slaets, 1994; Selten et al., 1999; Susser et al., 1994; Torrey
et al., 1988; Westergaard et al., 1999). However, the balance of evidence does suggest
that prenatal exposure to the 1957 epidemic was associated with a raised incidence
of schizophrenia, particularly if the exposure occurred in the second trimester of
pregnancy (reviewed by McGrath et al., 1995a; Wright et al., 1999). Studies of
longer-term trends in the association between the timing of influenza epidemics
and the birth dates of people with schizophrenia have yielded generally positive
results in most (Adams et al., 1993; Barr et al., 1990; Sham et al., 1992; Takei
et al., 1994) but not all (Morgan et al., 1997; Selten and Slaets, 1994; Torrey et al.,
1988) cases. In most studies where the sexes are examined separately, the positive
association was found mainly or exclusively in females (Adams et al., 1993; Izumoto
et al., 1999; Kendell and Kemp, 1989; McGrath et al., 1994; Mednick et al., 1990;
O’Callaghan et al., 1991; Takei et al., 1994), but no satisfactory explanation has yet
been offered for this.
The ecologic design has undoubted limitations, including the so-called “ecolog-
ical fallacy” (we cannot be certain that the individuals in the population who were
exposed to influenza in utero are the same individuals who are diagnosed with
schizophrenia as adults) and the possibility of unknown confounding with other
factors, such as maternal fever or medication.
However, the application of cohort and case–control methodology has not
shed much light on the problem. Two case–control studies replicated the associa-
tion between second-trimester exposure to influenza and later schizophrenia but
relied on maternal recall for information about the exposure (Stöber et al., 1992;
193 Early environmental risk factors for schizophrenia
Wright et al., 1995). Two cohort studies have failed to support the influenza–
schizophrenia association (Cannon et al., 1996; Crow and Done, 1992) but had low
power to examine this effect (Adams and Kendell, 1996).
in the first half of pregnancy and an increased (threefold) risk of adult schizophrenia
(Brown et al., 2003).
Prenatal malnutrition
A series of ecological studies of exposure to prenatal famine (Susser and Lin, 1992,
1994; Susser et al., 1996) have demonstrated dose–response relationships between
maternal nutritional deprivation during the Nazi blockade of the Netherlands in
the winter of 1944–5 and risk of later schizophrenia in the offspring. The initial
finding to emerge from these studies was that birth cohorts exposed to the famine in
early but not late gestation had a twofold increase in risk for schizophrenia (Susser
and Lin, 1992; Susser et al., 1996). A subsequent study using military conscription
data demonstrated that prenatal famine during early gestation was associated with
a twofold elevation in risk for schizoid or schizotypal personality disorders (Hoek
et al., 1996). The authors postulated that severe nutritional deprivation is the etio-
logical mechanism involved (Butler et al., 1999; Hoek et al., 1999). This possibility
has received indirect support from a finding that a short interval between siblings
is associated with an increased risk of schizophrenia (Westergaard et al., 1999).
There is biological plausibility for this putative cause; congenital CNS defects in
this population were related to famine exposure in a similar fashion (Susser et al.,
195 Early environmental risk factors for schizophrenia
1985). Interaction with maternal genetic effects or exposure to maternal stress may
also be components of any causal association (Koenig et al., 2002).
Rhesus incompatibility
Rhesus (Rh) incompatibility, characterized by an Rh-negative mother preg-
nant with an Rh-positive fetus, has been associated with an elevated risk for
schizophrenia. Hollister et al. (1996) used data on males from the Danish Perinatal
Cohort Study and found an elevated risk for schizophrenia among offspring of
Rh-incompatible pregnancies compared with Rh-compatible pregnancies. Rhesus
incompatibility can give rise to hemolytic disease of the newborn, which results,
among other things, in childhood neuromotor abnormalities and behavioral dis-
orders such as emotional instability. It is possible that schizophrenia is yet another
possible consequence of rhesus hemolytic disease (Hollister and Brown, 1999).
Rhesus hemolytic disease occurs most commonly in mothers who have already
delivered a Rh-positive child, thus triggering the production of the antibody against
the Rh(D) antigen. As hypothesized, the risk for schizophrenia among males in the
Danish Perinatal Cohort Study was increased over threefold in second and later-
born offspring from Rh-incompatible pregnancies, but there was no increased risk
for first-born offspring (Hollister et al., 1996).
helix, ear protrusion, abnormal eye fissure inclination, abnormal binocular diam-
eter, wide skull base, and hypoplastic ear lobe size. A novel method for studying
craniofacial dymorphology is the use of three-dimensional laser surface scanning
and geometric morphometric analysis (Hennessy et al., 2002), and its application
to psychosis is in progress. Elevated rates of MPAs have also been reported in
other psychoses (McGrath et al., 2002) and disorders such as schizotypal personal-
ity disorder (Weinstein et al., 1999); MPAs appear to be relatively independent of
obstetric complications in their association with later development of schizophre-
nia (McNeil and Cantor-Graae, 1999, 2000). MPAs are increased in those at genetic
risk for schizophrenia in some (Lawrie et al., 2001; Schiffman et al., 2002) but not
all (Griffiths et al., 1998) studies that have examined this issue. The association
between MPAs and schizophrenia does not appear to be confounded by ethnicity
(Dean et al., 2003).
risk factors for schizophrenia. This effect is possibly mediated by uterine artery
vasoconstriction and reduced placental blood flow (Teixeira et al., 1999). Prenatal
stress may be particularly pathogenic in those already at genetic risk of schizophrenia
(Mednick and Schulsinger, 1968; Mednick et al., 1971).
Done et al. UK Cohort: NCDS No overall association between risk factors for perinatal
(1991) death and later schizophrenia. Low maternal weight
and medications given to baby were associated with
narrowly defined schizophrenia
Buka et al. US Cohort: NCPP Non-significant increased risk of psychosis in those
(1993) (Providence exposed to chronic fetal hypoxia (OR, 2.6)
center)
Sacker et al. UK Cohort: NCDS Re-analysis of Done et al. (1991). The following
(1995) variables were associated with increased risk of
narrowly defined schizophrenia: low maternal
weight; maternal psychological problems; smoking in
pregnancy; poor antenatal attendance; rhesus
negative; parity > 2; previous births < 2500 g;
bleeding in pregnancy; untrained person delivering;
baby’s weight < 2500 g; other drugs given to baby
Jones et al. Finland Cohort: 1966 North Low birth weight (OR, 2.4; CI, 1–5.6): combination of
(1998) Finland Birth low birth weight and prematurity (OR, 3.5; CI,
Cohort 1.3–9.6) and perinatal brain damage (OR, 6.9; CI,
2.9–16.3) were associated with schizophrenia
Hultman et al. Sweden Case–control Schizophrenia was associated with multiparity (OR,
(1999) 2.0); bleeding during pregnancy (OR, 3.5); winter
birth (OR, 1.4); SGA (males only OR, 3.2); parity > 4
(males only OR, 3.6)
Dalman et al. Sweden Cohort Increased risk of pre-eclampsia (OR, 2.5); vacuum
(1999) extraction (OR, 1.7); malformations (OR, 2.4);
parity of 1 (OR, 1.3); bleeding during pregnancy
(OR, 2.0); threatened premature delivery (OR, 2.3);
gestational age < 32 weeks (OR, 2.7); prolonged
delivery (OR, 1.6); uterine inertia (OR, 2.4; ponderal
index < 20 (OR, 3.4); respiratory illness (OR, 1.5);
birth weight < 2500 g (males only OR, 2.2); birth
weight < 1500 g (females only OR, 6.0); SGA (males
only OR, 1.9). Preeclampsia remained significant
after adjusting for all other complications
Kendell et al. Scotland Case–control No association found between any OC and
(2000) schizophrenia
Kendell et al. Scotland Case–control Emergency Cesarian section (OR, 3.7; CI, 1.02–13.1)
(2000) and labor > 12 hours were associated with
subsequent schizophrenia
199 Early environmental risk factors for schizophrenia
NCPP, National Collaborative Perinatal Project: NCDS, National Child and Development Study; OR, odds
ratio; CI, confidence interval; SGA, small for gestational age; OC, obstetric complication.
one exposure based on a prior hypothesis, and these have showed significant effects
for the putative risk increasing mechanism of hypoxic–ischemic damage (Buka
et al., 1993; Cannon et al., 2000; Rosso et al., 2000; Zornberg et al., 2000b).
The standardized fashion of reporting results and the methodological similarities
of the population-based studies listed in Table 11.1 lend themselves to a meta-
analytic approach. Meta-analysis provides a method for integrating quantitative
data from multiple studies by using a weighted average of the results in which
larger studies have more influence than smaller studies. It improves the estimates
of effect size, increases the statistical power, and helps to make sense out of studies
with conflicting conclusions (Egger et al., 1997; Fleiss, 1993).
Cannon et al. (2002a) carried out a meta-analytic synthesis of the prospective
“population-based” studies that had published data on individual obstetric com-
plications and found that three groups of complications were significantly associ-
ated with schizophrenia: (i) complications of pregnancy (bleeding, diabetes, rhe-
sus incompatibility, pre-eclampsia); (ii) complications of delivery (uterine atony,
asphyxia, emergency cesarean section); and (iii) abnormal fetal growth and develop-
ment (low birth weight, congenital malformations, reduced head circumference).
200 M. Cannon, K. Dean, and P. B. Jones
Pooled estimates of effect sizes were generally small (odds ratios 2). Interactive
effects and independence of obstetric risk factors could not be examined using this
design. An individual patient-data meta-analysis on these population-based stud-
ies, such as that carried out by Geddes et al. (1999) on case–control studies, may
help to elucidate such issues.
The period of risk during which hypoxic brain damage may lead to later
schizophrenia appears to extend beyond birth. In the North Finland 1966 cohort, a
group of CNS insults that had hypoxia as a common mechanism was identified and
termed perinatal brain damage (Rantakallio et al., 1987). Of 125 survivors of such
perinatal brain damage, six (4.8%) developed schizophrenia in adult life: a sevenfold
relative risk (Jones et al., 1998). The estimate of the proportion of schizophrenia
in the general population that may be attributable to this mechanism was 5–8% in
this study.
Conclusions
It seems that many pre- and perinatal risk factors are somehow involved in increas-
ing the risk for schizophrenia in later life. The best evidence to date is for prenatal
(probably second-trimester) exposure to influenza and other respiratory infections,
prenatal rubella, hypoxia-related obstetric complications and low birth weight or
intrauterine growth retardation. Evidence is less secure for prenatal stress or prena-
tal malnutrition, principally because of the difficulties in obtaining suitable samples
in which to examine these exposures.
201 Early environmental risk factors for schizophrenia
Table 11.2. Estimate of approximate effect sizes for pre- and perinatal risk
factors for schizophrenia
Infection
Prenatal influenza 2.0
Prenatal respiratory infection (2◦ ) 2.1
Prenatal rubella 5.2
Prenatal poliovirus (2◦ ) 1.05
Neonatal and childhood CNS infection 4.0
Malnutrition
Prenatal famine (1◦ ) 2.0
Prenatal stress
Bereavement of spouse 6.2
Flood (2◦ ) 1.8
War (2◦ )
“Unwantedness” 2.4
Maternal depression (3◦ ) 1.8
Obstetric complications
General 2.0
Rhesus incompatibility 2.8
Hypoxia-related 2.1–4.4
Perinatal brain damage 7.0
Low birth weight (< 2500 g) 1.6
Pre-eclampsia 2.5
RR, relative risk; OR, odds ratio; 1◦ , 2◦ , 3◦ , first, second, and third trimester,
respectively.
The effect sizes for these prenatal and perinatal risk factors are small, with odds
ratios or relative risks of approximately 2 (Table 11.2). Similarly, genetic studies
since the early 1990s have suggest that multiple genes with small effect sizes are
involved in causation of schizophrenia. These and other findings indicate that we
are likely to be dealing with interactive effects of prenatal and genetic factors (Jones
and Murray, 1991; Tsuang, 2000). Recent findings confirm earlier reports that
increasing paternal age is associated with an increased risk of schizophrenia (Hare
202 M. Cannon, K. Dean, and P. B. Jones
and Moran, 1979; Malaspina et al., 2001). The most parsimonious interpretation
attributes the effect to mutations in the male sperm line prior to conception. These
mutations, in turn, are to a large extent environmentally determined, by both the
cumulative life experience (age) and specific environmental exposures (e.g. toxins)
of the biological father.
Techniques for investigating genetic and environmental risk factors are beginning
to converge, with case–control and cohort designs being used for genetic associa-
tion studies. Molecular genetic studies are including measures of environment and
cohort studies are collecting both DNA samples and information on early and later
environmental risk factors (Caspi et al., 2002, 2003). Over the next few years, we
should see the first reports of studies examining precisely measured genetic and
early environmental causes of schizophrenia in the same populations.
Acknowledgements
This work was made possible by the support of the Wellcome Trust (MC), NARSAD
(MC) and the Theodore and Vada Stanley Foundation (PBJ).
REFERENCES
Adams, W., Kendell, R. E. (1996). Influenza and schizophrenia (letter). Br J Psychiatry 169:
791–792.
Adams, W., Kendell, R. E., Hare, E. H. (1993). Epidemiological evidence that maternal influenza
contributes to the aetiology of schizophrenia: an analysis of Scottish, English and Danish
data. Br J Psychiatry 163: 522–534.
Agid, O., Shapira, B., Zislin, J., et al. (1999). Environment and vulnerability to major psychiatric
illness: a case control study of early parental loss in major depression, bipolar disorder and
schizophrenia. Mol Psychiatry 4: 163–172.
Barr, C. E., Mednick, S. A., Munk-Jorgensen, P. (1990). Exposure to influenza epidemics during
gestation and adult schizophrenia: a 40 year study. Arch Gen Psychiatry 47: 869–874.
Bowlby, J. (1944). Forty four juvenile thieves: their characters and home life. Int J Psychoanalysis
25: 19–52, 107–127.
Brown, A. S., Cohen, P., Greenwald, S., Susser, E. (2000a). Nonaffective psychosis after prenatal
exposure to rubella. Am J Psychiatry 157: 438–443.
Brown, A. S., Schaefer, C. A., Wyatt, R. J., et al. (2000b). Maternal exposure to respiratory
infections and adult schizophrenia spectrum disorders: a prospective birth cohort study.
Schizophr Bull 26: 287–296.
Brown, A. S., Begg, M., Gravenstein, S., et al. (2003). Serologic evidence for prenatal influenza in
the etiology of schizophrenia. Schizophr Res 60: 34.
203 Early environmental risk factors for schizophrenia
Buka, S. L., Tsuang, M. T., Lipsitt, L. P. (1993). Pregnancy/delivery complications and psychiatric
diagnosis. A prospective study. Arch Gen Psychiatry 50: 151–156.
Buka, S. L., Tsuang, M. T., Torrey, E. F., et al. (2001). Maternal infections and subsequent psychosis
among offspring. Arch Gen Psychiatry 58: 1032–1037.
Buka, S. L., Tsuang, M. T., Goldstein, J. M., et al. (2003). Maternal exposure to herpes simplex
virus type 2 and psychosis among adult offspring: replication and reanalysis. Schizophr Res
60: 35.
Butler, P. D., Printz, D., Klugewicz, D., Brown, A. S., Susser, E. S. (1999). Plausibility of early
nutritional deficiency as a risk factor for schizophrenia. In Prenatal Exposures in Schizophre-
nia, ed. E. S. Susser, A. S. Brown, J. M. Gorman. Washington, DC: American Psychiatric
Press, pp. 163–193.
Byrne, M., Browne, R., Mulryan, N., et al. (2000). Labour and delivery complications and
schizophrenia. Case–control study using contemporaneous labour ward records. Br J Psy-
chiatry 176: 531–536.
Cannon, M., Jones, P. (1996). Schizophrenia: neuroepidemiology of schizophrenia. J Neurol
Neurosurg Psychiatry 61: 604–613.
Cannon, M., Cotter, D., Sham, P. C., et al. (1996). Schizophrenia in an Irish sample following
prenatal exposure to the 1957 influenza epidemic: a case-controlled, prospective follow-up
study. Schizophr Res 11: 95.
Cannon, M., Jones, P. B., Murray R. M. (2002a). Obstetric complications and schizophrenia.
Historical and meta-analytic review. Am J Psychiatry 159: 1080–1092.
Cannon, M., Caspi, A., Moffott, T. E., et al. (2002b). Evidence for early-childhood, pan-
developmental impairment specific to schizophreniform disorder. Results from a longi-
tudinal birth cohort. Arch Gen Psychiatry 59: 449–456.
Cannon, T. D. (1997). On the nature and mechanisms of obstetric influences in schizophrenia:
a review and synthesis. Int Rev Psychiatry 9: 387–397.
Cannon, T. D., Rosso, I. M., Hollister, J. M., et al. (2000). A prospective cohort study of
genetic and perinatal influences in the etiology of schizophrenia. Schizophr Bull 26: 351–
366.
Caspi, A., McClay, J., Moffitt, T. E., et al. (2002). Role of genotype in the cycle of violence in
maltreated children. Science 297: 851–854.
Caspi A., Sugden, K., Moffitt, T. E., et al. (2003). Influence of life stress on depression: moderation
by a polymorphism in the 5-HTT gene. Science 301: 386–389.
Cotter, D., Larkin, C., Waddington, J. L., O’Callaghan, E. (1996). Season of birth in schizophrenia:
clue or cul-de-sac. In The Neurodevelopmental Basis of Schizophrenia, ed. J. L. Waddington,
P. B. Buckley. Austin, TX: Landes, pp. 17–33.
Crow, T. J., Done, J. (1992). Prenatal exposure to influenza does not cause schizophrenia. Br J
Psychiatry 161: 390–393.
Dalman, C., Allebeck, P., Culberg, J., Grunewald, C., Köster, M. (1999). Obstetric complications
and the risk of schizophrenia: a longitudinal study of a national birth cohort. Arch Gen
Psychiatry 56: 234–240.
Dalman, C., Thomas, H. V., David, A. S., et al. (2001). Signs of asphyxia at birth increase the risk
of schizophrenia. A population-based case–control study. Br J Psychiatry 179: 403–408.
204 M. Cannon, K. Dean, and P. B. Jones
Dean, K., Fearon, P., Dazzan, P., et al. (2003). Increased minor physical anomalies in all ethnic
groups in the AESOP first-onset study. Schizophr Res 60.
Done, J., Johnstone, E. C., Frith, C. D., et al. (1991). Complications of pregnancy and delivery in
relation to psychosis in adult life: data from the British Perinatal Mortality Survey. Br Med
J 302: 1576–1580.
Egger, M., Davey Smith, G., Phillips, A. N. (1997). Meta-analysis: principles and procedures.
Br Med J 315: 1533–1537.
Erlenmeyer-Kimling, L., Folnegovic, Z., Hrabic-Zerjavic. V., Borcic, B., Folnegovic-Smalc, V.
(1994). Schizophrenia and prenatal exposure to the 1957 influenza epidemic in Croatis. Am
J Psychiatry 151: 1496–1498.
Fine, P. E. M., Adelstein, A. M., Snowman, J., Clarkson, J. A., Evans, S. M. (1985). Long term
effects of exposure to viral infections in utero. Br Med J 290: 509–511.
Fleiss, J. L. (1993). The statistical basis of meta-analysis. Stat Meth Med Res 2: 121–145.
Geddes, J. R., Lawrie, S. M. (1995). Obstetric events in schizophrenia: a meta-analysis. Br J
Psychiatry 167: 786–793.
Geddes, J. R., Verdoux, H., Takei, N., et al. (1999). Schizophrenia and complications of
pregnancy and labor: an individual-patient data meta-analysis. Schizophr Bull 25: 413–
423.
Griffiths, T. D., Sigmundsson, T., Takei, N., et al. (1998). Minor physical anomalies in familial
and sporadic schizophrenia: the Maudsley family study. J Neurol Neurosurg Psychiatry 64:
56–60.
Gualtieri, C. T., Adams, A., Shen, C. D., Loiselle, D. (1982). Minor physical anomalies in alcoholic
and schizophrenic adults and hyperactive and autistic children. Am J Psychiatry 139: 640–
643.
Hare, E. H., Moran, P. A. (1979). Raised parental age in psychiatric patients: evidence for the
constitutional hypothesis, Br J Psychiatry 134: 169–177.
Hennessy, R. J., Kinsella, A., Waddington, J. L. (2002). 3D laser surface scanning and geometric
morphometric analysis of craniofacial shape as an index of cerebro-craniofacial morpho-
genesis: initial application to sexual dimorphism. Biol Psychiatry 51: 507–514.
Hoek, H. W., Susser, E., Buck, K. A., et al. (1996). Schizoid personality disorder after prenatal
exposure to famine. Am J Psychiatry 153: 1637–1639.
Hoek, H. W., Brown, A. S., Susser, E. S. (1999). The Dutch famine studies: prenatal nutritional
deficiency and schizophrenia. In Prenatal Exposures in Schizophrenia, ed. E. S. Susser, A. S.
Brown, J. M. Gorman. Washington, DC: American Psychiatric Press, pp. 135–161.
Hollister, J. M., Brown, A. S. (1999). Rhesus incompatibility and schizophrenia. In Prenatal
Exposures in Schizophrenia, ed. E. S. Susser, A. S. Brown, J. M. Gorman. Washington, DC:
American Psychiatric Press, pp. 197–214.
Hollister, J. M., Laing, P., Mednick, S. A. (1996). Rhesus incompatibility as a risk factor for
schizophrenia in male adults. Arch Gen Psychiatry 53: 19–24.
Hultman, C. M., Sparen, P., Takei, N., Murray, R. M., Cnattingius, S. (1999). Prenatal and
perinatal risk factors for schizophrenia, affective psychosis and reactive psychosis of early
onset: case–control study. Br Med J 318: 421–426.
205 Early environmental risk factors for schizophrenia
Huttunen, M., Niskanen, P. (1978). Prenatal loss of father and psychiatric disorders. Arch Gen
Psychiatry 35: 427–431.
Imamura, Y., Nakane, Y., Ohta, Y., Kondo, H. (1999). Lifetime prevalence of schizophrenia among
individuals exposed to atomic bomb radiation in Nagasaki. Acta Psychiatr Scand 100: 344–
349.
Ismail, B., Cantor-Graae, E., McNeil, T. F. (1998). Minor physical anomalies in schizophrenic
patients and their siblings. Am J Psychiatry 155: 1695–1702.
(2000). Minor physical anomalies in schizophrenia: cognitive, neurological and other clinical
correlates. J Psychiatr Res 34: 45–56.
Izumoto, Y., Inoue, S., Yasuda, N. (1999). Schizophrenia and the influenza epidemics of 1957 in
Japan. Biol Psychiatry, 46: 119–124.
Jones, P., Murray, R. (1991). The genetics of schizophrenia is the genetics of neurodevelopment,
Br J Psychiatry 158: 615–623.
Jones, P., Rodgers, B., Murray, R., Marmot, M. (1994). Childhood developmental risk factors for
schizophrenia in the British 1946 birth cohort. Lancet 344: 1398–1402.
Jones, P. B., Rantakallio, P., Hartikainen, A. L., et al. (1998). Schizophrenia as a long-term outcome
of pregnancy, delivery and perinatal complications: a 28-year follow-up of the 1966 North
Finland general population birth cohort. Am J Psychiatry 155: 355–364.
Jones, P. B., Pang, D., Piriach, S., Fine, P. E. M. (1999). Prenatal viral infection and subsequent
mental illness: a long-term cohort study of 6152 subjects. Schizophr Res 36: 45.
Kendell, R. E., Adams, W. (1991). Unexplained fluctuations in the risk for schizophrenia by month
and year of birth. Br J Psychiatry 158: 758–763.
Kendell, R. E., Kemp, J. W. (1989). Maternal influenza in the aetiology of schizophrenia. Arch
Gen Psychiatry 46: 878–882.
Kendell, R. E., McInneny, K., Jusczak, E., Bain, M. (2000). Obstetric complications and schizophre-
nia: two case–control studies based on structured obstetric records. Br J Psychiatry 174:
516–522.
Kendler, K. S., Neale, M. C., Kessler, R. C., Heath, A. C., Eaves, L. J. (1992). Childhood parental
loss and adult psychopathology in women. A twin study perspective. Arch Gen Psychiatry
49: 109–116.
Kinney, D. K. (2001). Prenatal stress and risk for schizophrenia. Int J Mental Health 29: 62–71.
Kinney, D. K., Hyman, W., Greetham, C., Tramer, S. (1999). Increased relative risk for schizophre-
nia and prenatal exposure to a severe tornado. Schizophr Res 36: 45–46.
Koenig, J. I., Kirkpatrick, B., Lee, P. (2002). Glucocorticoid hormones and early brain development
in schizophrenia. Neuropsychopharmacology 27: 309–318.
Kunugi, H., Nanko, S., Takei, N., et al. (1995). Schizophrenia following in utero exposure to the
1957 influenza epidemics in Japan. Am J Psychiatry 152: 450–452.
Lane, A., Kinsella, A., Murphy, P., et al. (1997). The anthropometric assessment of dysmorphic
features in schizophrenia as an index of its developmental origins. Psychol Med 27: 1155–
1164.
Lawrie, S. M., Byrne, M. Miller, P. et al. (2001). Neurodevelopmental indices and the development
of psychotic symptoms in subjects at high risk of schizophrenia. Br J Psychiatry 178: 524–530.
206 M. Cannon, K. Dean, and P. B. Jones
Lewis, G., David, A., Andreasson, S., Allebeck, P. (1992). Schizophrenia and city life. Lancet 340:
137–140.
Lou, H., Hansen, D., Nordentoft, M., et al. (1994). Prenatal stressors of human life affect fetal
brain development. Dev Med Child Neurol 36: 826–832.
Malaspina, D., Harlap, S., Fennig, S., et al. (2001). Advancing paternal age and the risk of
schizophrenia. Arch Gen Psychiatry 58: 313–412.
Marcelis, M., Takei, N., van Os, J. (1999). Urbanization and risk for schizophrenia: does the effect
operate before or around the time of illness onset? Psychol Med 29: 1197–1203.
Marcelis, M., Navarro-Mateu, F., Murray, R., Selten, J. P., van Os, J. (1998). Urbanization and
psychosis: a study of 1942–1978 birth cohorts in the Netherlands. Psychol Med 28: 871–
879.
Marenco, S., Weinberger, D. R. (2000). The neurodevelopmental hypothesis of schizophrenia:
following a trail of evidence from cradle to grave. Dev Psychopathol 12: 501–527.
McDonald, C., Fearon, P., Murray, R. M. (2000). Neurodevelopmental hypothesis of schizophre-
nia 12 years on: data and doubts. In Childhood Onset of Adult Psychopathology, ed. J. L.
Rapaport. Washington, DC: American Psychiatric Press, pp. 193–222.
McGrath, J. J., Welham, J. L. (1999). Season of birth and schizophrenia: a systematic review and
meta-analysis of data from the Southern Hemisphere. Schizophr Res 35: 237–242.
McGrath, J. J., Pemberton, M. R., Welham, J. L., Murray, R. M. (1994). Schizophrenia and the
influenza epidemics of 1954, 1957 and 1959: a southern hemisphere study. Schizophr Res
14: 1–8.
McGrath, J. J., Castle, D., Murray, R. M. (1995a). How can we judge whether or not prenatal expo-
sure to influenza causes schizophrenia. In Neural Development and Schizophrenia. Theory
and Research, ed. S. A. Mednick, J. M., Hollister. New York: Plenum Press, pp. 203–214.
McGrath, J. J., van Os, J., Hoyos, C., et al. (1995b). Minor physical anomalies in psychoses:
associations with clinical and putative aetiological variables. Schizophr Res 18: 9–20.
McGrath, J., El-Saadi, O., Grim, V., et al. (2002). Minor physical anomalies and quantitative
measures of the head and face in patients with psychosis. Arch Gen Psychiatry 59: 458–464.
McNeil, T. F. (1988). Obstetric factors and perinatal injuries. In Handbook of Schizophrenia,
Vol. 3: Nosology, Epidemiology and Genetics, ed. M. T. Tsuang, J. C. Simpson. North Holland:
Elsevier Science, pp. 319–311.
(1995). Perinatal risk factors and schizophrenia: selective review and methodological concerns.
Epidemiol Rev 17: 107–112.
McNeil, T. F., Cantor-Graae, E. (1999). Does preexisting abnormality cause labor–delivery com-
plications in fetuses who will develop schizophrenia? Schizophr Bull 25: 425–435.
(2000). Minor physical anomalies and obstetric complications in schizophrenia. Aust NZ J
Psychiatry 34(Suppl.): S65–S73.
McNeil, T., Cantor-Graae, E., Ishmail, B. (2000). Obstetric complications and congenital mal-
formations in schizophrenia. Brain Res Rev 31: 166–178.
Mednick, S. A., Schulsinger F. (1968). Some premorbid characteristics related to breakdown in
children with schizophrenic mothers. J Psychiat Res 6(Suppl. 1): 267–291.
Mednick, S. A., Mura, E., Schulsinger, F., Mednick, B. (1971). Perinatal conditions and infant
development in children with schizophrenic parents. Soc Biol 18: S103–S113.
207 Early environmental risk factors for schizophrenia
Mednick, S. A., Machon, R. A., Huttunen, M. O., Bonett, D. (1988). Adult schizophrenia following
prenatal exposure to an influenza epidemic. Arch Gen Psychiatry 45: 189–192.
Mednick, S. A., Machon, R. A., Huttunen, M. O. (1990). An update on the Helsinki influenza
project. Letter to Editor. Arch Gen Psychiatry 47: 292.
Morgan, V., Castle, D., Page, A., et al. (1997). Influenza epidemics and incidence of schizophrenia,
affective disorders and mental retardation in Western Australia: no evidence of a major effect.
Schizophr Res 26: 25–39.
Mortensen, P. B., Pedersen, C. B., Westergaard, T., et al. (1999). Effects of family history and place
and season of birth on the risk of schizophrenia. N Engl J Med 340: 603–608.
Murray, L., Fiori-Crowley, A., Hooper, R., Cooper P. J. (1996). The impact of post natal depression
and associated adversity on early mother–infant interactions and later infant outcome. Child
Dev 67: 2512–2526.
Myhrman, A., Rantakallio, P., Isohanni, M., Jones, P. B., Partanen, U. (1996). Unwantedness of a
pregnancy and schizophrenia in the child. Br J Psychiatry 169: 637–640.
Nemeroff, C. (1999). The pre-eminent role of early untoward experience on vulnerability to major
psychiatric disorders: the nature–nurture controversy revisited and soon to be resolved. Mol
Psychiatry 4: 106–108.
O’Callaghan, E., Sham, P., Takei, N., Glover, G., Murray, R. M. (1991). Schizophrenia after
prenatal exposure to 1957 A2 influenza epidemic. Lancet 337: 1248–1250.
O’Connor, T., Heron, J., Golding, J., Beveridge, M., Glover, V. (2002). Maternal antenatal anx-
iety and children’s behavioural/emotional problems at 4 years. Br J Psychiatry 180: 502–
508.
Rantakallio, P. (1969). Groups at risk in low birth weight infants and perinatal mortality. Acta
Paediatr Scand Suppl 193: 1–71.
Rantakallio, P., von Wendt, L., Koivu, M. (1987). Prognosis of perinatal brain damage: a prospec-
tive study of a one year birth cohort of 12 000 children. Early Hum Dev 15: 75–84.
Rantakallio, P., Jones, P., Moring, J., von Wendt, L. (1997). Association between central nervous
system infections during childhood and adult onset schizophrenia and other psychoses: a
28-year follow-up. Int J Epidemiol 26: 837–843.
Rosso, I. M., Cannon, T. D., Huttunen, T., et al. (2000). Obstetric risk factors for early-onset
schizophrenia in a Finnish birth cohort. Am J Psychiatry 157: 801–807.
Rutter, M. (2002). The interplay of nature, nurture and development. Arch Gen Psychiatry 59:
996–1001.
Sacker, A., Done, D. J., Crow, T. J., Golding, J. (1995). Antecedents of schizophrenia and affective
illness: obstetric complications. Br J Psychiatry 166: 734–741.
Schiffman, J., Abrahamson, A., Cannon, T., et al. (2001). Early rearing factors in schizophrenia.
Int J Mental Health 30: 3–16.
Schiffman, J., Ekstrom, M., LaBrie, J., et al. (2002). Minor physical anomalies and schizophrenia
spectrum disorders: a prospective investigation. Am J Psychiatry 159: 238–243.
Schore, A. N. (2001). Effects of early relational trauma on right brain development, affect regu-
lation, and infant mental health. Infant Ment Health J 22, 201–269.
Selten, J. P. C. J., Slaets, J. P. J. (1994). Evidence against maternal infection as a risk factor for
schizophrenia. Br J Psychiatry 164: 674–676.
208 M. Cannon, K. Dean, and P. B. Jones
Selten, J. P., van der Graaf, Y., van Duursen, R., Gispen-de Wied, C. C., Kahn, R. S. (1999).
Psychotic illness after prenatal exposure to the 1953 Dutch Flood Disaster. Schizophr Res 35:
243–245.
Sham, P. C., O’Callaghan, E., Takei, N., et al. (1992). Schizophrenia following pre-natal exposure
to influenza epidemics between 1939 and 1960. Br J Psychiatry 160: 461–466.
Siegel, D. J. (2001). Toward an interpersonal neurobiology of the developing mind: attachment
relationships, “mindsight”, and neural integration. Infant Ment Health J 22: 67–94.
Stöber, G., Franzek, E., Beckmann, J. (1992). The role of maternal infectious diseases during
pregnancy in the aetiology of schizophrenia in offspring. Eur Psychiatry 7: 147–152.
Suomi, S. J. (1997). Long-term effects of different early rearing experiences on social, emotional,
and physiological development in nonhuman primates. In M. S. Keshavan and R. M. Murray
(Eds.) Neurodevelopment and Adult Psychopathology, Ch. 8, ed. M. S. Keshavan, R. M. Murray.
Cambridge: Cambridge University Press, pp. 104–116.
Susser, E., Lin, S. P. (1992). Schizophrenia after prenatal exposure to the Dutch Hunger Winter
of 1944–1945. Arch Gen Psychiatry 49: 983–988.
(1994). Schizophrenia after prenatal exposure to the Dutch hunger winter of 1944–1945. Arch
Gen Psychiatry 51: 333–334.
Susser, E. S., Lin, S. P., Brown, A. S., Lumey, L. H., Erlenmeyer-Kimling, L. (1994). No relation
between risk of schizophrenia and prenatal exposure to influenza in Holland. Am J Psychiatry
151: 922–924.
Susser, E. S., Neugebauer, R., Hoek, H. W., et al. (1996). Schizophrenia after prenatal famine. Arch
Gen Psychiatry 53: 25–31.
Susser, E. S., Schaefer, C., Brown, A., Begg, M., Wyatt, R. J. (2000). The design of the Prenatal
Determinants of Schizophrenia Study (PDS). Schizophr Bull 26: 257–273.
Susser, M., Hauser, W. A., Keity, J. L., Paneth, N., Stein, Z. (1985). Quantitative estimates of
prenatal and perinatal risk factors for perinatal mortality, cerebral palsy, mental retardation
and epilepsy. In Prenatal and Perinatal Factors Associated with Brain Disorder, ed. J. Freeman.
Washington, DC: Institute of Health, pp. 359–432.
Suvisaari, J., Haukka, J., Tanskanen, A., Hovi, T., Lönnqvist, J. (1999). Association between
prenatal exposure to poliovirus infection and adult schizophrenia. Am J Psychiatry 156:
1100–1102.
Takei, N., Sham, P., O’Callaghan, E., et al. (1994). Prenatal exposure to influenza and the devel-
opment of schizophrenia: is the effect confined to females? Am J Psychiatry 151: 117–119.
Takei, N., Sham, P., O’Callaghan, E., Glover, G., Murray, R. M. (1995). Schizophrenia: increased
risk associated with winter and city birth: a case–control study in 12 regions within England
and Wales. J Epidemiol Community Health 49: 106–107.
Taylor, A., Fisk, N., Glover, V. (2000). Mode of delivery and later stress response. Lancet 355:
120.
Teixeira, J. M. A., Fisk, N. M., Glover, V. (1999). Association between maternal anxiety in preg-
nancy and increased uterine artery resistance index: cohort based study. Br Med J 318:
153–157.
Tienari, P. (1991). Interaction between genetic vulnerability and family environment: the Finnish
adoptive family study of schizophrenia. Acta Psychiatr Scand 84: 460–465.
209 Early environmental risk factors for schizophrenia
Torrey, E. F., Bowler, A. (1990). Geographical distribution of insanity in America: evidence for
an urban factor. Schizophr Bull 16: 591–604.
Torrey, E. F., Rawlings, R., Waldman, I. N. (1988). Schizophrenic births and viral diseases in two
states. Schizophr Res 1: 73–77.
Torrey, E. F., Bowler, A. E., Rawlings, R. (1992). Schizophrenia and the 1957 influenza epidemic.
Schizophr Res 6: 100–107.
Torrey, E. F., Miller, J., Rawlings. R., Yolken, R. H. (1997). Seasonality of birth in schizophrenia
and bipolar disorder: a review. Schizophr Res 28: 1–38.
Tsuang, M. (2000). Schizophrenia: genes and environment. Biological Psychiatry 47: 210–220.
van Os, J., Selten, J.-P. (1998). Prenatal exposure to maternal stress and later schizophrenia: the
May 1940 invasion of the Netherlands. Br J Psychiatry 172: 324–326.
Wahlberg, K.-E., Wynne, L. C., Oja, H., et al. (1997). Gene–environment interaction in vulnera-
bility to schizophrenia: findings from the Finnish Adoptive Family Study of Schizophrenia.
Am J Psychiatry 154: 355–362.
Weinstein, D. D., Diforio, D., Schiffman, J., Wlaker, E., Bonsall, R. (1999). Minor physical anoma-
lies, dermatoglyphic asymmetries and cortisol levels in adolescents with schizotypal per-
sonality disorder. Am J Psychiatry 156: 617–623.
Westergaard, T., Mortensen, P. B., Pedersen, C. B., Wohlfahrt, J., Melbye, M. (1999). Exposure to
prenatal and childhood infections and the risk of schizophrenia. Arch Gen Psychiatry 56:
993–998.
Wright, P., Rifkin, L., Takei, N., Murray, R. (1995). Maternal influenza, obstetric complications
and schizophrenia. Am J Psychiatry 152: 1714–1720.
Wright, P., Takei, N., Murray, R. M., Sham, P. C. (1999). Seasonality, prenatal influenza exposure
and schizophrenia. In Prenatal Exposures in Schizophrenia, ed. E. S. Susser, A. S. Brown,
J. M. Gorman. Washington, DC: American Psychiatric Press, pp. 89–112.
Zornberg, G., Buka, S. L., Tsuang, M. T. (2000a). The problem of obstetrical complications and
schizophrenia. Schizophr Bull 26: 249–256.
(2000b). Hypoxic-ischaemia-related fetal/neonatal complications and risk of schizophrenia
and other nonaffective psychoses: a 19 year longitudinal study. Am J Psychiatry 157: 196–
202.
12
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
210
211 Transcriptomes in schizophrenia
et al., 1996; Schena et al., 1995). Microarray technology is based on the well-
established principle of complementary hybridization between nucleic acids; each
microarray probe is capable of recognizing the complementary sequence through
base pairing (Southern et al., 1999). The “target” (e.g. mRNA extracted from a tissue
sample) is hybridized to the immobilized DNA sequence probes on the array sur-
face, an approach made possible by the miniaturization process and development of
high-throughput printing robots. After a series of high-stringency washes, only the
highly complementary probe–target complexes remain tightly bound. The amount
of the retained label is then quantified over each probe spot, with the amount of
retained label or the signal intensity providing a direct reflection of the abundance
of that mRNA species in the sample (Cheung et al., 1999).
Thus, microarrays permit an extensive and relatively rapid screening of the
expression levels of thousands of mRNA transcripts. Indeed, it is now possible to
analyze the expression profile in cells from a schizophrenia patient using microar-
rays that represent a majority of the human genome. Although this approach
is not informative regarding the potential contribution of translational or post-
translational processing events to the disease state, proteins in the postmortem
brain are likely to be less intact than mRNA, and their large-scale analysis is much
more complex. Furthermore, microarrays analyze transcripts that are intrinsic to
the harvested brain region, whereas protein profiles reflect intrinsic and extrinsic
sources, making it more difficult to discern the anatomical source of the observed
disease-related changes.
Two types of microarray platform have been used in studies of schizophrenia.
Complementary DNA (cDNA) microarrays are composed of relatively long cDNA
probes (typically ranging from approximately 100 to 2000 base pairs) that are
usually DNA clones amplified by the polymerase chain reaction (PCR) or expressed
sequence tags (ESTs) derived from the 3 end of RNA targets. Thousands of these
probes are each deposited or synthesized in very small, unique locations on a solid
support (e.g. glass or membrane structure) (Duggan et al., 1999; Schena and Davis,
1999). The sample analysis can be performed using a single label or a dual fluores-
cent method. In a dual-color analysis, two different fluorescent tags (e.g. cyanin 3
and 5 [Cy3 and Cy5, respectively]) are used to label, through a reverse transcription
procedure (Schena et al., 1996; DeRisi et al., 1997), the mRNA species present in
each of the two tissue samples to be compared (e.g. a subject with schizophrenia
and a matched normal control). The samples containing the labeled targets are
combined and hybridized together onto the same microarray. Using two independ-
ent scans that are selective for each of two incorporated fluorescent tags’ emission
wavelengths, a high-resolution fluorescent intensity measurement is obtained from
each spot on the microarray. This signal is quantified, standardized, and compared
between the two images, and the intensity differences are interpreted as relative
212 D. A. Lewis, K. Mirnics, and P. Levitt
for the selection of appropriate comparison subjects, including both normal con-
trols and subjects with other disorders who share some features (e.g. medication
history) with the schizophrenia subjects under investigation. In addition, the
detailed clinical information suggests the types of parallel study that may need to be
conducted in animal model systems, where factors such as medication exposure can
be assessed in a controlled fashion. For example, we have used macaque monkeys
treated chronically with antipsychotic drugs in a manner that directly mimics their
use in humans as one way of assessing the potential contribution of these thera-
peutic agents to our microarray findings in subjects with schizophrenia (Mirnics
et al., 2000).
Second, in order to avoid the introduction of potential confounds secondary
to geographic factors, differences in the handling of tissue specimens, or other
variables, all tissue specimens ideally are obtained from the same source. A stan-
dardized approach to blocking the brain facilitates examination and sampling for
neuropathological studies and permits a uniform dissection of the region(s) of
interest at the time that a microarray study is initiated. Photographic documen-
tation of the tissue blocks provides a means to confirm and record that the same
macroscopic landmarks are present in the tissue blocks from all subjects. These
procedures also facilitate the design of cutting and sampling procedures to best
approximate the acquisition of systematic and uniform random samples of the
region of interest. In addition, the preparation of slide-mounted sections from the
tissue blocks that are used for the isolation of RNA provides Nissl-stained material
that can be used to verify that the tissue block contains the characteristic cytoarchi-
tecture of the brain region targeted for study. Other sections are used to confirm
array findings for individual transcripts by in situ hybridization.
Third, it is important to assess variables that may affect the expression or integrity
of mRNA. These variables include agonal state events, the cause and manner of
death, and the postmortem interval (PMI), the period between the time of death
and the freezing of the tissue specimens. Although most mRNA species exhibit
minimal to modest degradation following PMIs of more than 24 hours (comparable
to freshly frozen animal brain tissue), premortem events can have a substantial
negative impact. In particular, antemortem hypoxia and/or acidosis, conditions
frequently found in individuals who die in hospital following medical complications
or sustained periods of illness, appear to be associated with the loss of at least some
mRNA species. Available studies suggest that postmortem brain pH provides an
easily measured and reliable assessment of the severity of these factors (Harrison
et al., 1995). Because the factors monitored by brain pH or PMI may not have
linear effects on mRNA integrity, we prefer to match individual pairs of subjects as
closely as possible on these variables, in addition to including pre- and postmortem
conditions as covariates in statistical analyses.
214 D. A. Lewis, K. Mirnics, and P. Levitt
genes within the PSYN group appeared to be subject specific. That is, although
all subjects with schizophrenia showed decreased expression of multiple PSYN
genes, the specific combination of affected transcripts varied from subject to sub-
ject. This phenotypic diversity may reflect a continuum of molecular phenotypes
in schizophrenia, perhaps similar to the variability in the clinical manifestations
of the illness across subjects. In addition to this general molecular diversity seen
across subjects, consistent expression deficits in several genes, such as those for
N-ethylmaleimide-sensitive factor (NSF), synapsin 2, and vesicular ATPase, were
found in almost all subjects with schizophrenia. NSF and its attachment proteins
are critical to provide energy for synaptic vesicle fusion; synapsin II regulates vesicle
availability for exocytosis; and vesicular ATPase is responsible for synaptic vesicle
homeostasis. These expression deficits were confirmed by in situ hybridization,
indicating that they did not represent artifacts of the microarray data analysis or
interpretation procedures.
Associated with these alterations in PSYN genes, robust decreases were also found
in two groups comprising much smaller numbers of genes represented on the arrays.
These two gene groups encode proteins that are specifically related to neurotrans-
mission for gamma-aminobutyric acid (GABA) and glutamate, which together
account for over 95% of cortical synapses. In the glutamate transmission gene
group, glutamate receptor 2 (AMPA2) showed the largest expression decrease, and
the 67 kDa isoform of glutamic acid decarboxylase (GAD67 ) was the most robustly
and consistently affected gene related to GABA neurotransmission (Mirnics et al.,
2000). These single gene findings replicated previous reports using more conven-
tional methods (Akbarian et al., 1995; Eastwood et al., 1995; Volk et al., 2000). The
etiology of these changes remains unknown, yet it is possible that these group-wise
changes reflect changes secondary to the broader alterations in presynaptic function.
For example, a reduction in synaptic activity may lead to the findings of reduced
prefrontal brain-derived neurotrophic factor (BDNF) signaling in schizophrenia
(Hashimoto et al., 2002; Weickert et al., 2003). This, in turn, may account for the
marked and selective reduction in GAD67 mRNA expression observed in the sub-
population of prefrontal GABA neurons (Hashimoto et al., 2003) that express TrkB,
the receptor for BDNF.
Because of the high metabolic demands placed on neurons by the processes
involved in synaptic communication, and given the in vivo evidence for alterations
in prefrontal cortical metabolism in schizophrenia (Berman et al., 1986; Buchs-
baum et al., 1992), we used the well-accepted listing of metabolic groups from the
Kyoto Encyclopedia of Genes and Genomes (KEGG) to identify 70 different clusters
of genes whose protein products are involved in metabolic functions (Middleton et
al., 2002). Remarkably, only five of these metabolic gene groups displayed signifi-
cant alteration in expression in the prefrontal cortex of subjects with schizophrenia.
217 Transcriptomes in schizophrenia
These included the mitochondrial malate shuttle system, transcarboxylic acid cycle,
ornithine/polyamine, aspartate/alanine, and ubiquitin metabolism groups. Veri-
fication of these microarray data by in situ hybridization confirmed reduced
mRNA levels for soluble malate dehydrogenase (MAD1), mitochondrial glutamate–
oxaloacetate transaminase type 2, ornithine decarboxylase antizyme inhibitor,
and ornithine aminotransferase. Interestingly, assessment of expression levels of
individual genes in these pathways for changes in haloperidol-exposed monkeys
revealed an unexpected increase in the expression of MAD1, raising the intriguing
possibility that the antipsychotic treatment may counteract the MAD1 expres-
sion decrease observed in schizophrenia. The MAD1 expression increase was most
prominent in the deep cortical layers, including layer 5, which contains the highest
density of dopamine D2 receptors (Lidow et al., 1989). Together, these findings
suggest that altered prefrontal metabolism may be an important component of the
disease process in schizophrenia, contributing to or reflecting alterations in presyn-
aptic function in the same brain region, and that the reversal of at least some of
these abnormalities may be involved in the therapeutic effects of antipsychotic
medications.
The same microarray data set also revealed multiple changes in genes that play
a role in chemical neurotransmission from the postsynaptic side. In addition to
changes in a subset of glutamate and GABA receptors, we found a marked and
highly consistent reduction in the transcript for a regulator of G-protein signal-
ing protein (RGS4) (Mirnics et al., 2001a). RGS proteins are GTPase-activating
proteins that facilitate hydrolysis of GTP bound to Gα -protein and thus limit the
duration and timing of signaling through G-protein-coupled receptors (De Vries
et al., 2000). Hence, downregulation of RGS4 may compensate for decreased synap-
tic efficacy in the prefrontal cortex by increasing the duration of signaling following
activation of G-protein-coupled receptors. However, a similar magnitude of RGS4
transcript reduction was also present in motor and visual cortices of the same sub-
jects with schizophrenia, regions that differ substantially from the prefrontal cortex
in structure, connectivity, and function. In addition, these changes in RGS4 expres-
sion were not observed in the prefrontal cortex of subjects with major depression
or in monkeys exposed chronically to haloperidol. Consequently, these findings
suggest that decreased RGS4 expression in schizophrenia is not restricted to the
prefrontal cortex, and that this decrease may be specific to the disease process and
not a consequence of potentially confounding factors such as comorbid depression
or antipsychotic medications. Together, these observations support the hypothe-
sis that RGS4 represents a schizophrenia susceptibility gene. Interestingly, RGS4 is
present at chromosomal locus 1q21–22, which has been linked to schizophrenia in
some cohorts (Brzustowicz et al., 2000). Consistent with this hypothesis, evidence
of association between allelic variations in the 5 region of RGS4 and schizophrenia
218 D. A. Lewis, K. Mirnics, and P. Levitt
was found in three family samples, although the pattern of transmission differed
across them (Chowdari et al., 2002). Although replication studies are required to
confirm this finding, these studies do exemplify how expression information from
microarray studies, in concert with positional and functional data, can be used to
identify schizophrenia susceptibility genes.
The concomitant reduction in expression of presynaptic, energy metabolism,
and postsynaptic genes in the same subjects strongly suggests that these findings
are interrelated components of the disease process of schizophrenia, although the
direction of causality among them is unclear at present. It is possible that a primary
deficit in the PSYN group, leading to decreased or inefficient synaptic activity, could
lead to secondary downregulation of energy metabolism and altered signaling at
the postsynaptic membrane. Indeed, we have proposed a testable model (Mirnics
et al., 2001b) which argues that a primary reduction in synaptic efficacy could
account for both the developmental trajectory of schizophrenia and the findings
of decreased markers of cortical synaptic number (Glantz and Lewis, 1997, 2000).
Specifically, we suggested that impaired synaptic function in certain cortical cir-
cuits would initially be compensated, at least to a certain degree, by the normal
exuberant production of synapses, but that these connections would be rendered
more vulnerable to additional alterations as a result of later developmental events.
For example, the adolescence-related pruning of cortical excitatory synapses
(Huttenlocher, 1979; Rakic et al., 1986) would not only reduce the total com-
plement of cortical synapses but might also do so to a greater degree than normal
because of the diminished function of the affected circuits (Mirnics et al., 2001b).
In this regard, the evidence of cognitive abnormalities during childhood of indi-
viduals who later become ill with schizophrenia may be considered to reflect both
the disease process and a risk factor for later developmental disturbances required
for the clinical manifestations of the illness (Lewis and Levitt, 2002). Once pruning
is completed, normally during the second decade of life (although the temporal
trajectory could itself be altered in schizophrenia), the manifestations of the ill-
ness would be expected to be relatively stable; however, the effects of normal aging
might be more severe, perhaps accounting for the non-Alzheimer’s dementia seen
in elderly individuals with schizophrenia (Arnold et al., 1998). Consistent with this
hypothesis, altering neuronal excitability during synaptogenesis has a profound
effect on the numbers of synapses that form, but this effect is not seen if excitability
is altered after synaptogenesis (Burrone et al., 2002).
As in all studies of schizophrenia, replication of findings in additional subjects
and by other investigators is essential. Other investigators, using different cohorts
of subjects, observed some of our microarray findings. For example, Sklar and co-
workers (Sklar, 2001) reported that NSF was decreased in the prefrontal cortex of
schizophrenia subjects; an analysis of single cells from subjects with schizophrenia
219 Transcriptomes in schizophrenia
showed decreased expression levels of several synaptic markers (Hemby et al., 2002);
and altered expression of genes involved in ubiquitinization was reported by Vawter
and colleagues (2001, 2002). Such replication studies are challenging for microar-
ray techniques for a variety of reasons including a lack of standardized methods
and modes of analysis and differences in sensitivity of different platforms. Indeed,
the rapid advances in microarray technology suggest that in the short-term we
are more likely to see improvement in, rather than replication of, results (Branca,
2003). One of the biggest challenges to replication of findings from gene investi-
gators at present is the fact that different sets of gene are represented on different
microarrays. For example, using a microarray containing 1127 brain-related genes
to probe prefrontal area 9 from subjects with schizophrenia, Vawter et al. (2002)
found reduced expression of several of the PSYN (e.g. NSF, vesicular ATPase, synap-
togyrin), GABA (e.g. GAD67 ), and glutamate (e.g. AMPA2) genes identified in our
initial study. However, perhaps because the specific genes constituting these groups
in this study were different and smaller in number than those used in our study,
significant effects at the level of gene groups were not observed.
The impact that the genes represented on the arrays can have on the outcome of
a study may also be reflected in a study using a different platform, the Affymetrix
GeneChip. Investigators from the Mt Sinai group reported deficient expression of
six genes whose protein products are thought to play important roles in myelina-
tion (Hakak et al., 2001). Consistent with these observations, we found reduced
expression of proteolipid protein 1 (PLP1) across the majority of subjects with
schizophrenia (Pongrac et al., 2002). This PLP1 expression deficit did not appear
to be related to antipsychotic medication since it was not found in haloperidol-
treated monkeys. PLP1 appears to have a dual function, participating in both glial
cell development and the assembly of myelin (Schneider et al., 1992). These deficits
in myelin-related genes also raise the interesting question of the possible factors that
may account for such an alteration in the transcriptome. Although such deficits are
frequently discussed as evidence of alterations in transcription, they may be caused
by more fundamental problems. For example, the Mt Sinai group has also observed
a decreased number of glial cells in layer 3 and the underlying white matter in area
9 of subjects with schizophrenia (Hof et al., 2003), suggesting that the reduction
in myelin-related transcripts may be the consequence of a process that leads to
decreased numbers of oligodendrocytes.
Another major study, using a custom-made candidate gene array comprising 300
genes, found upregulated expression of members of the apo L lipoprotein family
(apo L1, apo L2 and apo L4, which play a central role in cholesterol transport) in
several independent cohorts of subjects with schizophrenia (Mimmack et al., 2002).
Although the function of apo L proteins in the brain is unknown, the findings
are of particular interest given that the genes for apo L proteins are located on
220 D. A. Lewis, K. Mirnics, and P. Levitt
Acknowledgements
The interactions between our laboratories that are reflected in this chapter were
supported by NIMH Conte Center Grant MH45156 and by MH43784.
REFERENCES
Akbarian, S., Kim, J. J., Potkin, S. G., et al. (1995). Gene expression for glutamic acid decarboxylase
is reduced without loss of neurons in prefrontal cortex of schizophrenics. Arch Gen Psychiatry
52: 258–266.
Arnold, S. E., Trojanowski, J. Q., Gur, R. E., et al. (1998). Absence of neurodegeneration and
neural injury in the cerebral cortex in a sample of elderly patients with schizophrenia. Arch
Gen Psychiatry 55: 225–232.
Berman, K. F., Zec, R. F., Weinberger, D. R. (1986). Physiological dysfunction of dorsolateral
prefrontal cortex in schizophrenia. II. Role of neuroleptic treatment, attention and mental
effort. Arch Gen Psychiatry 43: 126–135.
Branca, M. (2003). Genetics and medicine: putting gene arrays to the test. Science 300: 238.
221 Transcriptomes in schizophrenia
Brown, P. O., Bottenstein, D. (1999). Exploring the new world of the genome with DNA microar-
rays. Nat Genet 21S: 33–37.
Brzustowicz, L. M., Hodgkinson, K. A., Chow, E. W. C., Honer, W. G., Bassett, A. S. (2000).
Location of a major susceptibility locus for familial schizophrenia on chromosome 1q21–
q22. Science 288: 678–682.
Buchsbaum, M. S., Haier, R. J., Potkin, S. G., et al. (1992). Frontrostriatal disorder of cerebral
metabolism in never-medicated schizophrenics. Arch Gen Psychiatry 49: 935–942.
Burrone, J., O’Byren, M., Murthy, V. N. (2002). Multiple forms of synaptic plasticity triggered
by selective suppression of activity in individual neurons. Nature 420: 414–418.
Cheung, V. G., Morley, M., Aguilar, F., et al. (1999). Making and reading microarrays. Nat Genet
21S: 20–32.
Chowdari, K. V., Mirnics, K., Semwal, P., et al. (2002). Association and linkage analyses of RGS4
polymorphisms in schizophrenia. Hum Mol Genet 11: 1373–1380.
DeRisi, J., Iyer, V. R., Brown, P. O. (1997). Exploring the metabolic and genetic control of gene
expression on a genomic scale. Science 278: 680–686.
DeRisi, J., Penland, L., Brown, P. O., et al. (1996). Use of a cDNA microarray to analyse gene
expression patterns in human cancer. Nat Genet 14: 457–460.
De Vries, L., Zheng, B., Fischer, T., Elenko, E., Farquhar, M. G. (2000). The regulator of G protein
signaling family. Annu Rev Pharmacol Toxicol 40: 235–271.
Duggan, D. J., Bittner, M., Chen, Y., Meltzer, P., Trent, J. M. (1999). Expression profiling using
cDNA microarrays. Nat Genet 21S: 10–14.
Eastwood, S. L., McDonald, B., Burnet, P. W. J., et al. (1995). Decreased expression of mRNAs
encoding non-NMDA glutamate receptors GluR1 and GluR2 in medial temporal lobe neu-
rons in schizophrenia. Mol Brain Res 29: 211–223.
Glantz, L. A., Lewis, D. A. (1997). Reduction of synaptophysin immunoreactivity in the prefrontal
cortex of subjects with schizophrenia: regional and diagnostic specificity. Arch Gen Psychiatry
54: 943–952.
(2000). Decreased dendritic spine density on prefrontal cortical pyramidal neurons in
schizophrenia. Arch Gen Psychiatry 57: 65–73.
Hakak, Y., Walker, J. R., Li, C., et al. (2001). Genome-wide expression analysis reveals dysregulation
of myelination-related genes in chronic schizophrenia. Proc Natl Acad Sci USA 98: 4746–
4751.
Harrison, P. J., Heath, P. R., Eastwood, S. L., et al. (1995). The relative importance of premortem
acidosis and postmortem interval for human brain gene expression studies: selective mRNA
vulnerability and comparison with their encoded proteins. Neurosci Lett 200: 151–154.
Hashimoto, T., Volk, D. W., Buchheit, S. E., Lewis, D. A. (2002). Expression of BDNF and trkB
mRNAs in prefrontal cortex of subjects with schizophrenia. Soc Neurosci Abstr 28: 703.7.
Hashimoto, T., Volk, D. W., Eggan, S. M., et al. (2003). Gene expression deficits in a subclass
of GABA neurons in the prefrontal cortex of subjects with schizophrenia. J Neurosci 23:
6315–6326.
Hemby, S. E., Ginsberg, S. D., Brunk, B., et al. (2002). Gene expression profile for schizophrenia:
discrete neuron transcription patterns in the entorhinal cortex. Arch Gen Psychiatry 59:
631–640.
222 D. A. Lewis, K. Mirnics, and P. Levitt
Hof, P. R., Haroutunian, V., Friedrich, Jr., V. L., et al. (2003). Loss and altered spatial distribution
of oligodendrocytes in the superior frontal gyrus in schizophrenia. Biol Psychiatry 53: 1075–
1085.
Huttenlocher, P. R. (1979). Synaptic density in human frontal cortex: developmental changes and
effects of aging. Brain Res 163: 195–205.
Kamme, F., Salunga, R., Yu, J., et al. (2003). Single-cell microarray analysis in hippocampus CA1:
demonstration and validation of cellular heterogeneity. J Neurosci 23: 3607–3615.
Lewis, D. A. (2002). The human brain revisited: opportunities and challenges in postmortem
studies of psychiatric disorders. Neuropsychopharmacology 26: 143–154.
Lewis, D. A., Levitt, P. (2002). Schizophrenia as a disorder of neurodevelopment. Annu Rev
Neurosci 25: 409–432.
Lewis, D. A., Lieberman, J. A. (2000). Catching up on schizophrenia: natural history and neuro-
biology. Neuron 28: 325–334.
Lidow, M. S., Goldman-Rakic, P. S., Rakic, P., Innis, R. B. (1989). Dopamine D(2) receptors in the
cerebral cortex: distribution and pharmacological characterization with [(3)H]raclopride.
Proc Natl Acad Sci USA 86: 6412–6416.
Lipshutz, R. J., Fodor, S. P. A., Gingeras, T. R., Lockhart, D. J. (1999). High density synthetic
oligonucleotide arrays. Nat Genet 21S: 20–24.
Lockhart, D. J., Barlow, C. (2001). DNA arrays and gene expression analysis in the brain. In
Methods in Genomic Neuroscience, ed. H. R. Chin, S. Moldin. New York: CRC Press, pp.
143–170.
Lockhart, D. J., Dong, H., Byrne, M. C., et al. (1996). Expression monitoring by hybridization to
high-density oligonucleotide arrays. Nat Biotechnol 14: 1675–1680.
Luo, L., Salunga, R. C., Guo, H., et al. (1999). Gene expression profiles of laser-captured adja-
cent neuronal subtypes. Nat Med 5: 117–122.
Middleton, F. A., Mirnics, K., Pierri, J. N., Lewis, D. A., Levitt, P. (2002). Gene expression profiling
reveals alterations of specific metabolic pathways in schizophrenia. J Neurosci 22: 2718–2729.
Mimmack, M. L., Ryan, M., Baba, H., et al. (2002). Gene expression analysis in schizophrenia:
reproducible up-regulation of several members of the apolipoprotein L family located in a
high-susceptibility locus for schizophrenia on chromosome 22. Proc Natl Acad Sci USA 99:
4680–4685.
Mirnics, K., Middleton, F. A., Marquez, A., Lewis, D. A., Levitt, P. (2000). Molecular characteriza-
tion of schizophrenia viewed by microarray analysis of gene expression in prefrontal cortex.
Neuron 28: 53–67.
Mirnics, K., Middleton, F. A., Stanwood, G. D., Lewis, D. A., Levitt, P. (2001a). Disease-specific
changes in regulator of G-protein signaling 4 (RGS4) expression in schizophrenia. Mol
Psychiatry 6: 293–301.
Mirnics, K., Middleton, F. A., Lewis, D. A., Levitt, P. (2001b). Analysis of complex brain disorders
with gene expression microarrays: schizophrenia as a disease of the synapse. Trends Neurosci
24: 479–486.
Pongrac, J., Middleton, F. A., Lewis, D. A., Levitt, P., Mirnics, K. (2002). Gene expression profiling
with DNA microarrays: advancing our understanding of psychiatric disorders. Neurochem
Res 27: 1049–1063.
223 Transcriptomes in schizophrenia
Rakic, P., Bourgeois, J. P., Eckenhoff, M. F., Zecevic, N., Goldman-Rakic, P. S. (1986). Concurrent
overproduction of synapses in diverse regions of the primate cerebral cortex. Science 232:
232–235.
Schena, M., Davis, R. W. (1999). Microgenes, genomes and chips. In DNA Microarrays: A Practical
Approach, ed. M. Schena. Oxford: Oxford University Press, pp. 1–16.
Schena, M., Shalon, D., Davis, R. W., Brown, P. O. (1995). Quantitative monitoring of gene
expression patterns with a complementary DNA microarray. Science 270: 467–470.
Schena, M., Shalon, D., Heller, R., et al. (1996). Parallel human genome analysis: microarray-based
expression monitoring of 1000 genes. Proc Natl Acad Sci USA 93: 10614–10619.
Schneider, A., Montague, P., Griffiths, I., et al. (1992). Uncoupling of hypomyelination and glial
cell death by a mutation in the proteolipid protein gene. Nature 358: 758–761.
Sklar, P. (2001). Microarray analysis of the Stanley brain collection. Abstract. In 7th Symposium on
the Neurobiology and Neuroimmunology of Schizophrenia and Bipolar Disorder. Washington,
DC: Stanley Foundation.
Southern, E., Mir, K., Shchepinov, M. (1999). Molecular interactions on microarrays. Nat Genet
21: 5–9.
Vawter, M. P., Barrett, T., Cheadle, C., et al. (2001). Application of cDNA microarrays to examine
gene expression differences in schizophrenia. Res Bull 55: 641–650.
Vawter, M. P., Crook, J. M., Hyde, T. M., et al. (2002). Microarray analysis of gene expression in
the prefrontal cortex in schizophrenia: a preliminary study. Schizophr Res 58: 11–20.
Volk, D. W., Austin, M. C., Pierri, J. N., Sampson, A. R., Lewis, D. A. (2000). Decreased
GAD67 mRNA expression in a subset of prefrontal cortical GABA neurons in subjects with
schizophrenia. Arch Gen Psychiatry 57: 237–245.
Waterworth, D. M., Bassett, A. S., Brzustowicz, L. M. (2002). Recent advances in the genetics of
schizophrenia. Cell Mol Life Sci 59: 331–348.
Weickert, C. S., Hyde, T. M., Lipska, B. K., et al. (2003). Reduced brain-derived neurotrophic
factor in prefrontal cortex of patients with schizophrenia. Mol Psychiatry 8: 592–610.
13
In the 1950s and 1960s, there was much extravagant discussion of the role of social
factors in the etiology of schizophrenia. However, there was little scientific basis
to this speculation, and it was swept away by the demonstration that people with
schizophrenia showed abnormalities of brain structure on computed tomographic
scans (Johnstone et al., 1976). A decade later, the neurodevelopmental model of
schizophrenia was proposed, and it subsequently became the dominant etiological
and pathogenetic model (Murray and Fearon, 1999; Murray et al., 1992). As a
result of these two developments, researchers have come to regard schizophrenia as
a brain disease, and social factors have been largely ignored as putative etiological
agents.
It is increasingly clear, however, that the neurodevelopemental model, an essen-
tially neurological concept, does not explain all the available data about schizophrea-
nia. One consequence has been a revival during the 1990s, particularly in Europe, of
research into the role of social factors as causal agents in schizophrenia. It is oppor-
tune, therefore, to draw together this disparate research and to examine critically
whether any of it stands up to scientific scrutiny. We will review both those social
factors that are postulated to operate early in life and those which may act more
proximal to the onset of the disorder (see Table 13.1 for an overview). First we will
briefly consider how animal research has informed on psychiatric conditions.
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
224
225 Social factors and development of schizophrenia
Unwanted child OR 2.4 (1.2–4.8) DSM III-R Birth cohort Myhrman et al.
schizophrenia (1996)
Poor mothering OR 2.65 (1.2–5.6) Schizophreniform Birth cohort Cannon et al. (2002)
disorders DSM IV
Early parental loss OR 3.8∗ DSM III-R Case–control Agid et al. (1999)
schizophrenia
Childhood abuse OR 7.3 (1.1–49) Positive psychotic Population Janssen et al. (2003)
symptoms cohort
reaching clinical
significance
City birth RR 2.4 (2.13–2.7) ICD 8 schizophrenia Population Mortensen et al.
cohort (1999)
City upbringing RR 2.75 (2.31–3.28) ICD 8 schizophrenia Population Pedersen and
cohort Mortensen (2001)
Ethnic minority IRR 4.4 (2.49–7.75) RDC Schizophrenia Ecological Boydell et al. (2001)
status
Racial harassment OR 2.86 (1.69–4.83) Psychosis diagnosed Cross-sectional Karlsen and Nazroo
(verbal abuse) by semistructured (2002)
interview
CI, confidence interval; OR, odds ratio; RR, relative risk; IRR, incidence rate ratio; DSM III-R, Diagnostic and
Statistical Manual III–Revised; ICD, International Statistical Classification of Diseases, Injuries, and Causes of
Death.
∗
p = 0.001.
evidence for the role of social factors in brain development has come from animal
experiments. For example, there is ample evidence that rats reared in isolation for a
period after birth have structural and physiological differences in their hippocampi
compared with rats reared in groups (Whitaker-Azmitia et al., 2000). Isolation
reared rats also show behavioral changes, anxiety, learning deficits, and sensory
changes. Furthermore, Matthews et al. (2001) found increased levels of dopamine
in the dorsal and ventral striatum in adult rats who were separated from their
mothers early in life. There is also evidence that rats reared in isolation, but not
other rats, respond to antipsychotic drugs in such a way as to normalize their
behavior (Heidbreder et al., 2001).
Therefore, by analogy, these data suggest that the social factors in the envi-
ronment of the developing child may similarly affect physical brain development.
These factors are now being revisited by psychiatric researchers searching for the
brain–environment interactions that shape vulnerability to conditions such as
226 J. Boydell, J. van Os, and R. M. Murray
psychosis. For example, experiences embedded in social relationships may alter the
prefrontal brain systems that mediate emotional self-regulation (Lyons et al., 2002);
similarly, the early social environment appears to program aspects of neurobiolog-
ical development that, in turn, affect behavioral, emotional, cognitive, and physio-
logical development (Sanchez et al., 2001). The early social environment has been
shown to induce synaptic changes that may be indicative, and perhaps the cause,
of alterations of behavioral and cognitive capacities (Ovtscharoff and Braun, 2001).
Although much of this work comes from animal research, there is evidence that
the early social environment can mediate the establishment of neural networks
that regulate a child’s response to stress and capacity for self-control (DiPietro,
2000). Furthermore, studies of exposure to institutional deprivation have shown
dramatic effects on a range of child behavioral outcomes, with clear dose–response
relationships between adverse outcomes and length of exposure (Beckett et al.,
2002). As Eisenberg (1995) states “The human brain is constructed socially.”
Family factors
Mother–child relationship
The British 1946 cohort study followed up all children (5362) born in 1 week in
the UK. By age 43, there were 30 cases of schizophrenia. The quality of the mother-
child relationship at 4 years of age, as rated by health visitors, was one of the most
powerful risk factors for later schizophrenia; a poor relationship (less skilled and
less understanding of the child) carried a sixfold increase in risk of schizophrenia
(Jones et al., 1994). Similarly, the Dunedin study (Cannon et al., 2002), which
followed up about 1000 children from birth to age 26 with comprehensive medical
and psychiatric assessments, found that the mothers of children who developed
schizophreniform disorders were rated as having poorer attitudes and behavior
towards their children at age 3 (odds ratio [OR], 2.65; 95% confidence interval
[CI], 1.2–5.6).
The British 1946 and the Dunedin studies are particularly interesting as they are
population-based and not high-risk studies. However, we do not know the direction
of the relationship. Is the increased risk an effect of poor mothering or is it that the
preschizophrenic child could not form a normal relationship with the mother?
Unwantedness
Several studies have claimed that being an unwanted child is associated with emo-
tional and social deprivation (Kubicka et al., 1995; Myhrman, 1992). Myhrman
et al. (1996), therefore, investigated whether unwantedness of pregnancy was a
risk factor for schizophrenia in the child as defined in the Diagnostic and Statistical
227 Social factors and development of schizophrenia
Manual III (DSM III-R; American Psychiatric Association, 1987). Their sample was
the Northern Finland 1966 Birth Cohort, which comprised 11 017 individuals who
were followed up until they were 28. Their mothers had been asked by a midwife
(when 6 or 7 months pregnant) whether the pregnancy was wanted, mistimed but
wanted, or unwanted. The risk of schizophrenia among the unwanted group was
considerably raised even after adjusting for sociodemographic, pregnancy (includ-
ing depression), and perinatal variables (OR, 2.4; 95% CI, 1.2–4.8). Unfortunately,
genetic risk was not considered in the study and, as with much social research, there
may have been residual confounding.
Communal upbringing
In the Israeli High Risk Study, Mirsky et al. (1985) carried out a follow-up study of
46 children with high genetic risk, half of whom were brought up communally
228 J. Boydell, J. van Os, and R. M. Murray
on a kibbutz (where their parents lived) and half in family homes, and com-
pared them with controls. Children with known genetic risk for schizophrenia
were significantly more likely to develop a psychotic disorder and, interestingly,
also an affective disorder if they were brought up on a kibbutz, rather than a
family home. The control kibbutz children did not have a higher risk (although
the numbers were small), suggesting genetic vulnerability to the social environ-
ment in the high-risk children. Similar findings emerged from the Copenhagen
High Risk Project, which followed up 207 high-risk children and compared them
with controls (Mednick et al., 1987). The high-risk children who had been sep-
arated from their parents were either brought up in public care institutions or
foster placements with families. The latter group had a better outcome (especially
for males) but of course the placement was not random. Nevertheless, these two
high-risk studies are among the few studies into social factors during childhood
that are less likely to have been confounded by the inherited characteristics of the
child.
Expressed emotion
It is well known that social overstimulation (Wing, 1978) and high expressed emo-
tion within families (Vaughn and Leff, 1976, 1982) are associated with relapse in
schizophrenia. Some assume that these factors are also implicated in the first episode
(Wing, 1978). As far as we are aware, there is no evidence to support or refute
this.
Childhood abuse
Several investigators have claimed an association between trauma and psychosis
(Read et al., 2001). First, studies of patients with psychosis have demonstrated that
they recall a high incidence of trauma in their lifetimes (Ross et al., 1994), and those
229 Social factors and development of schizophrenia
City upbringing
It is important but very difficult to separate place of birth and place of upbringing.
Astrup and Odegard (1961) found a stronger effect of city upbringing amongst
those who had moved to the city. A recent analysis of the Danish population cohort
just described has now addressed this question. Pedersen and Mortensen (2001)
used a comprehensive national registration system that accurately recorded every
change of residence to show that schizophrenia risk increased with the number of
years (between 0 and 20 years of age) that an individual lived in an urbanized area,
and with increasing degree of urbanization. Relative risk for those who had spent
their entire childhood in the capital was 2.75 (95% CI, 2.31–3.28) compared with
those who had always lived in the most rural areas. There was no evidence that
urbanicity was particularly toxic for any particular age group. The dose–response
relationship found in this study suggests that etiological factors of a pervasive and
long-term (either continuous or repeated) nature are operating in urbanized areas.
areas. However, this effect cannot account for the results of recent studies that
looked at place of birth and upbringing. For example, Dauncey and colleagues
(1993) investigated where patients had lived 5 years before their first admission
in Nottingham, UK and were unable to find evidence for systematic geographic
drift. Furthermore, it is unlikely that the drift occurred in the previous parental
generation, as the magnitude of this movement would need to have been extremely
high to explain the findings. For example, in the Danish study, Mortensen and
colleagues (1999) calculated that nearly 50 000 children born in the capital and
its suburbs needed to have a parent who transmitted a genetic risk equal to that
transmitted by a parent with diagnosed schizophrenia to account for the urban
excess. Furthermore, a family history of schizophrenia did not explain or effect the
urban–rural difference in this and other studies (van Os et al., 2002). This is also
relevant for the social residue theory (that those at greater risk are left behind in
an area as it becomes less desirable because they do not have the resources to move
out).
Possible explanations
No adequate explanation has been found for the urban excess of schizophrenia.
Infectious agents were once considered likely etiological factors but they are unlikely
to account for much of the risk-increasing effect, given the fact that the urban envi-
ronment is not only associated with increased rates of schizophrenia but also with
increases in much more prevalent expressions of non-clinical psychotic experi-
ences (van Os et al., 2001). Other physical factors associated with city living such
as lead pollution require further investigation. However, while there is no direct
evidence that the urban excess is caused by social factors, the fact that its effect is
so widespread across the population is compatible with the notion that part of the
excess risk represents a psychological reaction to factors in the wider social environ-
ment. Certainly some of the major differences between urban and rural areas are
to do with social cohesion and social support. For example, it has been shown that
the physical and mental health of children growing up in an urban environment is
adversely influenced by low levels of social cohesion in the neighborhood (Drukker
et al., 2003), and within the city, the incidence of schizophrenia has been shown to
vary as a function of the social neighborhood environment (van Os et al., 2000).
Although the evidence for an early effect is strong, there may also be an effect
of urbanization around the time of onset for non-early-onset schizophrenia and
there is evidence of a cumulative effect of urban exposure throughout childhood
(Castle et al., 1993). As we will see below, social isolation, deprivation, and adverse
life events are associated with increased risk of psychosis. As individuals in big cities
may be more likely to be exposed to these factors (Takano and Nakamura, 2001), it is
possible that some of the urban excess might be attributed to such factors. However,
it is likely that the social factors that increase the risk do not act independently of
232 J. Boydell, J. van Os, and R. M. Murray
genetic risk, and a recent report suggested that the urban environment acts by
increasing the likelihood that schizophrenia is expressed in those who carry genetic
susceptibility (van Os et al., 2003).
Social isolation
During childhood
Numerous reports from Bleuler onwards have shown that children destined to
develop schizophrenia tend to be solitary, lack friends, and indeed often prefer to
be alone. For example, the British 1946 cohort study of Jones et al. (1994), described
above, found that preference for solitary play at age 4 and 6 was associated with
later schizophrenia (OR, 2.1 and 2.5, respectively). Self-reported anxiety at age 13
and teacher-rated anxiety at age 15 both showed linear associations with later risk
for schizophrenia (Jones et al., 1994).
to make social relationships, and then the social isolation itself may propel them
further toward frank psychosis.
At time of onset
Hare (1956) reported that social isolation, as measured by proportion of single
person households in a geographical area, was associated with increased rates of
schizophrenia. The findings were not accounted for by movement of people with,
or developing, schizophrenia into the area. There has been a recent resurgence of
interest in these findings. Thornicroft et al. (1993) noted that clustering of individ-
uals with schizophrenia in deprived areas occurs only in urban areas and suggested
that social isolation is an important mediator of this. However, it is difficult to
distinguish between cause and effect in this context. Related to these ideas is the
theory that disruption of social networks decreases an individual’s capacity to cope
with psychosocial stress and increases the risk of schizophrenia.
Van Os et al. (2000) found that people who were single had a slightly higher
risk of developing psychosis if they lived in a neighborhood with fewer single
people compared with a neighborhood with many other single people. The authors
suggested that single status might give rise to perceived (or actual) social isolation
if most other people are living with a partner. The question of whether social
isolation may increase risk of schizophrenia (or whether a close relationship may
be protective) is also raised by Jablensky and Cole (1997), who showed that marriage
had a protective effect and that this was not simply a consequence of better adjusted
males being able to marry.
Everywhere you are surrounded by people with strange and unfamiliar ways. . . . They do not
seem to be as friendly as the people at home and many of them do their best to profit by your lack
of experience. Even if you have not had any disagreeable experiences yourself, your imagination
is stirred by all the stories you have heard about how crooked and dangerous they may be.
You notice that your own appearance, clothing and language points you out to everyone as a
greenhorn.
Studies from different countries and continents have now shown that the inci-
dence of psychosis is high amongst many migrant groups (Castle et al., 1991;
234 J. Boydell, J. van Os, and R. M. Murray
Harrison et al., 1988, 1997; King et al., 1994; Murphy, 1977; Selten and Sijben,
1994; Selten et al., 1998, 2001; Thomas et al., 1993; van Os et al., 1996a,b). Even
when migration between very similar cultures is considered, there is still evidence of
an increased risk. For example, Bruxner et al. (1997) found higher rates of hospital
admission with psychosis in British, Irish and southern European migrants to West.
Australia; the risk persisted many years after migration.
Methodological issues
There are many methodological pitfalls in migration and minority group research,
for example the “category fallacy” (that the categories of mental illness used in
one culture cannot be applied to another), misdiagnosis (whether psychiatrists in
the host country may wrongly diagnose schizophrenia in migrants because of preju-
dice or a lack of understanding of their cultural norms), or inaccurate estimation of
the denominator (migrants may be under-represented in general population data).
However, studies that have overcome these problems have still found an excess of
psychosis among migrant groups (Hickling et al., 1999; Mortensen et al., 1997;
Sharpley et al., 2001; van Os et al., 1996a,b; Wessely et al., 1991).
Increased rates of schizophrenia amongst migrants and ethnic minorities might
also be attributed (at least in part) to the urban effect, as most migrants live in cities
and often in the most deprived areas. However, Malzberg (1969) found that con-
trolling for urbanicity reduced but did not explain the excess of psychosis amongst
migrants to the USA, and similar findings were reported more recently in the
Netherlands (van Os et al., 2001)
African-Caribbeans in the UK
Most work has investigated the increased rates of psychosis in the African-
Caribbeans in the UK and the Netherlands. Genetic predisposition cannot be the
sole explanation since the increased risk is not shared by the population of origin
in the Caribbean (Bhugra et al., 1997; Mahy et al., 1999); in addition the morbid
risk for second-generation siblings is much higher than for their first-generation
equivalents (Hutchinson et al., 1996; Sugarman and Crawford, 1994). Selective
migration has also been largely ruled out. For example, Selten et al. (2002) found
that, even if the entire Surinamese population had migrated to the Netherlands,
this still would not account for the absolute number of cases amongst Surinam
migrants unless the incidence had increased. Increased exposure, or susceptibil-
ity, to neurodevelopmental insult such as obstetric complications (Glover, 1989;
Hutchinson et al., 1997) and viral infections (Selten et al., 1998) have largely been
excluded as possible explanations. McGuire et al. (1995) found no difference in drug
abuse between their white and African- Caribbean patients in South London. Simi-
larly, Selten et al. (1997) found that consumption of cannabis was lower amongst
235 Social factors and development of schizophrenia
Caribbean migrants to the Netherlands than amongst the native population, but
their incidence rate of schizophrenia was higher.
Discrimination
The combination of more affective symptoms and a less-deteriorated course has
led to the idea that factors in the social environment might cause psychosis in those
migrants predisposed but who might not otherwise develop the disease (McKenzie
et al., 1995). Racism (overt and institutionalized), social isolation, and reduced
social networks are among the factors that some consider may contribute (Hutchin-
son et al., 1999). Boydell et al. (2001) have found that incidence rates of schizophre-
nia increased in ethnic minorities as the proportion of ethnic minorities in the
locality fell, suggesting social experience (perhaps of isolation or discrimination)
contributes to the development of the disorder.
Janssen et al. (2003) measured subjective experience of discrimination and sub-
sequent development of psychotic illness 3 years later. Experience of discrimination
strongly predicted for the development of delusional ideation (OR, 2.3; 95% CI,
236 J. Boydell, J. van Os, and R. M. Murray
1.2–4.2). In another analysis in the same sample, it was shown that the effect of
ethnic minority status on psychosis was no longer significant when controlling for
experience of discrimination (Janssen et al., 2002). Karlsen and Nazroo (2002) stud-
ied the experience of racial harassment and perception of discrimination among
a UK representative sample of 5000 people from ethnic minorities. They found
significantly increased OR values for a number of health problems but particularly
psychosis (experience of verbal abuse OR, 2.86 [95% CI, 1.69–4.83] experience of
physical attack OR, 4.77 [95% CI, 2.32–9.8]).
Some people may be more susceptible to the deleterious effects of such events
than others because they are less able to derive support from those around them
(Sharpley et al., 2001). Mallett et al. (1998) demonstrated that one of the main
distinguishing features of first-onset patients of Caribbean origin with psychosis in
London was that they lived alone and additionally had been separated from their
mother at an early age.
Unemployment
Bhugra et al. (1997) found that unemployment was particularly high amongst peo-
ple of Caribbean origin first presenting with schizophrenia in the UK. They then
went on to compare the unemployment rates in African-Carribean people present-
ing with their first episode in London and Trinidad, relative to local unemployment
rates. In London, the rate was much higher amongst the patients than in the local
population, but this was not the case in Trinidad (Bhugra et al., 2000). It is pos-
sible, therefore, that the high unemployment rate amongst African-Caribbeans in
London (and subsequent social isolation and stigmatization) might have etiological
relevance. Alternatively, the social milieu in Trinidad might be more sympathetic
to those at risk of developing schizophrenia.
Life events
The first study to find an association between schizophrenia and life events reported
an excess of events limited to 3 weeks before onset (Brown and Birley, 1968).
Prospective studies have also found an association between life events and relapse
of psychosis (Malla et al., 1990; Ventura et al., 1989). Bebbington (2000) carried out
a comprehensive case–control study in London and found a significant excess of
events and, importantly, independent life events in the 3 months prior to onset of
schizophrenia. Not all studies have concurred however (Chung et al., 1986; Jacobs
and Myers, 1976).
There is evidence that the kinds of stress that affect individuals with schizophrenia
vulnerability are not so much the major life events but rather the hassles and stress
that individuals experience in the flow of daily life (Malla et al., 1990). Studies using
237 Social factors and development of schizophrenia
intensive field methods to examine subjective experience of stress in the flow of daily
life have found that individuals with schizophrenia, and individuals with genetic
susceptibility to schizophrenia, display greater levels of emotional reactivity to small
daily life stressors than do control subjects (Myin-Germeys et al., 2001). Individuals
with schizophrenia may be more sensitive to such stressors than individuals with
affective disorder (Myin-Germeys et al., 2003a). In addition, part of the sensitivity
to daily life stressors may be determined by prior exposure to major life events
(Myin-Germeys et al., 2003b), suggesting synergistic environment–environment
interactions.
Other research suggests that some individuals carry traits that make them more
likely to experience life events, such as the personality trait neuroticism (van Os and
Jones, 1999). This finding appears to be relevant to schizophrenia, as several studies
have demonstrated that neuroticism is a risk factor for schizophrenia (Krabbendam
et al., 2002; van Os and Jones, 2001) and is more prevalent in the first-degree relatives
of patients with schizophrenia (Maier et al., 1994). Therefore, the role of life events
and daily life hassles as a risk factor for schizophrenia is complex. Stress may be
generated in part by underlying personality traits, whose genetic contribution may
overlap with that of schizophrenia. Alternatively, individuals with vulnerability to
schizophrenia may be more sensitive to the effects of stress, while sensitivity to stress
is also determined by the degree of prior exposure to stress, possibly including stress
in early life (Janssen et al., 2004).
(i.e. most people exposed to each of the risk factors remain well and never develop
the illness). A unifying explanation seems to be that the environmental factors
operate upon genetic risk. There are many forms that this interaction could take:
synergistic, additive, multiplicative (van Os and Sham, 2003). We have reviewed
evidence from the Finnish and Danish adoption studies and the Israeli High Risk
Study that the social environment can operate on genetic factors to increase risk
of schizophrenia. Genes might also influence the likelihood of exposure to some
social risk factors. According to the model proposed by van Os and Marcelis (1998),
individuals vary in their sensitivity to adverse environmental circumstances, and
genetically sensitive individuals are more likely to develop psychiatric illness when
exposed to certain environments than those without genetic predisposition. In the
case of schizophrenia, being different in some way from one’s neighbors, and/or
experiencing discrimination seem to be important factors.
Conclusions
A range of social factors have been reported to be implicated in the etiology of
schizophrenia, but the evidence for many of them remains scanty. Even for those
where the evidence is more substantial (e.g. urban living, social isolation, and
discrimination), it is unclear how these factors contribute to the risk. Indeed, it is
likely that more than one mechanism operates, and there may be many different
interactions with pre-existing genetic/neurodevelopmental vulnerabilities. It is now
clear, however, that, in order to understand the causes of schizophrenia, the role of
the social environment cannot continue to be ignored. In saying this, we are not
proposing an oppositional social instead of biological approach, which we consider
as futile as arguing whether poverty or mycobateria cause tuberculosis! Rather, we
suggest that both social and biological factors need to be studied as well as their
interaction.
We need to recognize that (i) social factors can impact on brain development,
(ii) some social factors give rise to psychological vulnerabilities, and (iii) many
social factors act over the life course, creating developmental liabilities. We have
presented evidence that early social adversity can affect brain development. For
example, structural abnormalities can be demonstrated in adult life in individuals
with a history of childhood abuse (Anderson et al., 2002; Bremner et al., 1997; Stein
et al., 1997). Now that evidence is emerging that childhood abuse may contribute
to schizophrenia risk, it is attractive to speculate that some of the neuroimaging
findings associated with abuse are relevant to schizophrenia; for example, decreased
hippocampal volume has been reported in both. It is possible that the social environ-
ment creates psychological vulnerabilities that act additively to the risk function
in combination with genetic or non-genetic neurodevelopmental impairments.
For example, chronic exposure to discrimination may give rise to a psychological
vulnerability to delusion formation, which in individuals with additional neurode-
velopmental impairment may result in overt psychotic disorder. Finally, it is also
240 J. Boydell, J. van Os, and R. M. Murray
REFERENCES
Agid, O., Shapira, B., Zislin, J., et al. (1999). Environment and vulnerability to major psychiatric
illness: a case–control study of early parental loss in major depression, bipolar disorder and
schizophrenia. Mol Psychiatry 4: 163–172.
Allardyce, J., Boydell, J., van Os, J., et al. (2001). A comparison of the incidence of schizophrenia
in rural Dumfries and Galloway and urban Camberwell 179: 335–340.
American Psychiatric Association (1987). Diagnostic and Statistical Manual III–Revised.
Washington, DC: American Psychiatric Press.
Anderson, C. M., Teicher, M. H., Polcari, A., et al. (2002). Abnormal T2 relaxation time in the
cerebellar vermis of adults sexually abused in childhood: potential role of the vermis in
stress-enhanced risk for drug abuse. Psychoneuroendocrinology 27: 231–244.
Astrup C, Odegard, O. (1961). Internal migration and mental illness in Norway. Psychiatr Q 34:
116–130.
Bateson, G., Jackson, D., Haley, J., et al. (1956). Toward a theory of schizophrenia. Behav Sci 1:
251–264.
Bebbington, P. (2003). In Schizophrenia – not just biological. In Proceedings of Conference held
at the Institute of Psychiatry. London: Institute of Psychiatry, p. 2.
Beckett, C., Bredenkamp, D., Castle, J., et al. (2002). Behavior patterns associated with institutional
deprivation: a study of children adopted from Romania. J Dev Behav Pediatr 23: 297–303.
Berenbaum, H. (1999). Peculiarity and reported childhood maltreatment. Psychiatry 62: 21–35.
Bhugra, D., Leff, J., Mallett, R., et al. (1997). Incidence and outcome of schizophrenia in Whites,
African Caribbeans and Asians in London. Psychol Med 27: 791–798.
Bhugra, D., Hilwig, M., Mallett, R., et al. (2000). Factors in the onset of schizophrenia: a com-
parison between London and Trinidad samples. Acta Psychiatr Scand 101: 135–141.
Blackwood, N., Howard, R., Bentall, R., et al. (2001). Cognitive neuropsychiatric models of
persecutory delusions. Am J Psychiatry 158: 527–539.
241 Social factors and development of schizophrenia
Boardman, A., Hodgson, R., Lewis, M., et al. (1997). Social indicators and the prediction of
psychiatric admission in different diagnostic groups. Br J Psychiatry 171: 457–462.
Boydell, J., van Os, J., McKenzie, K., et al. (2001). Incidence of schizophrenia in ethnic minorities
in London: ecological study into interactions with environment. Br Med J 7325: 1336–1338.
Boydell, J., van Os, J., McKenzie, K., Murray, R. (2003). The influence of inequality on the
incidence of schizophrenia: an ecological study. Schizophr Bull 60: Suppl. 33.
Bremner, J. D., Randall, P., Vermetten, E., et al. (1997). Magnetic resonance imaging-based
measurement of hippocampal volume in posttraumatic stress disorder related to childhood
physical and sexual abuse: a preliminary report. Bio Psychiatry 41: 23–32.
Brown, G. W., Birley, J. L. (1968). Crises and life changes and the onset of schizophrenia. J Health
Soc Behav 9: 203–214.
Bruxner, G., Burvill, P., Fazio, S., et al. (1997). Aspects of psychiatric admissions of migrants to
hospitals in Perth. West Aust N Z J Psychiatry 31: 532–542.
Bryer, J. B., Nelson, B. A., Miller, J. B., et al. (1987). Childhood sexual and physical abuse as
factors in adult psychiatric illness. Am J Psychiatry 144: 1426–1430.
Butler, R. W., Mueser, K. T., Sprock, J., et al. (1996). Positive symptoms of psychosis in posttrau-
matic stress disorder. Biol Psychiatry 39: 839–844.
Cannon, M., Caspi, A., Moffitt, T., et al. (2002). Evidence for early-childhood, pan-developmental
impairment specific to schizophreniform disorder. Arch Gen Psychiatry 59: 449–456.
Castle, D., Wessley, S., Der, G., Murray, R. M. (1991). The incidence of operationally defined
schizophrenia in Camberwell, 1965–1984. Br J Psychiatry 159: 790–794.
Castle, D. J., Scott, K., Wessely, S., et al. (1993). Does social deprivation during gestation and
early life predispose to later schizophrenia? Soc Psychiatry Psychiatr Epidemiol 28: 1–4.
Chung, R. K., Langeluddecke, P., Tennant, C., et al. (1986). Threatening life events in the onset of
schizophrenia, schizophreniform psychosis and hypomania. Br J Psychiatry 148: 680–685.
Croudace, T. J., Kayne, R., Jones, P. B., et al. (2000). Non-linear relationship between an index of
social deprivation, psychiatric admission prevalence and the incidence of psychosis. Psychol
Med 30: 177–185.
Dauncey, K., Giggs, J., Baker, K., et al. (1993). Schizophrenia in Nottingham: lifelong residential
mobility of a cohort. Br J Psychiatry 163: 613–619.
DiPietro, J. A. (2000). Baby and the brain: advances in child development. Annu Rev Publ Health
21: 455–471.
Dohrenwend, B. P., Levav, I., Shrout, P. E., et al. (1992). Socioeconomic status psychiatric disorders
the causation-selection issue. Science 255: 946–952.
Drukker, M., Kaplan, C. D., Feron, F. J. M., et al. (2003). Children’s health-related quality of life,
neighbourhood socio-economic deprivation and social capital. A contextual analysis. Soc
Sci Med 57: 825–841.
Eisenberg, L. (1995). The social construction of the human brain. Am J Psychiatry 152: 1563–
1575.
Ellason, J. W., Ross, C. A. (1997). Childhood trauma and psychiatric symptoms. Psychol Rep 80:
447–450.
Farris, R., Dunham, H. (1939). Mental Disorders in Urban Areas. Chicago, IL: University of
Chicago Press.
242 J. Boydell, J. van Os, and R. M. Murray
Janssen, I., Krabbendam, L., Bak, M., et al. (2004). Childhood abuse as a risk factor for psychotic
experience. Acta Psychiatr Scand in press.
Johns, L. C., van Os, J. (2001). The continuity of psychotic experiences in the general population.
Clin Psychol Rev 21: 1125–1141.
Jones, P., Rodgers, B., Murray, R., et al. (1994). Child developmental risk factors for adult
schizophrenia in the British 1946 birth cohort. Lancet 344: 1398–1402.
Johnstone, E. C., Crow, T. J., Frith, C. D., et al. (1976). Cerebral ventricular size and cognitive
impairment in chronic schizophrenia. Lancet, ii: 924–926.
Karlsen, S., Nazroo, J. (2002). Relation between racial discrimination and health among ethnic
minority groups. Am J Publ Health 92: 624–631.
King, M., Coker, E., Leavey, G. et al. (1994). Incidence of psychotic illness in London: a comparison
of ethnic groups. Br Med J 309: 1115–1119.
Koppel, S., McGuffin, P. (1999). Socioeconomic factors that predict psychiatric admissions. Psy-
chol Med 29: 1235–1241.
Krabbendam, L., Janssen, I., Bak, M., et al. (2002). Neuroticism and low self-esteem as risk factors
for psychosis. Soc Psychiatry Psychiatr Epidemiol 37: 1–6.
Kubicka, L., Matejcek, Z., David, H.P., et al. (1995). Children from unwanted pregnancies in
Prague, Czech Republic revisted at age 30. Acta Psychiatr Scand 91: 361–369.
Lewis, G., David, A., Andreasson, S., et al. (1992). Schizophrenia and city life. Lancet 340: 137–
140.
Lidz, T. (1958). Schizophrenia and the family. Psychiatry 21: 21–27.
Lyons, D. M., Afarian, H., Schatzberg, A. F., et al. (2002). Experience-dependent asymmetric
variation in primate prefrontal morphology. Behav Brain Res 136: 51–59.
Mahy, G., Mallett, R., Leff, J., Bhugra, D. (1999). First-contact incidence rate of schizophrenia on
Barbados. Br J Psychiatry 175: 28–33.
Maier, W., Minges, J., Lichtermann, D., et al. (1994). Personality variations in healthy relatives of
schizophrenics. Schizophr Res 12: 81–88.
Malla, A. K., Cortese, L., Shaw, T. S., et al. (1990). Life events and relapse in schizophrenia. A one
year prospective study. Soc Psychiatry Psychiatr Epidemiol 25: 221–224.
Mallett, R., Hutchinson, G., Leff, J., et al. (1998). Social conditions and schizophrenia in African-
Caribbeans: Edward Hare revisited. In Proceedings of Winter Meeting 39 of the Royal College
of Psychiatrists. London: Royal College of Psychiatrists.
Mallett, R., Leff, J., Bhugra, D., Pang, D., Zhao, J. (2002). Social environment, ethnicity and
schizophrenia. A case-control study. Soc Psychiatry Psychiatr Epidemiol 37: 329–335.
Malmberg, A., Lewis, G., David, A., et al. (1998). Premorbid adjustment and personality in people
with schizophrenia. Br J Psychiatry 172: 308–313.
Malzberg, B. (1969). Are immigrants psychologically disturbed? In Changing Perspectives in Men-
tal illness, ed. S. Plog, R. Edgerton. New York: Holt, Rinehart and Winston, pp. 395–421.
Marcelis, M., Navarro-Mateu, F., Murray, R., et al. (1998). Urbanization and psychosis: a study
of 1942–1978 birth cohorts in the Netherlands. Psychol Med 28: 871–879.
Marcelis, M., Takei, N., van Os, J., et al. (1999). Urbanization and risk for schizophrenia: does
the effect operate before or around the time of illness onset? Psychol Med 29: 1197–1203.
244 J. Boydell, J. van Os, and R. M. Murray
Matthews, M. K., Dalleny, J. W., Matthews, C., et al. (2001). Periodic maternal separation of
neonatal rats produces region- and genetic-specific effects on bioamine content in post-
mortem adult brain. Synapse 40: 1–10
McGuire, P., Jones, P., Harvey, M., et al. (1995). Morbid risk of schizophrenia in relatives of
patients with cannabis-associated psychosis. Schizophr Res 15: 277–281.
McKenzie, K., van Os, J., Fahy, T., et al. (1995). Psychosis with good prognosis in Afro-Caribbean
people now living in the United Kingdom. [See comments] Br Med J 311: 1325–1328.
Mednick, S.A., Parnas, J., Schulsinger, F., et al. (1987). The Copenhagen High Risk Project 1962–
1986. Schizophr Bull 133: 485–495.
Mirsky, A. F., Silberman, E. K., Latz, A., et al. (1985). Adult outcomes of high-risk children:
differential effects of town and kibbutz rearing. Schizophr Bull 11: 150–156.
Mortensen, P. B., Cantor-Graae, E., McNeil, T. F., et al. (1997). Increased rates of schizophrenia
among immigrants: some methodological concerns raised by Danish findings. Psychol Med
274: 813–820.
Mortensen, P., Pedersen, C., Westergaard, T., et al. (1999). Effects of family history and place and
season of birth on the risk of schizophrenia. N Eng J Med 340: 603–608. [Sec correspondence
in N Engl J Med (1999) 341: 370–372].
Moser, K. (2001). Inequalities in treated heart disease and mental illness in England and Wales
1994–1998. Br J Gen Pract 51: 438–444.
Murphy, H. B. (1977). Migration, culture and mental health. Psychol Med 7: 677–684.
Murray, R., Fearon, P. (1999). The developmental “risk factor” model of schizophrenia. J Psychiatr
Res 33: 497–499.
Murray, R. M., O’Callaghan, E., Castle, D. J., et al. (1992). A neurodevelopmental approach to
the classification of schizophrenia. Schizophr Bull 18: 319–332.
Myhrman, A. (1992). Unwanted pregnancy, its occurrence and significance for the family and
the child. Acta Univ Ouluen Ser 249: 1–46.
Myhrman, A., Rantakallio, P., Isohanni, M., et al. (1996). Unwantedness of pregnancy and
schizophrenia in the Child. Br J Psychiatry 169: 637–640.
Myin-Germeys, I., van Os, J., Schwartz, J. E., et al. (2001). Emotional reactivity to daily life stress
in psychosis. Arch Gen Psychiatry 58: 1137–1144.
Myin-Germeys, I., Peeters, F., Havermans, R., et al. (2003a). Emotional reactivity to daily life stress
in psychosis and affective disorder: an experience sampling study. Acta Psychiatr Scand 107:
124–131.
Myin-Germeys, I., Krabbendam, L., Delespaul, P. A., et al. (2003b). Do life events have their
effect on psychosis by influencing the emotional reactivity to daily life stress? Psychol Med
33: 327–333.
Ødegård, O. (1932). Emigration and insanity: a study of mental disease among Norwegian born
population in Minnesota. Acta Psychiatr Neurol Scand 7(Suppl. 4): 1–206.
Ovtscharoff, Jr., W., Braun, K. (2001). Maternal separation and social isolation modulate the
postnatal development of synaptic composition in the infralimbic cortex of Octodon degus.
Neuroscience 104: 33–40.
Pedersen, C. B., Mortensen, P. B. (2001). Evidence of a dose–response relationship between
urbanicity during upbringing and schizophrenia risk. Arch Gen Psychiatry 58: 1039–1046.
245 Social factors and development of schizophrenia
Putnam, F. W., Guroff, J. J., Silberman, E. K., et al. (1986). The clinical phenomenology of multiple
personality disorder: review of 100 recent cases. J Clin Psychiatry 47: 285–293.
Read, J., Argyle, N. (1999). Hallucinations, delusions, and thought disorder among adult psychi-
atric inpatients with a history of child abuse. Psychiatr Serv 50: 1467–1472.
Read, J., Perry, B. D., Moskowitz, A., et al. (2001). The contribution of early traumatic events to
schizophrenia in some patients: a traumagenic neurodevelopmental model. Psychiatry 64:
319–345.
Read, J., Agar, K., Argyle, N., et al. (2003). Sexual and physical abuse during childhood and
adulthood as predictors of hallucinations, delusions and thought disorder. Psychol Psychother
Theory Res Pract 76: 1–22.
Rosenthal, D., Wender, P., Kety, S., et al. (1971). The adopted-away offspring of schizophrenics.
Am J Psychiatry 128: 307–311.
Ross, C. A., Joshi, S. (1992). Schneiderian symptoms and childhood trauma in the general popu-
lation. Compr Psychiatry 33: 269–273.
Ross, C. A., Norton, G. R., et al. (1989). Multiple personality disorder: an analysis of 236 cases.
Can J Psychiatry Rev Can Psychiatry 34: 413–418.
Ross, C. A., Anderson, G., Clark, P., Wozney, K. (1994). Childhood abuse and the positive symp-
toms of schizophrenia. Hosp Community Psychiatry 45: 489–491.
Sanchez, M. M., Ladd, C. O., Plotsky, P. M. (2001). Early adverse experience as a developmen-
tal risk factor for later psychopathology: evidence from rodent and primate models. Dev
Psychopathol 13: 419–449.
Sansonnet-Hayden, H., Haley, G., Marriage, K., et al. (1987). Sexual abuse and psychopathology
in hospitalized adolescents. J Am Acad Child Adolesc Psychiatry 26: 753–757.
Selten, J. P., Sijben, N. (1994). First admission rate for schizophrenia in immigrants to the Nether-
lands: the Dutch National Register. Soc Psychiatry Psychiatr Epidemiol 29: 71–72.
Selten, J. P., Slaets, J. P., Kahn, R. S., et al. (1997). Schizophrenia in Surinamese and Dutch
Antillean immigrants to the Netherlands: evidence of an increased incidence. Psychol Med
27: 807–811.
(1998). Prenatal exposure to influenza and schizophrenia in Surinamese and Dutch Antillean
immigrants to the Netherlands. Schizophr Res 30: 101–103.
Selten, J. P., Veen, N., Feller, W., et al. (2001). Incidence of psychotic disorders in immigrant
groups to the Netherlands. Br J Psychiatry 178: 1–7.
Selten, J. P., Cantor-Graae, E., Slaets, J., et al. (2002). Ødegårds selection hypothesis revisited:
schizophrenia in Surinamese immigrants to the Netherlands. Am J Psychiatry 159: 669–671.
Sharpley, M., Hutchinson, G., McKenzie, K., et al. (2001). Understanding the excess of psychosis
among the African-Caribbean population in England. Review of current hypotheses. Br J
Psychiatry 178(Suppl. 40): 60–68.
Startup, M. (1999). Schizotypy, dissociative experiences and childhood abuse: relationships
among self-report measures. Br J Clin Psychol 38: 333–344.
Stein, M. B., Koverola, C., Hanna, C., et al. (1997). Hippocampal volume in women victimized
by childhood sexual abuse. Psychol Med 27: 951–959.
Sugarman, P., Crawford, D. (1994). Schizophrenia in the Afro-Caribbean community. Br J Psy-
chiatry 164: 474–480.
246 J. Boydell, J. van Os, and R. M. Murray
Swett, Jr., C. , Surrey, J., Cohen, C., et al. (1990). Sexual and physical abuse histories and psychiatric
symptoms among male psychiatric outpatients. Am J Psychiatry 147: 632–636.
Takano, T., Nakamura, K. (2001). An analysis of health levels and various indicators of urban
environments for healthy cities projects. J Epidemiol Community Health 55: 263–270.
Thomas, C. S., Stone, K., Osborn, M., et al. (1993). Psychiatric morbidity and compulsory
admission among UK born Europeans, Afro-Caribbeans and Asians in central Manchester.
Br J Psychiatry 163: 91–99.
Thornicroft, G., Bisoffi, G., De Salva, D., et al. (1993). Urban–rural differences in the associa-
tions between social deprivation and psychiatric service utilization in schizophrenia and all
diagnoses: a case-register study in Northern Italy. Psychol Med 23: 487–496.
Tienari, P. (1991). Interaction between genetic vulnerability and family environment: The Finnish
adoptive family study of schizophrenia. Acta Psychiatrica Scand 84: 460–465.
Tienari, P., Wynne, L., Laksy, K., et al. (2000). Finnish Adoptive Family Study. Acta Psychiatr
Scand 101 433–443.
van Os, J., Jones, P. B. (1999). Early risk factors and adult person–environment relationships in
affective disorder. Psychol Med 29: 1055–1067.
(2001). Neuroticism as a risk factor for schizophrenia. Psychol Med 31: 1129–1134.
van Os, J., Marcelis, M. (1998). The ecogenetics of schizophrenia: a review. Schizophr Res 32:
127–135.
van Os, J., Sham, P. (2003). Gene–environment correlation and interaction in schizophrenia. In
The Epidemiology of Schizophrenia, ed. R. Murray, P. Jones, E. Susser, J. van Os, M. Cannon.
Cambridge, UK: Cambridge. University Press.
van Os, J., Castle, D. J., Takei, N., et al. (1996a). Psychotic illness in ethnic minorities: clarification
from the 1991 census. Psychol Med 26: 203–208.
van Os, J., Takei, N., Castle, D. J., et al. (1996b). The incidence of mania: time trends in relation
to gender and ethnicity. Soc Psychiatry Psychiatr Epidemiol 31: 129–136.
van Os, J., Driessen, G., Gunther, N., et al. (2000). Neighbourhood variation in incidence of
schizophrenia. Evidence for person–environment interaction. Br J Psychiatry 176: 243–248.
van Os, J., Hanssen, M., Bijl, R. V., et al. (2001). Prevalence of psychotic disorder and com-
munity level of psychotic symptoms: an urban–rural comparison. Arch Gen Psychiatry 58:
663–668.
van Os, J., Hanssen, M., de Graaf, R., et al. (2002). Does the urban environment independently
increase the risk for both negative and positive features of psychosis? Soc Psychiatry Psychiatr
Epidemiol 37: 460–464.
van Os, J., Hanssen, M., Bak, M., et al. (2003). Do urbanicity and familial liability coparticipate
in causing psychosis? Am J Psychiatry 160: 477–482.
Vaughn, C. E., Leff, J. P. (1976). The influence of family and social factors on the course of
psychiatric illness. Br J Psychiatry 129: 125–137.
(1982). Patterns of emotional responses in the relatives of schizophrenic patients. Schizophr
Bull 7: 43–44.
Ventura, J., Nuechterlein, K. H., Lukoff, D., et al. (1989). A prospective study of stressful life
events and schizophrenic relapse. J Abnorm Psychol 98: 407–411.
247 Social factors and development of schizophrenia
Wahlberg, K. E., Wynne, L., Oja, H., et al. (1997). Gene–environment interaction in vulnerability
to schizophrenia: findings from the Finnish Adoptive Family Study of Schizophrenia. Am J
Psychiatry 154: 355–362.
Wahlberg, K. E., Wynne, L., Oja, H., et al. (2000). Thought disorder index of Finnish adoptees
and communication deviance of their adoptive parents. Psychol Med 30: 127–136.
Weingarten, M. A., Orron, D. E. (1983). Schizophrenia in a Yemenite immigrant town in Israel.
Int J Soc Psychiatry 29: 249–254.
Wessely, S., Castle, D., Der, R., et al. (1991). Schizophrenia and Afro-Caribbeans. A case-control
study. Br J Psychiatry 159: 795–801.
Whitaker-Azmitia, P., Zhou, F., Hobin, J., et al. (2000). Isolation rearing of rats produces deficits
as adults in the serotonergic innervation of hippocampus. Peptides 21: 1755–1759.
Wiesel, T. N., Hubel, D. H. (1974). Ordered arrangement of orientation columns in monkeys
lacking visual experience. J Comp Neurol 158: 307–318.
Wing, J. K. (1978). The social context of schizophrenia. Am J Psychiatry 135: 1333–1339.
World Health Organization (1967). Manual of International Statistical Classification of Diseases,
Injuries and Causes of Death, 8th edn. Geneva: World Health Organization.
Wynne, L., Ryckoff, I., Day, J., et al. (1958). Pseudo-mutuality in the family relations of
schizophrenics. Psychiatry 21: 205–220.
14
248
249 Drug abuse and development of schizophrenia
Elsewhere in the world, rates of substance abuse are generally lower, but again
consumption is more frequent in those with psychosis than in the rest of the popu-
lation. In the UK, in South London, the 1-year prevalence rate among patients with
psychotic illnesses was found to be 36.3% for any substance problem and 15.8%
for drug problems (Menezes et al., 1996). A more recent study from Scotland
(McCreadie, 2002) revealed that problem use of drugs by people with schizophre-
nia was greater than in the general population for both the previous year (7% versus
2%) and at any time previously (20% versus 6%). Cantwell et al. (1999) examined
168 subjects with first-episode psychosis in the UK and reported that 37% of the
sample met criteria for drug use, drug misuse, or alcohol misuse; 8.4% of the sub-
jects received a primary diagnosis of substance-related psychotic disorder. The rates
of substance use among individuals with schizophrenia in Continental Europe are
similar to this and rarely as high as those reported in US studies (Cassano et al.,
1998; Modestin et al., 1997; Soyka et al., 1993; Verdoux et al., 1996a).
Self-medication
A long-standing notion has been that substance abuse is often the product of
psychopathology, resulting from the patient’s attempt to “self-medicate” dysphoric
symptoms (Khantzian, 1985). Patients suffering from schizophrenia are postulated
to use substances either to improve their negative, depressive, or anxiety symptoms
(DeQuardo et al., 1994; Dixon et al., 1991; Mueser et al., 1998; Schneier and Siris,
1987) or to alleviate the side effects of antipsychotic drugs (Dixon et al., 1991; Lopez
and Jeste, 1997). For example, small amounts of cannabis are said to attenuate
anxiety, depression, and negative symptoms in schizophrenia (Linszen et al., 1994;
Peralta and Cuesta, 1992). However, empirical studies (Fischman and Schuster,
1982; Griffith et al., 1972) on the effects of cannabis and stimulants have often
failed to produce any evidence to substantiate the self-medication hypothesis.
by cohort studies (Jones et al., 1994). So far there is little evidence for a common
genetic vulnerability for schizophrenia and drug abuse.
Vulnerability hypothesis
As with other common chronic diseases such as diabetes and coronary artery disease,
many consider that the appropriate etiological model (Murray and Fearon, 1999)
for schizophrenia is one involving multiple genes and environmental risk factors.
As discussed elsewhere in this book, genetic and/or early environmental factors are
postulated to cause the development of anomalous neural networks, which interact
in the growing child with inherited schizotypal traits to establish a trajectory towards
an increasingly solitary and deviant lifestyle. The vulnerablity hypothesis postulates
that stressors, including the use of drugs, then precipitate the individual across the
threshold for expression of psychosis (e.g. Bebbington et al., 1993; Jones, et al.,
1994; Tsuang et al., 2001; Verdoux et al., 1996b).
Amphetamine-induced psychosis
Amphetamine psychosis is a reaction that usually occurs following chronic use
of amphetamines, though it can occur after only a short period of amphetamine
consumption. Patients with amphetamine psychosis show a picture very similar
to paranoid schizophrenia (Bell, 1965; Connell, 1958). Indeed, amphetamine psy-
chosis so closely resembles schizophrenia, especially the paranoid type, that it has
been viewed as a model of schizophrenia (Crow et al., 1976; Snyder, 1973).
Connell (1958) and Bell (1965) originally conceptualized amphetamine psy-
chosis as lasting no more than 1 week after the urine becomes negative for
amphetamines. However, the psychosis does not always remit so readily, and use of
methamphetamine has been reported in several studies to result in psychotic symp-
toms lasting several months or more (Iwanami et al., 1994; Nakatani et al., 1989; Sato
et al., 1983; Tatetsu et al., 1956). Methamphetamine is a derivative of amphetamine,
251 Drug abuse and development of schizophrenia
Cannabis-induced psychosis:
Of the plant-based drugs, the illicit consumption of cannabis is most widespread
(United Nations International Drug Control Programme, 1997). Many studies
(Dixon et al., 1991; Linszen, et al., 1994; Longhurst, 1997; Mathers et al., 1991;
Rolfe et al., 1993; Schneier and Siris, 1987) have reported a positive association
between cannabis use and psychosis. Furthermore, in a prospective cohort study of
psychotic patients, Linszen et al. (1994) found that significantly more and earlier
relapses occurred in the cannabis-abusing group. In all but one of their subjects,
cannabis abuse preceded the onset of the first psychotic symptoms by at least
1 year. The authors, therefore, concluded that cannabis abuse, and particularly
heavy abuse, can elicit relapse in patients with schizophrenia and related disorders,
and it may possibly precipitate the initial psychosis.
Thornicroft (1990), in a review of the evidence for an association between
cannabis and psychosis, concluded that there is no convincing support for a separate
clinical diagnosis of “cannabis psychosis.” A retrospective study of 272 psychotic
inpatients (70 with cannabis-related psychosis) also failed to find any consistent
pattern of symptoms associated with cannabis use (Imade and Ebie, 1991). How-
ever, arguments against the use of this particular diagnostic label should not be
confused with arguments against any association between the drug and psychosis
252 C.-K. Chen and R. M. Murray
(Thomas, 1993). Chronic psychosis following prolonged cannabis abuse may reflect
the precipitation of schizophreniform disorder or schizophrenia in a vulnerable
individual, rather than a separate diagnosis of cannabis psychosis (Thornicroft,
1990). To establish conclusively whether cannabis consumption actually causes the
onset of schizophrenia, one must obtain prospective data and establish whether
there is evidence of a dose–response relationship between the amount consumed
and the risk of psychosis.
The first prospective cohort study was reported in 1987. Andreasson et al. (1987)
followed up almost 50 000 individuals conscripted into the Swedish Army and
reported the relative risk of developing schizophrenia was 2.4 for cannabis users
compared with non-users, rising to 6.0 for heavy users. An extension of this Swedish
Conscript Study has recently been carried out (Zammit et al., 2002). Consistent with
previous findings, this showed that “heavy cannabis users” by the age of 18 years
were 6.7 times more likely than non-users to be diagnosed with schizophrenia in the
ensuing 27 years. The risk was reduced but remained significant after controlling
for other potential confounding factors such as disturbed behavior, low intelligence
quotient score, growing up in a city, cigarette smoking, other drug use, and poor
social integration.
However, this study was largely ignored for 15 years. Then, in the Netherlands,
van Os and his colleagues (2002) examined the effect of cannabis use on psychotic
symptoms among the general population, assessing 4045 subjects at baseline and
again 3 years later. Compared with non-users, individuals using cannabis at baseline
were nearly three times more likely to manifest psychotic symptoms at follow-up.
This risk remained significant after statistical adjustment for a range of confounders.
There was a dose–response relationship with the highest risk (odds ratio, 6.81) being
observed for the highest level of cannabis use. Further analysis revealed that lifetime
history of cannabis use at baseline, as opposed to use of cannabis at follow-up, was a
stronger predictor of psychosis 3 years later than cannabis use at the later point. This
suggests that the association between cannabis use and psychosis was not merely a
short-term effect of cannabis use leading to an acute psychotic episode.
The Swedish and Dutch studies were open to criticism on a number of points
(Johnson et al., 1988; Negrete, 1989), notably that both the cannabis use and the
schizophrenia may have been related to an underlying predisposing factor such as
premorbid personality. This criticism has been at least partly overcome in a study
from New Zealand, which followed-up a general population birth cohort of 1037
individuals born in Dunedin, New Zealand in 1972–1973 (96% follow-up rate at age
26). Although much smaller, this study has information on self-reported psychotic
symptoms at age 11 years (i.e. before the likely onset of cannabis use) and then
information on cannabis use (self-reports) at 15 and 18 years. Thus, it allows the
age of onset of cannabis use to be examined in relation to later psychiatric outcome.
253 Drug abuse and development of schizophrenia
Results showed that both cannabis users by age 15 and cannabis users by age 18
had elevated schizophrenia symptoms at age 26, which remained significant after
controlling for psychotic symptoms predating the onset of cannabis use (Arseneault
et al., 2002). The effect was much stronger with earlier use, and onset of cannabis use
by age 15 was associated with an increased likelihood of meeting diagnostic criteria
for schizophreniform disorder at age 26. Indeed, 10.3% of those using cannabis at
age 15 in this cohort were diagnosed with schizophreniform disorder at age 26, as
opposed to 3% of the controls.
Phencyclidine-induced psychosis
PCP is a dissociative anesthetic originally employed in human anesthesia in the late
1950s. It can produce hallucinations, relaxation, feeling of dissociation from the
environment, and sometimes intense euphoria (Jacobs et al., 1987). Further char-
acterization of the psychomimetic actions of PCP was facilitated by the epidemic
of PCP abuse that occurred in the USA during the late 1960s and 1970s. Although
hallucinations, agitation, and paranoid delusions occur after PCP administration,
the most striking and consistent behavioral effects of PCP are alterations in body
image, disorganization of thought, negativism, and apathy (Javitt and Zukin, 1991).
Javitt and Zukin (1991) suggested that the symptoms induced by PCP are similar
to the “four A’s” proposed by Bleuler as the primary symptoms of schizophrenia:
affective blunting, ambivalence, autism, and disturbance of association. They also
suggested that PCP psychosis may provide a neurochemical model corresponding
uniquely to the Bleulerian conception of schizophrenia.
PCP interacts selectively with a specific binding site (PCP receptor) that is associ-
ated with the N-methyl-D-aspartate (NMDA)-type excitatory amino acid receptor.
Occupation of its receptor by PCP induces non-competitive inhibition of NMDA
receptor-mediated neurotransmission (Snell et al., 1988). Primary dysfunction of
NMDA receptor functioning could potentially account for the deficit in cognitive
functioning associated with schizophrenia, as well as for the dopaminergic hyper-
activity and dysregulation associated with acute schizophrenia (Kleinman et al.,
1988).
In short, PCP psychosis has been an attractive fashionable model for psychosis.
Unfortunately, there have been few studies of its role in precipitating long-lasting
schizophrenia-like psychoses in real life.
person will “taste” green, “smell” grey or “see” music (Jacobs et al., 1987; Slaby,
1991). However, the picture in some patients remains sufficiently complex to ren-
der LSD psychosis very difficult to distinguish from acute schizophrenia (Vardy
and Kay, 1983). Many believe that LSD alone does not produce an enduring psy-
chosis but rather precipitates schizophrenia in predisposed personalities (Slaby,
1991). Strassman (1984) suggested that the development of chronic LSD psychosis
is multifactorial in origin and that premorbid factors such as family history and per-
sonality characteristics, as well as exposure to a multitude of drugs, play important
roles.
I could not fail to note, however, that the individual disposition plays a major role in the effects of
cocaine, perhaps a more important role than with other alkaloids. The subjective phenomena after
ingestion of cocaine differ from person to person, and only a few persons experience, like myself, a
pure euphoria without alteration. Others already experience slight intoxication, hyperkinesia and
talkativeness after the same amount of cocaine, while still others have no subjective symptoms
of the effects of cocaine at all.
6.0
Mean PSST
5.5
5.0
4.5
4.0
3.5
MNP MIP-B MIP-P
Fig. 14.1. Premorbid schizoid/schizotypal traits between methamphetamine users with and without
psychosis. MNP, methamphetamine users with no experience of psychotic symptom; MIP-B,
subjects with brief methamphetamine psychosis; MIP-P, subjects with prolonged metham-
phetamine psychosis; PSST, score of Premorbid Schizoid and Schizotypal Traits.
Familial predisposition
There have been only a few competent studies of the relationship between familial
risk of psychosis and the occurrence and duration of drug-related psychoses. Tsuang
et al. (1982) revealed that the familial risks of schizophrenia and affective disorder
were greater in drug abusers with prolonged psychosis than those without psychosis
or with psychosis of short duration. McGuire et al. (1995) compared the lifetime
morbid risk of psychiatric disorder among the first-degree relatives of patients
admitted with acute psychosis who were cannabis positive on urinary screening
with that of psychotic controls who screened negatively for all substances. Patients
with cannabis-associated psychosis had a significantly increased familial morbid
risk for schizophrenia. This might be taken to imply that cannabis abuse acts on
genetic predisposition to trigger psychosis.
In an early study, Tatetsu et al. (1956) reported that the prevalence of schizophre-
nia in the families of methamphetamine psychotics (4.7% for siblings, 3.4% for
parents) was lower than that for schizophrenia patients (13.1% for siblings, 10.1%
for parents) but higher than that for normal controls (0.6% for siblings, 0.25 for
parents). Their interpretation was that methamphetamine psychosis is not geneti-
cally identical to schizophrenia, and that the higher prevalence of schizophrenia in
families of patients with methamphetamine psychosis than in families of normal
controls may have resulted from a small number of schizophrenia patients being
included in the group of methamphetamine psychotics. An alternative formulation
is that the psychosis predisposition of methamphetamine psychotics falls between
that of schizophrenia patients and that of the normal population.
We assessed 445 methamphetamine users with the Diagnostic Interview for
Genetic Studies and the Family Interview for Genetic Study. Familial morbid risk
for psychotic disorders and other mental illnesses of first-degree relatives was com-
pared after age correction between the methamphetamine users with a lifetime
diagnosis of methamphetamine psychosis and those without. This study obtained
psychiatric information for 1983 first-degree relatives of the methamphetamine
258 C.-K. Chen and R. M. Murray
users. The relatives of those with methamphetamine psychosis had higher morbid
risks for schizophrenia than those of methamphetamine abusers without psychosis
(3.1% versus 0.6%). The familial morbid risk for schizophrenia was even higher
(6.5%) among those with prolonged methamphetamine psychosis.
Therefore, it appears that familial loading is an important factor in determining
not only the occurrence of psychosis associated with drug abuse but also its dura-
tion. Furthermore, these findings are consistent with the notion of an underlying
continuum of genetic liability that has schizophrenia as only one of its possible
outcomes (Tsuang et al., 2001; van Os et al., 1998).
for the candidate genes of these disorders. It is likely that certain drugs change
the expressions of genes related to neurotransmitter systems such as dopamine or
glutamic acid and also for transcription factors, cell proliferation, apoptosis, cell
adhesion, and the synapse (Ito, 2002). Hopefully, identifying these alterable gene
expressions may facilitate the discovery of the genes that underlie the development
of drug-associated psychosis.
Dopamine sensitization
Behavioral sensitization is a phenomenon whereby exposure to a given stimu-
lus such as a drug or a stressor results in an enhanced response at subsequent
exposures (Wolf et al., 1993). Repeated cocaine use leads to progressive sensitiza-
tion, with short-term psychotic symptoms triggered more rapidly and after smaller
amounts of cocaine (Bartlett et al., 1997). The process of sensitization does not
necessarily require subsequent exposures to exactly the same stimulus that induced
the process. For example, Gorriti et al. (1999) showed that chronic treatment with
261 Drug abuse and development of schizophrenia
Conclusions
It is generally not disputed that the use of certain drugs, particularly cannabis
and psychostimulants, which have major effects on dopamine is associated with
schizophrenia. We regard the vulnerability hypothesis as the most likely explanation
for this association. Early environmental insults often compound the genotype for
psychosis to produce developmental deviance, but this is in itself sometimes not
sufficient to produce a psychotic illness. Later environmental factors, such as drug
abuse, then act on the underlying predisposition to precipitate frank psychosis.
262 C.-K. Chen and R. M. Murray
Recent research suggests that dopamine sensitization may underlie both craving
and the onset of drug-associated psychosis. Whether an individual develops psy-
chosis after drug abuse may be determined by various factors, such as drug-use
patterns, premorbid mental health, and the individual’s place on a continuum of
psychosis proneness. The liability for psychosis of each individual falls within a
normal distribution. A drug abuser with low liability to psychosis may use psy-
chostimulant drugs regularly for long periods without developing psychosis or, at
worst may have just brief psychotic symptoms. By comparison, an individual with
heavy liability may develop psychosis after using drugs only a few times and may
have a prolonged psychosis in spite of subsequently remaining abstinent from the
drug.
REFERENCES
Andreasson, S., Alleback, A., Engstrom, A., Rydberg, U. (1987). Cannabis and schizophrenia: a
longitudinal study of Swedish conscripts. Lancet i: 1483–1485.
Angrist, B. M., Gershon, S. (1970). The phenomenology of experimentally induced amphetamine
psychosis: preliminary observations. Biol Psychiatry 2: 95–107.
Arndt, S., Tyrrell, G., Flaum, M., Andreasen, N. C. (1992). Comorbidity of substance abuse and
schizophrenia: the role of premorbid adjustment. Psychol Med 22: 379–388.
Arseneault, L., Cannon, M., Poulton, R., et al. (2002). Cannabis use in adolescence and risk for
adult psychosis: longitudinal prospective study. Br Med J 325: 1212–1213.
Bartlett, E., Hallin, A., Chapman, B., Angrist, B. (1997). Selective sensitization to the psychosis-
inducing effects of cocaine: a possible marker for addiction relapse vulnerability? Neuropsy-
chopharmacology 16: 77–82.
Battaglia, M., Cavallini, M. C., Macciardi, F., Bellodi, L. (1997). The structure of DSM-III-R
schizotypal personality disorder diagnosed by direct interviews. Schizophr Bull 23: 83–92.
Bebbington, P., Wilkins, S., Jones, P., et al. (1993). Life events and psychosis. Initial results from
the Camberwell Collaborative Psychosis Study. Br J Psychiatry 162: 72–79.
Bell, D. S. (1965) Comparison of amphetamine psychosis and schizophrenia. Br J Psychiatry III:
701–707.
(1973). The experimental reproduction of amphetamine psychosis. Arch Gen Psychiatry 29:
35–40.
Berger, N., Vaillancourt, C., Boksa, P. (2000). Genetic factors modulate effects of C-section birth
on dopaminergic function in the rat. Neuroreport 11: 639–643.
Blanchard, J. J., Brown, S. A., Horan, W. P., Sherwood, A. R. (2000). Substance use disorders in
schizophrenia: review, integration, and a proposed model. Clin Psychol Rev 20: 207–234.
Boyle, M. H., Offord, D. R., Racine, Y. A., et al. (1992). Predicting substance use in late adolescence:
results from the Ontario child health study follow-up. Am J Psychiatry 149: 761–767.
Braff, D. L., Grillon, C., Geyer, M. A. (1992). Gating and habituation of the startle reflex in
schizophrenic patients. Arch Gen Psychiatry 49: 206–215.
263 Drug abuse and development of schizophrenia
Brake, W. G., Boksa, P., Gratton, A. (1997). Effects of perinatal anoxia on the acute locomotor
response to repeated amphetamine administration in adult rats. Psychopharmacology 133:
389–395.
Breakey, W. R., Goodell, H., Lorenz, P. C., McHugh, P. R. (1974). Hallucinogenic drugs as pre-
cipitants of schizophrenia. Psychol Med 4: 255–261.
Breier, A., Su, T. P., Saunders, R., et al. (1997). Schizophrenia is associated with elevated
amphetamine-induced synaptic dopamine concentrations: evidence from a novel positron
emission tomography method. Proc Natl Acad Sci USA 94: 2569–2574.
Camp, D. M., Browman, K. E., Robinson, T. E. (1994). The effects of methamphetamine and
cocaine on motor behavior and extracellular dopamine in the ventral striatum of Lewis
versus Fischer 344 rats. Brain Res 668: 180–193.
Cannon, T. D., Kaprio, J., Lonnqvist, J., Huttunen, M., Koskenvuo, M. (1998). The genetic
epidemiology of schizophrenia in a Finnish twin cohort. A population-based modeling
study. Arch Gen Psychiatry 55: 67–74.
Cantor-Graae, E., McNeil, T. F., Sjostrom, K., Nordstrom, L. G., Rosenlund, T. (1994). Obstetric
complications and their relationship to other etiological risk factors in schizophrenia. A
case–control study. J Nerv Ment Dis 182: 645–650.
Cantwell, R., Brewin, J., Glazebrook, C., et al. (1999). Prevalence of substance misuse in first-
episode psychosis. Br J Psychiatry 174: 150–153.
Cardno, A. G., Marshall, E. J., Coid, B., et al. (1999). Heritability estimates for psychotic disorders:
the Maudsley twin psychosis series. Arch of Gen Psychiatry 56: 162–168.
Carlsson, A. (1988). The current status of the dopamine hypothesis of schizophrenia. Neuropsy-
chopharmacology 1: 179–186.
Cassano, G. B., Pini, S., Saettoni, M., Rucci, P., Dell’Osso, L. (1998). Occurrence and clinical
correlates of psychiatric comorbidity in patients with psychotic disorders. J Clin Psychiatry
59: 60–68.
Chen, C. K., Lin, S. K., Sham, P., et al. (2003). Premorbid characteristics and comorbid-
ity of methamphetamine users with and without psychosis. Psychol Med 33: 1407–
1414.
Chou, P., Liou, M. Y., Lai, M. Y., Hsiao, M. L., Chang, H. J. (1999). Time trend of substance use
among adolescent students in Taiwan, 1991–1996. J Formos Med Assoc 98: 827–831.
Connell, P. H. (1958). Amphetamine Psychosis London: Chapman, Hall.
Crow, T. J., Johnstone, E. C., Deakin, J. F., Longden, A. (1976). Dopamine and schizophrenia.
Lancet ii: 563–566.
Cubells, J. F., Kranzler, H. R., McCance-Katz, E., et al. (2000). A haplotype at the DBH locus,
associated with low plasma dopamine beta-hydroxylase activity, also associates with cocaine-
induced paranoia. Mol Psychiatry 5: 56–63.
DeQuardo, J. R., Carpenter, C. F., Tandon, R. (1994). Patterns of substance abuse in schizophrenia:
nature and significance. J Psychiatr Res 28: 267–275.
Dixon, L., Haas, G., Weiden, P. J., Sweeney, J., Frances, A. J. (1991). Drug abuse in schizophrenic
patients: clinical correlates and reasons for use. Am J Psychiatry 148: 224–230.
Duaux, E., Gorwood, P., Griffon, N., et al. (1998). Homozygosity at the dopamine D3 receptor
gene is associated with opiate dependence. Mol Psychiatry 3: 333–336.
264 C.-K. Chen and R. M. Murray
Dumas, P., Saoud, M., Bouafia, S., et al. (2002). Cannabis use correlates with schizotypal person-
ality traits in healthy students. Psychiatry Res 109: 27–35.
Ebstein, R. P., Novick, O., Umansky, R., et al. (1996). Dopamine D4 receptor (D4DR) exon III
polymorphism associated with the human personality trait of novelty seeking. Nat Genet
12: 78–80.
Ewing, J. A., Mueller, R. A., Rouse, B. A., Silver, D. (1977). Low levels of dopamine beta-hydroxylase
and psychosis. Am J Psychiatry 134: 927–928.
Fischman, M. W., Schuster, C. R. (1982). Cocaine self-administration in humans. Fed Proc 41:
241–246.
Flaum, M., Schultz, S. K. (1996). When does amphetamine-induced psychosis become
schizophrenia? [Clinical conference] Am J Psychiatry 153: 812–815.
Franke, P., Nothen, M. M., Wang, T., et al. (2000). DRD4 exon III VNTR polymorphism-
susceptibility factor for heroin dependence? Results of a case–control and a family-based
association approach. Mol Psychiatry 5: 101–104.
Freud, S. (1885). On the general effects of cocaine. [Lecture before Psychiatric Union, 5 March,
1885. Translated to English.] Drug Depend 5: 15–17.
Geddes, J. R., Lawrie, S. M. (1995). Obstetric complications and schizophrenia: a meta-analysis.
Br J Psychiatry 167: 786–793.
Gold, M. S., Bowers, Jr. M. B. (1978). Neurobiological vulnerability to low-dose amphetamine
psychosis. Am J Psychiatry 135: 1546–1548.
Gorriti, M. A., de Rodriguez, F., Navarro, M., Palomo, T. (1999). Chronic (−)-delta9-
tetrahydrocannabinol treatment induces sensitization to the psychomotor effects of
amphetamine in rats. Eur J Pharmacol 365: 133–142.
Griffith, J. D., Cavanaugh, J., Oates, J. A. (1969). Schizophreniform psychosis induced by large-
dose administration of d-amphetamine. J Psychedel Drugs 2: 42–48.
Griffith, J. D., Cavanaugh, J., Held, J., Oates, J. A. (1972). Dextroamphetamine. Evaluation of
psychomimetic properties in man. Arch Gen Psychiatry 26: 97–100.
Guillin, O., Diaz, J., Carroll, P., et al. (2001). BDNF controls dopamine D3 receptor expression
and triggers behavioral sensitization. Nature 411: 86–89.
Harrison, P. J., Owen, M. J. (2003). Genes for schizophrenia? Recent findings and their patho-
physiological implications. Lancet 361: 417–419.
Imade, A. G. T., Ebie, J. C. (1991). A retrospective study of symptom patterns of cannabis-induced
psychosis. Acta Psychiatr Scand 83: 134–136.
Ito, C. (2002). Analysis of overall gene expression induced by amphetamine and phencyclidine:
novel targets for the treatment of drug psychosis and schizophrenia. Curr Pharm Des 8:
147–153.
Iwanami, A., Suga, I., Kaneko, T., Sugiyama, A., Nakatani, Y. (1994). P300 component of event-
related potentials in methamphetamine psychosis and schizophrenia. Prog Neuropsychophar-
macol Biol Psychiatry 18: 465–475.
Jacobs, M. R., Fehr, K. O., Cox, T. C., et al. (1987). Drugs and Drug Abuse. A Reference Text.
Toronto: Addiction Research Foundation.
Javitt, D. C., Zukin, S. Z. (1991). Recent advances in the phencyclidine model of schizophrenia.
Am J Psychiatry 148: 1301–1308.
265 Drug abuse and development of schizophrenia
Johnson, B. A., Smith, B. L., Taylor, P. (1988). Cannabis and schizophrenia. Lancet i: 593.
Jones, P., Rodgers, B., Murray, R., Marmot, M. (1994). Child developmental risk factors for adult
schizophrenia in the British 1946 birth cohort. Lancet 344: 1398–1402.
Jori, A., Garattini, S. (1973). Catecholamine metabolism and amphetamine effects on sensitive
and insensitive mice. In Frontiers in Catecholamine Research, ed. E. Usdin, S. H. Snyder. New
York: Pergamon Press, pp. 939–941.
Kendler, K. S., Diehl, S. R. (1993). The genetics of schizophrenia: a current, genetic-epidemiologic
perspective. Schizophr Bull 19: 261–285.
Khantzian, E. J. (1985). The self-medication hypothesis of addictive disorders: focus on heroin
and cocaine dependence. Am J Psychiatry 142: 1259–1264.
Kikuchi, K., Inada, T., Iijima, Y., et al. (2001). Association between dopamine D1 receptor fam-
ily (DRD1, DRD5) gene polymorphisms and methamphetamine psychosis. In Proceed-
ings of the International College of Neuro-Psychopharmacology Regional Meeting, Hiroshima,
p. 398.
Kleinman, J. E., Casanova, F. M., Jaskiv, G. E. (1988). The neuropathology of schizophrenia.
Schizophrenia Bulletin 14: 209–216.
Konuma, K. (1994). Use and abuse of amphetamines in Japan. In Amphetamine and its Analogs
pp. 415–435.
Kotler, M., Cohen, H., Segman, R., et al. (1997). Excess dopamine D4 receptor (D4DR) exon III
seven repeat allele in opioid-dependent subjects. Mol Psychiatry 2: 251–254.
Krebs, M. O., Sautel, F., Bourdel, M. C., et al. (1998). Dopamine D3 receptor gene variants and
substance abuse in schizophrenia. Mol Psychiatry 3: 337–341.
Kwapil, T. R. (1996). A longitudinal study of drug and alcohol use by psychosis-prone and
impulsive-nonconforming individuals. J Abnorm Psychol 105: 114–123.
Laruelle, M. (2000). The role of endogenous sensitization in the pathophysiology of schizophrenia:
implications from recent brain imaging studies. Brain Res Brain Res Rev 31: 371–384.
Lewis, S. W., Murray, R. M. (1987). Obstetric complications, neurodevelopmental deviance, and
risk of schizophrenia. J Psychiatr Res 21: 413–421.
Li, T., Zhu, Z. H., Liu, X., et al. (2000). Association analysis of polymorphisms in the DRD4 gene
and heroin abuse in Chinese subjects. Am J Med Genet 96: 616–621.
Lieberman, J. A., Kane, J. M., Alvir, J. (1987). Provocative tests with psychostimulant drugs in
schizophrenia. Psychopharmacology 91: 415–433.
Lieberman, J. A., Sheitman, B. B., Kinon, B. J. (1997). Neurochemical sensitization in the patho-
physiology of schizophrenia: deficits and dysfunction in neuronal regulation and plasticity.
Neuropsychopharmacology 17: 205–229.
Linszen, D. H., Dingemans, P. M., Lenior, M. E. (1994). Cannabis abuse and the course of recent-
onset schizophrenic disorders. Arch of Gen Psychiatry 51: 273–279.
Lipska, B. K., Jaskiw, G. E., Chrapusta, S., Karoum, F., Weinberger, D. R. (1992). Ibotenic acid
lesion of the ventral hippocampus differentially affects dopamine and its metabolites in the
nucleus accumbens and prefrontal cortex in the rat. Brain Res 585: 1–6.
Lipska, B. K., Halim, N. D., Segal, P. N., Weinberger, D. R. (2002). Effects of reversible inactivation
of the neonatal ventral hippocampus on behavior in the adult rat. J Neurosci 22: 2835–
2842.
266 C.-K. Chen and R. M. Murray
Persico, A. M., Bird, G., Gabbay, F. H., Uhl, G. R. (1996) D2 dopamine receptor gene TaqI A1 and
B1 restriction fragment length polymorphisms: enhanced frequencies in psychostimulant-
preferring polysubstance abusers. Biol Psychiatry 40: 776–784.
Pilla, M., Perachon, S., Sautel, F., et al. (1999). Selective inhibition of cocaine-seeking behavior
by a partial dopamine D3 receptor agonist. Nature 400: 371–375.
Post, R. M. (1975). Cocaine psychosis: a continuum model. Am J Psychiatry 132: 225–231.
Ralph, R. J., Varty, G. B., Kelly, M. A., et al. (1999). The dopamine D2 , but not D3 or D4 , receptor
subtype is essential for the disruption of prepulse inhibition produced by amphetamine in
mice. J Neurosci 19: 4627–4633.
Regier, D. A., Farmer, M. E., Rae, D. S., et al. (1990). Comorbidity of mental disorders with alcohol
and other drug abuse. Results from the Epidemiologic Catchment Area (ECA) Study. J Am
Med Assoc 264: 2511–2518.
Rolfe, M., Tang, C. M., Sabally, S., et al. (1993). Psychosis and cannabis abuse in The Gambia. A
case–control study. Br J Psychiatry 163: 798–801.
Sato, M., Chen, C. C., Akiyama, K., Otsuki, S. (1983). Acute exacerbation of paranoid psychotic
state after long-term abstinence in patients with previous methamphetamine psychosis. Biol
Psychiatry 18: 429–440.
Schneier, F. R., Siris, S. G. (1987). A review of psychoactive substance use and abuse in schizo-
phrenia. Patterns of drug choice. J Nerv Ment Dis 175: 641–652.
Sekine, Y., Iyo, M., Ouchi, Y., et al. (2001). Methamphetamine-related psychiatric symptoms
and reduced brain dopamine transporters studied with PET. Am J Psychiatry 158: 1206–
1214.
Selzer, J. A., Lieberman, J. A. (1993). Schizophrenia and substance abuse. Psychiatr Clin North
Am 16: 401–412.
Slaby, A. E. (1991). Dual diagnosis: fact or fiction? In Dual Diagnosis in Substance Abuse, ed. M.
S. Gold, A. E. Slaby. New York: Marcel Dekker, pp. 3–28.
Snell, L. D., Yi, S.-J., Johnson, K. M. (1988). Comparison of the effects of MK-801 and phencycli-
dine on catecholamine uptake and NMDA-induced norepinephrine release. Eur J Pharmacol
145: 223–226.
Snyder, S. H. (1973). Amphetamine psychosis: a “model” schizophrenia mediated by cate-
cholamines. Am J Psychiatry 130: 61–67.
Soyka, M., Albus, M., Kathmann, N., et al. (1993). Prevalence of alcohol and drug abuse in
schizophrenic inpatients. Eur Arch Psychiatry Clin Neurosci 242: 362–372.
Staley, J. K., Mash, D. C. (1996). Adaptive increase in D3 dopamine receptors in the brain reward
circuits of human cocaine fatalities. J Neurosci 16: 6100–6106.
Stefanis, N., Frangou, S., Yakeley, J., et al. (1999). Hippocampal volume reduction in schizophre-
nia: effects of genetic risk and pregnancy and birth complications. Biol Psychiatry 46:
697–702.
Strassman, R. J. (1984). Adverse reactions to psychedelic drugs. A review of the literature. J Nerv
Ment Dis 172: 577–595.
Suwaki, H., Fukui, S., Konuma, K. (1997) Methamphetamine abuse in Japan: its 45 year history
and the current situation. In Amphetamine Misuse: International Perspective on Current
Trends, ed. H. Klee. Amsterdam: Harwood Academic, pp. 199–214.
268 C.-K. Chen and R. M. Murray
Takahashi, M., Shirakawa, O., Toyooka, K., et al. (2000). Abnormal expression of brain-derived
neurotrophic factor and its receptor in the corticolimbic system of schizophrenic patients.
Mol Psychiatry 5: 293–300.
Tarter, R. E., Laird, S. B., Kabene, M., Bukstein, O., Kaminer, Y. (1990). Drug abuse severity in
adolescents is associated with magnitude of deviation in temperament traits. Br J Addiction
85: 1501–1504.
Tatetsu, S. (1963). Methamphetamine. Folia Psychiatr Neurol Jap Suppl 7: 377–380.
Tatetsu, S., Goto, A., Fujiwara, T. (1956). The Methamphetamine Psychosis. Tokyo: Igakushoin.
Thomas, H. (1993). Psychiatric symptoms in cannabis users. Br J Psychiatry 163: 141–149.
Thornicroft, G. (1990). Cannabis and psychosis: is there epidemiological evidence for an associ-
ation? Br J Psychiatry 157: 25–33.
Tsuang, M. T., Simpson, J. C., Kronfol, Z. (1982). Subtypes of drug abuse with psychosis. Demo-
graphic characteristics, clinical features, and family history. Arch Gen Psychiatry 39: 141–147.
Tsuang, M. T., Stone, W. S., Faraone, S. V. (2001). Genes, environment and schizophrenia. Br J
Psychiatry 178: 18–24.
Ujike, H., Harano, M., Inada, T., et al. (2003). Nine- or fewer repeat alleles in VNTR poly-
morphism of the dopamine transporter gene is a strong risk factor for prolonged metham-
phetamine psychosis. Pharmacogenom J 3: 242–247.
United Nations International Drug Control Programme (1997). World Drug Report. New York:
Oxford University Press.
Vaillancourt, C., Boksa, P. (1998). Caesarean section birth with general anesthesia increases
dopamine-mediated behavior in the adult rat. Neuroreport 9: 2953–2959.
(2000). Birth insult alters dopamine-mediated behavior in a precocial species, the guinea pig.
Implications for schizophrenia. Neuropsychopharmacology 23: 654–666.
van Os, J., Jones, P., Sham, P., Bebbington, P., Murray, R. M. (1998). Risk factors for onset and
persistence of psychosis. Soc Psychiatry Psychiatr Epidemiol 33: 596–605.
van Os, J., Bak, M., Hanssen, M., et al. (2002). Cannabis use and psychosis: a longitudinal
population-based study. Am J Epidemiol 156: 319–327.
Vardy, M. M., Kay, S. R. (1983). LSD psychosis or LSD-induced schizophrenia? A multimethod
inquiry. Arch Gen Psychiatry 40: 877–883.
Verdoux, H., Mury, M., Besancon, G., Bourgeois, M. (1996a). [Comparative study of substance
dependence comorbidity in bipolar, schizophrenic and schizoaffective disorders.] Encephale
22: 95–101.
Verdoux, H., van Os, J., Sham, P., et al. (1996b). Does familiality predispose to both emergence
and persistence of psychosis? A follow-up study. Br J Psychiatry 168: 620–626. [Published
erratum appears in Br J Psychiatry (1996). 169: 116].
Verdoux, H., Gindre, C., Sorbara, F., Tournier, M., Swendsen, J. D. (2002). Cannabis use and the
expression of psychosis vulnerability in daily life. Schizophr Res 53 (Suppl.): 225.
Volkow, N. D., Chang, L., Wang, G. J., et al. (2001). Association of dopamine transporter
reduction with psychomotor impairment in methamphetamine abusers. Am J Psychiatry
158: 377–382.
Williams, J. H., Wellman, N. A., Rawlins, J. N. (1996). Cannabis use correlates with schizotypy
in healthy people. Addiction 91: 869–877.
269 Drug abuse and development of schizophrenia
Wolf, M. E., White, F. J., Nassar, R., Brooderson, R. J., Khansa, M. R. (1993). Differential develop-
ment of autoreceptor subsensitivity and enhanced dopamine release during amphetamine
sensitization. J Pharmacol Exp Ther 264: 249–255.
Zammit, S., Allebeck, P., Andreasson, S., Lundberg, I., Lewis, G. (2002). Self reported cannabis
use as a risk factor for schizophrenia in Swedish conscripts of 1969: historical cohort study.
Br Med J 325: 1199.
Part III
Pathophysiology
15
Evidence suggests that the dopamine (DA) system plays a key role in the pathophys-
iology and treatment of schizophrenia. Therefore, drugs that increase DA trans-
mission are known to exacerbate this disorder and mimic paranoid psychosis in
normal individuals (Angrist et al., 1974, 1980), and drugs that are effective antipsy-
chotic agents all have DA receptor-blocking properties in common (Creese et al.,
1976; Grace et al., 1997; Seeman, 1987). However, despite substantial effort, there
has been little direct evidence for a pathology within at least the subcortical DA
system (Beuger et al., 1996; Post et al., 1975; Van Kammen et al., 1986). Nonetheless,
schizophrenia patients show significantly higher levels of DA release in the stria-
tum in response to low doses of amphetamine, particularly in patients in whom the
amphetamine leads to an exacerbation of their psychosis (Laruelle, 2000; Laruelle
and Abi-Dargham, 1999). This has led to the suggestion that the DA system may
be abnormally regulated in the brains of schizophrenia patients, leading to the
psychopathological state. One system in particular that has attracted attention as a
potential source of this dysregulatory influence is the cortical glutamatergic system.
This is based on evidence that drugs that interfere with glutamate transmission exert
a symptom-specific exacerbation of schizophrenia in susceptible patients and will
mimic the entire spectrum of schizophrenia symptoms in some normal individuals
(Goff and Coyle, 2001; Jentsch and Roth, 1999; Olney and Farber, 1995). There is
also evidence for a pathology within the limbic cortex of schizophrenia patients
(Weinberger, 1999).
Substantial evidence exists to suggest that the pathological processes contributing
to schizophrenia occur early in life, supporting the contention that schizophrenia
is a developmental disorder. Thus, the incidence of schizophrenia is higher in indi-
viduals with a genetic predisposition to the disorder, with the rate of occurrence
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
273
274 A. A. Grace
as much as 33% in this structure in the brains of schizophrenia patients (Akil et al.,
1999). Together, these data suggest that pathology within the prefrontal cortex may
be an etiological factor in the onset of schizophrenia. However, although prefrontal
cortical lesions in adults may mimic some of the negative symptoms of schizophre-
nia, there is little evidence that such an insult will lead to psychosis.
stress. Studies have shown that exposure to a stressor leads to increased DA release
in the prefrontal cortex, with substantially smaller increases in DA within subcor-
tical structures (Abercrombie et al., 1989; Imperato et al., 1989). However, if the
DA innervation of the prefrontal cortex is disrupted, two consequences are noted.
First, there is a decrease in the spontaneous activity of ventral tegmental DA neu-
rons (Harden et al., 1998); second, there is an increase in the stress-induced DA
response within subcortical limbic structures such as the nucleus accumbens (King
et al., 1997).
Together, these studies suggest that, in the absence of a normally functioning
prefrontal cortex, the system would over-react to potentially anxiogenic stimuli,
resulting in increased levels of stress. Indeed, such a model is consistent with obser-
vations of high-risk individuals by E. C. Johnstone (personal communication), who
found that those adolescents who eventually go on to develop schizophrenia typ-
ically show a higher reactivity to stress compared with those who do not develop
psychosis. What consequence would such an increase in stress produce? One region
that may be of particular significance in this regard is the ventral hippocampus.
One pathological finding that has been reported with some consistency is a
decrease in ventral hippocampal volume in schizophrenia patients, particularly
when comparing afflicted versus normal monozygotic twins (Suddath et al., 1990).
A focus on the ventral hippocampus is also supported by prospective studies, in
which an increase in hippocampal volume is observed in high-risk individuals
prior to the onset of psychosis (Pantelis et al., 2003; Phillips et al., 2002). Such
a condition suggests that the ventral hippocampus may undergo some type of
pathological alteration during the first episode that would lead to the expression
of psychosis. A potential etiological factor involving developmental disruption of
the ventral hippocampus or limbic cortex has been exploited in the advancement
of animal models of this disorder.
HPA axis in response to glucocorticoids (Sapolsky et. al. 1990), which are released
in response to stress (Cullinan et al., 1995). Indeed, in an animal model of this
disorder, Lipska et al. (1993) have shown that rats with neonatal damage to the hip-
pocampus exhibit hyperresponsiveness to stress. This interaction between stress
and hippocampal damage has been proposed as a model for delayed onset of disor-
ders such as schizophrenia (Walker and Diforio, 1997). In addition, a pathology of
prefrontal cortex function may also place increased demand on the hippocampus.
It has been suggested that some of the higher-order functions that are mediated by
specific cortical regions in the adult, such as the prefrontal cortex, may be mediated
by subcortical regions during developmental periods prior to the maturation of
the cortex under question (Goldman and Rosvold, 1972). Specifically, it has been
proposed that, early in life, the hippocampus may subsume some of the work-
ing memory functions of the individual while the prefrontal cortex is developing
during early life (Diamond, 1990), with this responsibility being off-loaded to the
prefrontal cortex as it reaches functional maturity. One consequence of a prefrontal
pathology would be that this region may not be capable of taking over such func-
tions from the hippocampus. Indeed, this is supported by data showing that some
working memory functions in the schizophrenia patient are mediated by the hip-
pocampus (Meyer-Lindenberg et al., 2001; Weinberger et al., 1993). Therefore, at
a time when stress may be taking an increasing toll on the hippocampus, it will
also be placed under abnormally high functional demand because of the increased
working memory load placed upon it. Indeed, the increased hippocampal volume
prior to the first episode of psychotic symptoms (Pantelis et al., 2003; Phillips et al.,
2002) may be an indicator of such a pathological condition.
The result is that maturation-related stressors could trigger a cascade whereby
prefrontal dysfunction interrupts coping mechanisms for stress at the level of the
amygdala, which, via projections from the central nucleus of the amygdala to the
hypothalamus, could augment stress hormone release. The hyperactivated sub-
cortical DA system could serve to overwhelm the system further with a flooding
of sensory input, thus increasing the stress-induced responses of the system. As
reviewed above, the glucocorticoids released in response to stress are known to
damage the hippocampus (Sapolsky et al., 1985), and it is known that the hip-
pocampus is involved in modulating the glucocorticoid response to stress (Hoschl
and Hajek, 2001; McEwen, 1998; Sapolsky, 1992; Sapolsky et al., 1990). There-
fore, the multiple systems that contribute to an augmented stress response and
the stress-induced release of glucocorticoids, when combined with increased func-
tional demand placed on the hippocampus, could lead to hippocampal damage,
which would reduce the hippocampal modulation of the stress response, thus lead-
ing to further heightened response to stress and more hippocampal damage in a
positive feedback system. Such an interaction could explain the delayed onset of
281 Developmental dysregulation of the dopamine system
Fig. 15.1. A model of the developmental pathophysiology of schizophrenia. This model consists of
a series of interconnected loops, with the common endpoint being a disruption in the
regulation of phasic dopamine (DA) release. One feature of this model is that the positive
feedback loop could be initiated from a number of starting points. For example, a perinatal
deficit in DA in the prefrontal cortex (PFC) is proposed to result in (i) disruption of tonic
DA regulation in the nucleus accumbens; (ii) removal of regulation of affective responses
within the basolateral amygdala; and (iii) a failure to offload working memory demand
from the hippocampus during maturation. This would lead to abnormally potentiated stress
responses, which would drive hippocampal pathology and DA imbalance further. Similarly,
an initiation within the hippocampal system would also be expected to exacerbate the
stress response, ultimately leading to disruption of PFC regulation and subcortical DA system
responsivity. Note that each of these feedback pathways loops through the stress response.
Thus, one potential means to alter the course of this pathological process would be to treat
the exacerbated stress response prior to the initiation of the damaging positive feedback
cascade that is proposed to lead to irreversible system damage and psychosis.
this pathology (Fig. 15.1). This is likely to have important implications for how it
ultimately affects DA system function.
Fig. 15.2. The dopamine (DA) system is under a complex set of regulatory influences. (A) In response
to a behaviorally salient stimulus, the DA neuron emits bursts of action potentials (1), leading
to a massive release of DA (in micro- to millimolar concentrations) into the synaptic cleft,
where it can stimulate postsynaptic receptors (2). The DA that is released is then rapidly taken
back up into the terminal by the dopamine transporter (DAT; 3) before it can escape the
synaptic cleft. The DAT achieves this because it is located at the periphery of the synaptic cleft
285 Developmental dysregulation of the dopamine system
Conclusions
I have presented a model whereby stress is proposed to interact with an existing
pathology within the limbic system to cause the delayed emergence of the posi-
tive symptoms of schizophrenia. The involvement of stress is demonstrated by the
finding that, of the children at risk for developing schizophrenia, those that do go
on to show the pathology exhibit higher levels of anxiety and stress premorbidly
(E. C. Johnstone, personal communication). Moreover, stress is known to exacer-
bate the symptoms of schizophrenia and lead to a relapse in patients under remis-
sion (Birley and Brown, 1970; Norman and Malla, 1993a,b). One implication of
this model is that, given that the DA system does not appear to be abnormal prior
to the first episode of psychotic symptoms, prophylactic treatment with antipsy-
chotic drugs may not be an effective course of action for preventing the ultimate
transition to schizophrenia. In contrast, drugs that act to reduce indices of stress or
anxiety may be more effective at preventing the triggering of the cascade of events
that eventually lead to the development of schizophrenia.
Acknowledgements
This work was supported by USPHS MH01055, MH57440, and MH45156. I thank
Stan Floresco for his help in producing the figures used in this manuscript.
(Nirenberg et al., 1997), where it can effectively limit diffusion of released transmitter.
(B) In contrast, slow, irregular firing of DA neurons (1) leads to substantially lower lev-
els of DA release into the synaptic cleft; this DA release is not as potently affected by
the DAT (2) but is modulated by glutamatergic (glu) afferents that synapse in the vicin-
ity of the DA terminal (3). As a result, a small amount of DA will escape the synaptic cleft
(4) or be released from non-synaptic sites on the terminal (5). This low (approximately
20 nM) concentration of tonic DA (6) is then free to diffuse away from the synaptic terminal.
(C) Although the levels of tonic DA (1) are too low to stimulate postsynaptic receptors, this
concentration is sufficient to activate D2 autoreceptors on the DA terminal, which would
cause an attenuation of burst firing-dependent phasic DA release (2). As a result, there is a
tonic inhibition in the amplitude of the phasic DA release (3). Systems that will attenuate
tonic DA release, either decreasing tonic DA neuron activity (i.e. less DA neurons active) or a
pathological decrease in cortical glutamatergic drive, would thereby remove tonic inhibition
of phasic DA release (Grace, 1991).
286 A. A. Grace
REFERENCES
Chang, M. S., Sved, A. F., Zigmond, M. J., Austin, M. C. (2000). Increased transcription of the
tyrosine hydroxylase gene in individual locus coeruleus neurons following footshock stress.
Neuroscience 101: 131–139.
Chrobak, J. J., Napier, T. C. (1993). Opioid and GABA modulation of accumbens-evoked ventral
pallidal activity. J Neur Transm Gen Sect 93: 123–143.
Cohen, J. D., Servan-Schreiber, D. (1992). Context, cortex and dopamine: a connectionist
approach to behavior and biology in schizophrenia. Psychol Rev 99: 45–77.
Correll, C. J., Rosenkranz, J. A., Grace, A. A. (2001). Chronic cold stress alters basal neuronal firing
rate and response to acute stressors of basolateral amygdala neurons in rats. Soc Neurosci
Abstr 27: 2540.
Creese, I., Burt, D. R., Snyder, S. H. (1976). Dopamine receptor binding predicts clinical and
pharmacological potencies of antischizophrenic drugs. Science 192: 596–598.
Cullinan, W. E., Herman, J. P., Helmreich, D. L., Watson, Jr., S. J. (1995). A neuroanatomy
of stress. In Neurobiological and Clinical Consequences of Stress: From Normal Adaptation
to PTSD, ed. M. J. Friedman, D. S. Charney, A. Y. Deutch. Philadelphia, PA: Lippincott,
pp. 3–25.
Curtis, A. L., Lechner, S. M., Pavcovich, L. A., Valentino, R. J. (1997). Activation of the locus
coeruleus noradrenergic system by intracoerulear microinfusion of corticotropin-releasing
factor: effects on discharge rate, cortical norepinephrine levels and cortical electroencephalo-
graphic activity. J Pharmacol Exp Ther 281: 163–172.
DeLisi, L. E. (1992). The significance of age of onset for schizophrenia. Schizophr Bull 18: 209–
215.
Diamond, A. (1990). The development and neural bases of memory functions as indexed by the
AB and delayed response tasks in human infants and infant monkeys. Ann NY Acad Sci 608:
267–309. [Discussion 309–317.]
Drevets, W. C. (1999). Prefrontal cortical–amygdalar metabolism in major depression. Ann N Y
Acad Sci 877: 614–637.
Dunn, A. J., Berridge, C. W. (1987). Corticotropin-releasing factor administration elicits a stress-
like activation of cerebral catecholaminergic systems.Pharmacol Biochem Behav 27: 685–691.
Eastwood, S. L., Cairns, N. J., Harrison, P. J. (2000). Synaptophysin gene expression in schizophre-
nia. Investigation of synaptic pathology in the cerebral cortex. Br J Psychiatry 176: 236–
242.
Edwards, H. (1970). The significance of brain damage in persistent oral dyskinesia. Br J Psychiatry
116: 271–275.
Emoto, H., Koga, C., Ishii, H., et al. (1993). A CRF antagonist attenuates stress-induced increases
in NA turnover in extended brain regions in rats. Brain Res 627: 171–176.
Ennis, M., Shipley, M. T., Aston-Jones, G., Williams, J. T. (1998). Afferent control of nucleus locus
ceruleus: differential regulation by “shell” and “core” inputs. Adv Pharmacol 42: 767–771.
Finlay J. M., Jedema H. P., Rabinovic, A. D., et al. (1997). Impact of corticotropin-releasing
hormone on extracellular norepinephrine in prefrontal cortex after chronic cold stress. J
Neurochem 69: 144–150.
Floresco, S. B., Todd, C. L., Grace, A. A. (2001). Glutamatergic afferents from the hippocampus
to the nucleus accumbens regulate activity of ventral tegmental area dopamine neurons.
J Neurosci 21: 4915–4922.
288 A. A. Grace
Floresco, S. B., West, A. R., Ash, B., Moore, H., Grace, A. A. (2003). Afferent modulation of
dopamine neuron firing differentially regulates tonic and phasic dopamine transmission.
Nat Neurosci 6: 968–973.
Garey, L. J., Ong, W. Y., Patel, T. S., et al. (1998). Reduced dendritic spine density on cerebral
cortical pyramidal neurons in schizophrenia. J Neurol Neurosurg Psychiatry 65: 446–453.
Ghajarnia, M., Moore, H., Grace, A. A. (1998). Enhanced behavioral effects of phencyclidine
(PCP) in rats with developmental abnormalities of the temporal lobe. Soc Neurosci Abstr 24:
2177.
Glantz, L. A., Lewis, D. A. (1997). Reduction of synaptophysin immunoreactivity in the pre-
frontal cortex of subjects with schizophrenia. Regional and diagnostic specificity. Arch Gen
Psychiatry 54: 943–952.
Goddard, A. W., Charney, D. S. (1997). Toward an integrated neurobiology of panic disorder. J
Clin Psychiatry 58(Suppl. 2): 4–11.
Goff, D. C., Coyle, J. T. (2001). The emerging role of glutamate in the pathophysiology and
treatment of schizophrenia. Am J Psychiatry 158: 1367–1377.
Goldman, P. S., Rosvold, H. E. (1972). The effects of selective caudate lesions in infant and juvenile
rhesus monkeys. Brain Res 43: 53–66.
Goldman-Rakic, P. S. (1995). Cellular basis of working memory. Neuron 14: 477–485.
(1996). Regional and cellular fractionation of working memory. Proc Natl Acad Sci USA 93:
13473–13480.
(1999). The physiological approach: functional architecture of working memory and disor-
dered cognition in schizophrenia. Biol Psychiatry 46: 650–661.
Grace, A. A. (1991). Phasic versus tonic dopamine release and the modulation of dopamine
system responsivity: a hypothesis for the etiology of schizophrenia. Neuroscience 41: 1–24.
(2000). Gating of information flow within the limbic system and the pathophysiology of
schizophrenia. [In Proceedings from the Nobel Symposium – Schizophrenia: Pathophysi-
ological Mechanisms.] Brain Res Rev 31: 330–361. Online at Brain Research Interactive:
http://www.elsevier.nl/gej-ng/29/19/30/26/43/article.html.
Grace, A. A., Moore, H. (1998). Regulation of information flow in the accumbens: a model for the
pathophysiology of schizophrenia. In Origins and Development of Schizophrenia: Advances
in Experimental Psychopathology, ed. M. F. Lenzenweger, R. H. Dworkin. Washington, DC:
American Psychological Association Press, pp. 123–157.
Grace, A. A., Bunney, B. S., Moore, H., Todd. C. L. (1997). Dopamine cell depolarization block
as a model for the therapeutic actions of antipsychotic drugs. Trends Neurosci 20: 31–37.
Gunne, L. M., Growdon, J., Glaeser, B. (1982). Oral dyskinesia in rats following brain lesions and
neuroleptic drug administration. Psychopharmacology 77: 134–139.
Harden, D. G., King, D., Finlay, J., Grace, A. A. (1998). Depletion of dopamine in the prefrontal
cortex decreases the basal electrophysiological activity of mesolimbic dopamine neurons.
Brain Res 794: 96–102.
Harrison, P. J. (1999). The neuropathology of schizophrenia. A critical review of the data and
their interpretation. Brain 122: 593–624.
Herman, J. P., Cullinan, W. E. (1997). Neurocircuitry of stress: central control of the hypothalamo–
pituitary–adrenocortical axis. Trends Neurosci 20: 78–84.
289 Developmental dysregulation of the dopamine system
Liddle, P. F., Friston, K. J., Frith, C. D., Frackowiak, R. S. (1992). Cerebral blood flow and mental
processes in schizophrenia. J Roy Soc Med 85: 224–227.
Lipska, B. K., Weinberger, D. R. (2000). To model a psychiatric disorder in animals: schizophrenia
as a reality test. Neuropsychopharmacology 23: 223–239.
Lipska, B. K., Jaskiw, G. E., Weinberger, D. (1993). Postpubertal emergence of hyperresponsiveness
to stress and to amphetamine after neonatal hippocampal damage: a potential animal model
of schizophrenia. Neuropsychopharmacology 9: 67–75.
Mana, M. J., Grace, A. A. (1997). Chronic cold stress alters the basal and evoked electrophysio-
logical activity of rat locus coeruleus neurons. Neuroscience 81: 1055–1064.
Mayer, M. L., Westbrook, G. L., Guthrie, P. B. (1984). Voltage-dependent block by Mg2+ of NMDA
responses in spinal cord neurones. Nature 309: 261–263.
McCarley, R. W., Wible, C. G., Frumin, M., et al. (1999). MRI anatomy of schizophrenia. Biol
Psychiatry 45: 1099–1119.
McDonald, C., Murray, R. M. (2000). Early and late environmental risk factors for schizophrenia.
Brain Res Rev 31: 130–137.
McEwen, B. S. (1998). Protective and damaging effects of stress mediators. N Engl J Med 338:
171–179.
McKernan, M. G., Shinnick-Gallagher, P. (1997). Fear conditioning induces a lasting potentiation
of synaptic currents in vitro. Nature 390: 607–611.
Meyer-Lindenberg, A., Poline, J. B., Kohn, P. D., et al. (2001). Evidence for abnormal cortical
functional connectivity during working memory in schizophrenia. Am J Psychiatry 158:
1809–1817.
Mirnics, K., Middleton, F. A., Marquez. A., Lewis, D. A., Levitt, P. (2000). Molecular characteriza-
tion of schizophrenia viewed by microarray analysis of gene expression in prefrontal cortex.
Neuron 28: 53–67.
Moore, H., Grace, A. A. (1997). Anatomical changes in limbic structures produced by methy-
lazoxymethanol acetate (MAM) during brain development are associated with changes in
physiological interactions among afferents to the nucleus accumbens. Soc Neurosci Abstr 23:
2378.
Moore, H., Ghajarnia, M., Geyer, M., Jentsch, J. D., Grace, A. A. (2001a). Selective disruption of
prefrontal and limbic corticostriatal circuits by prenatal exposure to the DNA methylation
agent methylazoxymethanol acetate (MAM): anatomical, neurophysiological and behavioral
studies. Schizophr Res 49(Suppl. 1–2): 48.
Moore, H., Rose, H. J., Grace, A. A. (2001b). Chronic cold stress reduces the spontaneous
activity of ventral tegmental dopamine neurons. Neuropsychopharmacology 24: 410–
419.
Nagata, Y., Matsumoto, H. (1969). Studies on methylazoxymethanol: methylation of nucleic acids
in the fetal rat brain. Proc Soc Exp Biol Med 132: 383–385.
Ninan, P. T. (1999). The functional anatomy, neurochemistry, and pharmacology of anxiety.
J Clin Psychiatry 60(Suppl. 22): 12–17.
Nirenberg, M. J., Chan, J., Pohorille, A., et al. (1997). The dopamine transporter: comparative
ultrastructure of dopaminergic axons in limbic and motor compartments of the nucleus
accumbens. J Neurosci 17: 6899–6907.
291 Developmental dysregulation of the dopamine system
Norman, R. M., Malla, A. K. (1993a). Stressful life events and schizophrenia: I. A review of the
research. Br J Psychiatry 162: 161–166.
(1993b). Stressful life events and schizophrenia: II. Conceptual and methodological issues.
Br J Psychiatry 162: 166–174.
O’Donnell, P., Grace, A. A. (1995). Synaptic interactions among excitatory afferents to nucleus
accumbens neurons: hippocampal gating of prefrontal cortical input. J Neurosci 15: 3622–
3639.
Olney, J. W., Farber, N. B. (1995). NMDA antagonists as neurotherapeutic drugs, psychotogens,
neurotoxins, and research tools for studying schizophrenia. Neuropsychopharmacology 13:
335–345.
Ono, T., Nishijo, H., Uwano, T. (1995). Amygdala role in conditioned associative learning. Prog
Neurobiol 46: 401–423.
Page, M. E., Abercrombie, E. D. (1999). Discrete local application of corticotropin-releasing factor
increases locus coeruleus discharge and extracellular norepinephrine in rat hippocampus.
Synapse 33: 304–313.
Pantelis, C., Velakoulis, D., McGorry, P. D., et al. (2003). Neuroanatomical abnormalities before
and after onset of psychosis: a cross-sectional and longitudinal MRI comparison. Lancet
361: 281–288.
Pare, D., Collins, D. R. (2000). Neuronal correlates of fear in the lateral amygdala: multiple
extracellular recordings in conscious rats. J Neurosci 20: 2701–2710.
Phillips, L. J., Velakoulis, D., Pantelis, C., et al. (2002). Non-reduction in hippocampal volume is
associated with higher risk of psychosis. Schizophr Res 58: 145–158.
Pilowsky, L. S., Kerwin, R. W., Murray, R. M. (1993). Schizophrenia: a neurodevelopmental
perspective. Neuropsychopharmacology 9: 83–91.
Pitkanen, A., Savander, V., LeDoux, J. E. (1997). Organization of intra-amygdaloid circuitries
in the rat: an emerging framework for understanding functions of the amygdala. Trends
Neurosci 20: 517–523.
Post, R. M., Fink, E., Carpenter, W. T., Goodwin, F. K. (1975). Cerebrospinal fluid amine metabo-
lites in acute schizophrenia. Arch Gen Psychiatry 32: 1063–1069.
Ramsooksingh M. D., Jedema, H. P., Moore, H., Sved, A. F., Grace, A. A. (2001). The effects
of chronic stress on the regulation of locus coeruleus neurons by peripheral, limbic, and
prefrontal cortical inputs. Soc Neurosci Abstr 27: 1961.
Ramsooksingh, M. D., Jedema, H. P., Moore, H., Sved, A. F., Grace, A. A. (2002). Chronic cold
stress leads to persistent changes in the behavioral response to footshock with coincident
changes in evoked spike activity in the locus coeruleus. Soc Neurosci Abstr 28: [Program No.]
669.4.
Richfield, E. K., Penney, J. B., Young, A. B. (1989). Anatomical and affinity state comparisons
between dopamine D1 and D2 receptors in the rat central nervous system. Neuroscience 30:
767–777.
Rogan, M. T., Stauble, U. V., LeDoux, J. E. (1997). Fear conditioning induces associative long-term
potentiation in the amygdala. Nature 390: 604–607.
Rosenkranz, J. A., Grace, A. A. (1999). Modulation of basolateral amygdala neuronal firing and
afferent drive by dopamine receptor activation in vivo. J Neurosci 19: 11027–11039.
292 A. A. Grace
(2001). Dopamine attenuates prefrontal cortical suppression of sensory inputs to the basolat-
eral amygdala of rats. J Neurosci 21: 4090–4103.
(2002a). Cellular mechanisms of infralimbic and prelimbic prefrontal cortical inhibition and
dopaminergic modulation of basolateral amygdala neurons in vivo. J Neurosci 22: 324–
337.
(2002b). Dopamine-mediated modulation of odour-evoked amygdala potentials during Pavlo-
vian conditioning. Nature 417: 282–287.
Sabban, E. L., Kvetnansky, R. (2001). Stress-triggered activation of gene expression in cate-
cholaminergic systems: dynamics of transcriptional events. Trends Neurosci 24: 91–98.
Sapolsky, R. (1992). Stress, The Aging Brain, and the Mechanisms of Neuron Death. Cambridge,
MA: MIT Press.
Sapolsky, R., Krey, L., McEwen, B. (1985). Prolonged glucocorticoid exposure reduces hippocam-
pal neural number: Implications for aging. J Neurosci 5: 1221–1224.
Sapolsky, R. M., Armanini, M. P., Packan, D. R., Sutton, W. S., Plotsky, P. M. (1990). Glucocorticoid
feedback inhibition of adrenocorticotropic hormone secretagogue release. Neuroendocrinol-
ogy 51: 328–336.
Saunders R. C., Kolachana, B. S., Bachevalier, J., Weinberger, D. R. (1998). Neonatal lesions of
the medial temporal lobe disrupt prefrontal cortical regulation of striatal dopamine. Nature
393: 169–171.
Seeman, P. (1987). Dopamine receptors and the dopamine hypothesis of schizophrenia. Synapse
1: 133–152.
Schultz, W. (1998). Predictive reward signal of dopamine neurons. J Neurophysiol 80: 1–27.
Selemon, L. D., Rajkowska, G., Goldman-Rakic, P. S. (1995). Abnormally high neuronal density
in the schizophrenic cortex. A morphometric analysis of prefrontal area 9 and occipital area
17. Arch Gen Psychiatry 52: 805–818.
Serova, L. I., Nankova, B. B., Feng, Z., et al. (1999). Heightened transcription for enzymes
involved in norepinephrine biosynthesis in the rat locus coeruleus by immobilization stress.
Biol Psychiatry 45: 853–862.
Shanks, N., Zalcman, S., Zacharko, R. M., Anisman, H. (1991). Alterations of central nore-
pinephrine, dopamine and serotonin in several strains of mice following acute stressor
exposure. Pharmacol Biochem Behav 38: 69–75.
Sherman, J. E., Kalin, N. H. (1986). ICV–CRH potently affects behavior without altering antinoci-
ceptive responding. Life Sci 39: 433–441.
Smagin, G. N., Swiergiel. A. H., Dunn, A. J. (1995). Corticotropin-releasing factor administered
into the locus coeruleus, but not the parabrachial nucleus, stimulates norepinephrine release
in the prefrontal cortex. Brain Res Bull 36: 71–76.
Smagin, G. N., Harris, R. B., Ryan, D. H. (1996). Corticotropin-releasing factor receptor antag-
onist infused into the locus coeruleus attenuates immobilization stress-induced defensive
withdrawal in rats. Neurosci Lett 220: 167–170.
Smagin, G. N., Zhou, J., Harris, R. B., Ryan, D. H. (1997). CRF receptor antagonist attenuates
immobilization stress-induced norepinephrine release in the prefrontal cortex in rats. Brain
Res Bull 42: 431–434.
293 Developmental dysregulation of the dopamine system
Suddath, R. L., Christison, G. W., Torrey, E. F., Casanova, M. F., Weinberger, D. R. (1990).
Anatomical abnormalities in the brains of monozygotic twins discordant for schizophrenia.
N Engl J Med 322: 789–794.
Susser, E., Neugebauer, R., Hoek, H. W., et al. (1996). Schizophrenia after prenatal famine. Arch
Gen Psychiatry 53: 25–31.
Tamminga, C. A., Thaker, G. K., Buchanan, R., et al. (1992). Limbic system abnormalities iden-
tified in schizophrenia using positron emission tomography with fluorodeoxyglucose and
neocortical alterations with deficit syndrome. Arch Gen Psychiatry 49: 522–530.
Tebartz van Elst, L., Woermann, F. G., Lemieux, L., Trimble, M. R. (1999). Amygdala enlargement
in dysthymia: a volumetric study of patients with temporal lobe epilepsy. Biol Psychiatry 46:
1614–1623.
Thierry, A. M., Javoy. F., Glowinski, J., Kety, S. S. (1968). Effects of stress on the metabolism
of norepinephrine, dopamine and serotonin in the central nervous system of the rat. I.
Modifications of norepinephrine turnover. J Pharmacol Exp Ther 163: 163–171.
Thune, J. J., Pakkenberg, B. (2000). Stereological studies of the schizophrenic brain. Brain Res
Rev 31: 200–204.
Torrey, E. F., Miller, J., Rawlings, R., Yolken R. H. (1997). Seasonality of births in schizophrenia
and bipolar disorder: a review of the literature. Schizophr Res 28: 1–38.
Vale, W., Spiess, J., Rivier, C., Rivier, J. (1981). Characterization of a 41-residue ovine hypothalamic
peptide that stimulates secretion of corticotropin and beta-endorphin. Science 213: 1394–
1397.
Valentino, R. J., Foote, S. L. (1986). Brain noradrenergic neurons, corticotropin-releasing factor,
and stress. In: Neural and Endocrine Peptides and Receptors, ed. T. W. Moody. New York:
Plenum Press, pp. 101–120.
Valentino, R. J., Wehby, R. G. (1988). Corticotropin-releasing factor: evidence for a neurotrans-
mitter role in the locus ceruleus during hemodynamic stress. Neuroendocrinology 48: 674–
677.
Valentino, R. J., Foote S. L., Aston-Jones G (1983). Corticotropin-releasing factor activates nora-
drenergic neurons of the locus coeruleus. Brain Res 270: 363–367.
Valentino, R. J., Page, M. E., Curtis, A. L. (1991). Activation of noradrenergic locus coeruleus
neurons by hemodynamic stress is due to local release of corticotropin-releasing factor.
Brain Res 555: 25–34.
Valentino, R. J., Foote, S. L., Page, M. E. (1993). The locus coeruleus as a site for integrating
corticotropin-releasing factor and noradrenergic mediation of stress responses. Ann NY
Acad Sci 697: 173–188.
Valentino, R. J., Chen, S., Zhu, Y., Aston-Jones, G. (1996). Evidence for divergent projections to
the brain noradrenergic system and the spinal parasympathetic system from Barrington’s
nucleus. Brain Res 732: 1–15.
Van Bockstaele, E. J., Colago, E. E., Valentino, R. J. (1996). Corticotropin-releasing factor-
containing axon terminals synapse onto catecholamine dendrites and may presynaptically
modulate other afferents in the rostral pole of the nucleus locus coeruleus in the rat brain.
J Comp Neurol 364: 523–534.
294 A. A. Grace
Since the early 1980s, neuroscience studies have begun to define the pathophys-
iology of schizophrenia and bipolar disorder by using a combination of highly
selective molecular probes and quantitative microscopy. It is now possible to define
how neural circuits have been altered in psychosis in terms of an evolving set of
neurobiological principles (Benes, 2000). In patients with schizophrenia, and more
recently bipolar disorder, a variety of anomalies have been detected in the limbic
lobe, a phylogenetically old portion of the cerebral cortex that includes the anterior
cingulate region and hippocampal formation (Fig. 16.1). In addition, the basolat-
eral subdivision of the amygdala (not shown), another component of the limbic
lobe, has been implicated in the pathophysiology of psychotic disorders, in part
because it sends important inputs to the cingulate cortex and hippocampus. It now
seems likely that the three major components of the limbic lobe play a central role
in generating disturbances in motivation, attention, emotion, and social interac-
tions in schizophrenia and bipolar disorder (Benes, 1993). The discussion below
describes studies in both human and rodent brains that have contributed to our
understanding of how this complex circuitry may be altered during development,
leading to the appearance of psychotic disorders during late adolescence and early
adulthood. A clear understanding of how the brain is normally changing during
the postnatal period is an essential ingredient in building plausible models for what
may be happening during the prodrome of psychotic disorders.
295
296 F. M. Benes
Neocortex
Fig. 16.1. The limbic lobe is shown on the midsagittal surface of the brains of a rat, cat, and monkey.
There is a remarkable degree of preservation of this structure across mammalian species. In
contrast, the surrounding neocortex show a marked expansion in primates when compared
with that in rodents. (Adapted from McLean, 1954.)
CONTROL PSYCHOSIS
DA DA
Pyramidal Cell
Fig. 16.2. A schematic diagram depicting a “mis-wired” circuit within layer II of the anterior cingulate
cortex of a normal control (left) and psychotic (right) subject. A pyramidal neuron in the
psychotic circuit is receiving a reduced inhibition from both dopaminergic (DA) and gamma-
aminobutyric acid (GABA)-ergic inputs, while the GABA cell is receiving increased inputs from
the mutually antagonistic amygdalar (excitatory) and dopaminergic (inhibitory) fibers. The
release of DA might prevent the GABA cell from responding to increased excitatory inflow
from the amygdala. Overall, the circuit associated with psychotic disorders has too much
excitation, too little inhibition, and would result in abnormal information processing and
possibly apoptotic changes. Antipsychotic drugs would ameliorate the effect of stress by
blocking DA receptors on GABA cells and allowing the GABA cells to inhibit the firing of
pyramidal neurons.
Fig. 16.3. The distribution of gamma-aminobutyric acid (GABA)-ergic cells in the medial prefrontal
cortex of rat brain during the postnatal period. Between day 1 (P1) and day 10 (P10),
the density of cells is much greater than that seen at days 20 to 41 (P20 to P41), the
postweanling period that is considered to be the equivalent in rats of human adolescence.
Although the overall number of cells at day 41 is identical to that found at day 5, the packing
density decreases dramatically as the cortical mantle increases its thickness. The GABAergic
cells are surrounded by neuropil, where axons and dendrites occupy most of the space.
increase in numerical density until postnatal day II (Chronwall and Wolff, 1980).
In contrast, the concentration of GABA and the specific activity of GAD (Coyle and
Enna, 1976), as well as the binding activity of the GABA receptor (Coyle and Enna,
1976; Palacios et al., 1979) and its mRNA (Gambarana et al., 1990), all increase
until the third postnatal week.
As shown in Fig. 16.3, the numerical density of GABA-immunoreactive neurons
in the anterior cingulate cortex of the rat reaches a peak at approximately postnatal
day 5 and then diminishes until day 20, when the thickness of the cortical mantle
is maximal (Vincent et al., 1995). During this same period, the relative amount
of neuropil surrounding all cell bodies in this region is expanding as dendritic
and axonal fibers are increasing. GABAergic cell bodies not only show a gradual
increase in their size, but by postnatal days 20 to 40, primary, secondary, and tertiary
branches of their dendritic tree can also be visualized. The expansion of neuropil in
rat medial prefrontal cortex probably involves an increase of both dendritic branches
301 “Mis-wired” limbic lobe circuitry
A B C
GABA-Immunoreactive Neurons
Dopamine-Immunoreactive Fibers
II
III
VI
P6 P16 P26 P36 P45 Adult
B
2000
r = 0.873
1500
1000
Fiber Density
500
0
0 50 100 150
Postnatal Day
Fig. 16.5. Amygdala projections to the anterior cingulate cortex after birth in rats. (A) Amygdalar fibers
visualized with anterograde tracing show a marked increase in layers II and V during the
postnatal period. (B) The increase in fiber density shows a highly significant curvilinear rise
between birth (postnatal day 0) and adulthood (postnatal day 120) in layer II. (See plate
section for color version.)
303 “Mis-wired” limbic lobe circuitry
Fig. 16.6. A set of schematic diagrams depicting the normal development of an intrinsic circuit in
the anterior cingulate cortex during early and late adolescence. Gamma-aminobutyric acid
(GABA) cell development is probably relatively complete by early adolescence while that
of dopamine fibers and amygdalar projections continues into late adolescence and early
adulthood. By late adolescence, the neural response to emotionally stressful situations may
be relatively complete in normal individuals. However, individuals exposed to stress early in
life may be sensitized to the effects of stress later. During late adolescence, the stressed circuit
could theoretically develop excessive dopaminergic and amygdalar inputs. Such changes
may be associated with a decompensation of the circuit if GABAergic cells are unable to
provide sufficient inhibitory modulation.
of the cingulate cortex, it will be relevant to know whether these two systems may
actually interact with one another through a presynaptic mechanism.
Acknowledgements
This work has been supported by grants from the National Institutes of Health
(MH00423, MH31862, MH31154), the Stanley Foundation, the Carmella and
Menachem Abraham Fund and the Taplin Fund.
REFERENCES
Benes, F. M., McSparren, J., Bird, E. D., SanGiovanni, J. P., Vincent, S. L. (1991). Deficits in
small interneurons in prefrontal and cingulate cortices of schizophrenic and schizoaffective
patients. Arch Gen Psychiatry 48: 996–1001.
Benes, F. M., Vincent, S. L., Alsterberg, G., Bird, E. D., SanGiovanni, J. P. (1992a). Increased GABAA
receptor binding in superficial layers of cingulate cortex in schizophrenics. J Neurosci 12:
924–929.
Benes, F. M., Sorensen, I., Vincent, S. L., Bird, E. D., Sathi, M. (1992b). Increased density of
glutamate-immunoreactive vertical processes in superficial laminae in cingulate cortex of
schizophrenic brain. Cereb Cortex 2: 503–512.
Benes, F. M., Vincent, S. L., Molloy, R. (1993). Dopamine-immunoreactive axon varicosities form
nonrandom contacts with GABA-immunoreactive neurons of rat medial prefrontal cortex.
Synapse 15: 285–295.
Benes, F. M., Khan, Y., Vincent, S. L., Wickramasinghe, R. (1996a). Differences in the subregional
and cellular distribution of GABAA receptor binding in the hippocampal formation of
schizophrenic brain. Synapse 22: 338–349.
Benes, F. M., Vincent, S. L., Molloy, R., Khan, Y. (1996b). Increased interaction of dopamine-
immunoreactive varicosities with GABA neurons of rat medial prefrontal cortex occurs
during the postweanling period. Synapse 23: 237–245.
Benes, F. M., Todtenkopf, M. S., Taylor, J. B. (1997). Differential distribution of tyrosine hydroxy-
lase fibers on small and large neurons in layer II of anterior cingulate cortex of schizophrenic
brain. Synapse 25: 80–92.
Benes, F. M., Kwok, E. W., Vincent, S. L., Todtenkopf, M. S. (1998). A reduction of nonpyramidal
cells in sector CA2 of schizophrenics and manic depressives. [See comments] Biol Psychiatry
44: 88–97.
Benes, F. M., Todtenkopf, M. S., Logiotatos, P., Williams, M. (2000). Glutamate decarboxylase(65)-
immunoreactive terminals in cingulate and prefrontal cortices of schizophrenic and bipolar
brain. J Chem Neuroanat 20: 259–269.
Benes, F. M., Vincent, S. L., Todtenkopf, M. S. (2001). The density of pyramidal and nonpyramidal
neurons in anterior cingulate cortex of schizophrenic and bipolar subjects. Biol Psychiatry
50: 395–406.
Berretta, S., Munno, D. W., Benes, F. M. (2001). Amygdalar activation alters the hippocampal
GABA system: “partial” modelling for postmortem changes in schizophrenia. J Comp Neurol
431: 129–138.
Brozoski, T., Brown, R. M., Rosvold, H. E., Goldman, P. S. (1979). Cognitive deficit
caused by depletion of dopamine in prefrontal cortex of rhesus monkey. Science 205:
929–931.
Bruinink, A., Lichtensteiner, W., Schlumpf, M. (1983). Pre- and postnatal ontogeny and char-
acterization of dopaminergic D2, serotonergic S2, and spirodecanone binding sites in rat
forebrain. J Neurochem 40: 1227–1237.
Chronwall, B., Wolff, J. R. (1980). Prenatal and postnatal development of GABA-accumulating
cells in the occipital neocortex of rat. J Comp Neurol 190: 187–208.
Coyle, J. T., Enna, S. (1976). Neurochemical aspects of the ontogenesis of GABAnergic neurons
in the rat brain. Brain Res 111: 119–133.
307 “Mis-wired” limbic lobe circuitry
Coyle, J. T., Puttfarcken, P. (1993). Oxidative stress, glutamate, and neurodegenerative disorders.
[Review] Science 262: 689–695.
Cunningham, M. G., Bhattacharyya, S., Benes, F. M. (2002). Amygdalo-cortical sprouting con-
tinues into early adulthood: Implications for the development of normal and abnormal
function during adolescence. J Comp Neurol 453: 116–130.
Daviss, S. R., Lewis, D. A. (1995). Local circuit neurons of the prefrontal cortex in schizophrenia:
selective increase in the density of calbindin-immunoreactive neurons. Psychiatry Res 59:
81–96.
Deskin, R., Seidler, F. J., Whitmore, W. L., Slotkin, T. A. (1981). Development of α-noradrenergic
and dopaminergic receptor systems depends on maturation of their presynaptic nerve
terminals in the rat brain. J Neurochem 36: 1683–1690.
Fatemi, S. H., Emamian, E. S., Kist, D., et al. (1999). Defective corticogenesis and reduction in
Reelin immunoreactivity in cortex and hippocampus of prenatally infected neonatal mice.
Mol Psychiatry 4: 145–154.
Gambarana, C., Pittman, R., Siegel, R. E. (1990). Development expression of the GABA-A receptor
γ 1 subunit mRNA in the rat brain. J Neurobiol 21: 1169–1179.
Guidotti, A., Auta, J., Davis, J. M., et al. (2000). Decrease in Reelin and glutamate acid
decarboxylase67 (GAD67 ) expression in schizophrenia and bipolar disorder. Arch Gen
Psychiatry 57: 1061–1069.
Heckers, S., Stone, D., Walsh, J., et al. (2002). Differential hippocampal expression of glutamic
acid decarboxylase 65 and 67 messenger RNA in bipolar disorder and schizophrenia. Arch
Gen Psychiatry 59: 521–529.
Impagnatiello, F., Guidotti, A. R., Pesold, C., et al. (1998). A decrease of reelin expression as a
putative vulnerability factor in schizophrenia. Proc Natl Acad Sci USA 95: 15718–15723.
Iritani, S., Kuroki, N., Ikeda, K., Kazamatsuri, H. (1999). Calbindin immunoreactivity in the
hippocampal formation and neocortex of schizophrenics. Prog Neuropsychopharmacol Biol
Psychiatry 23: 409–421.
Johnston, M. V. (1988). Biochemistry of neurotransmitters in cortical development. In Cerebral
Cortex, ed. E. G. Jones. New York: Plenum Press, pp. 211–236.
Jones, E. G. (1993). GABAergic neurons and their role in cortical plasticity in primates. Cereb
Cortex 3: 361–372.
Kalsbeek, A., Voorn, P., Buijs, R. M., Pool, C. W., Uylings, H. B. (1988). Development of the
dopaminergic innervation in the prefrontal cortex of the rat. J Comp Neurol 269: 58–72.
Kalsbeek, A., Matthijssen, M. A., Uylings, H. B. (1989). Morphometric analysis of prefrontal corti-
cal development following neonatal lesioning of the dopaminergic mesocortical projection.
Exp Brain Res 78: 279–289.
Longson, D., Deakin, J. F., Benes, F. M. (1996). Increased density of entorhinal glutamate-
immunoreactive vertical fibers in schizophrenia. J Neur Transm 103: 503–507.
Luhmann, H. J., Prince, D. A. (1991). Postnatal maturation of the GABAergic system in rat
neocortex. J Neurophysiol 65: 247–263.
McDonald, A. J., Beitz, A. J., Larson, A. A., et al. (1989). Co-localization of glutamate and tubulin
in putative excitatory neurons of the hippocampus and amygdala: an immunohistochemical
study using monoclonal antibodies. Neuroscience 30: 405–421.
308 F. M. Benes
McLean, P. D. (1954). Studies on limbic system (visceral brain) and their bearing on psycho-
somatic problems. In Recent Developments in Psychosomatic Medicine, ed. E. Wittkower,
R. Cleghorn. Philadelphia, PA: Lippincott, p. 106.
Palacios, J. M., Niehoff, D. L., Kuhar, M. J. (1979). Ontogeny of GABA and benzodiazepine
receptors: effects of Triton X-100, bromide and muscimol. Brain Res 179: 390–395.
Penit-Soria, J., Retaux, S., Maurin, Y. (1989). Effets de la stimulation des recepteurs D1 et D2
dopaminergiques sur la liberation d’acide γ -[3 H] aminobutyrique induite electriquement
dans le cortex prefrontal du rat. C R Acad Sci Paris 309: 441–446.
Perez-Jananay, J. M., Vives, F. (1991). Electrophysiological study of the response of medical
prefrontal cortex neurons to stimulation of the basolateral nucleus of the amygdala in the
rat. Brain Res 564: 97–101.
Pierri, J. N., Chaudry, A. S., Woo, T. U., Lewis, D. A. (1999). Alterations in chandelier neuron
axon terminals in the prefrontal cortex of schizophrenic subjects. Am J Psychiatry 156:
1709–1719.
Pitkanen, A., Pikkarainen, M., Nurminen, N., Ylinen, A. (2000). Reciprocal connections between
the amygdala and the hippocampal formation, perirhinal cortex, and postrhinal cortex in
rat. A review. Ann N Y Acad Sci 911: 369–391.
Retaux, S., Besson, M. J., Penit-Soria, J. (1991). Opposing effects of dopamine D2 receptor
stimulation on the spontaneous and the electrically evoked release of [3 H] GABA on rat
prefrontal cortex slices. Neuroscience 42: 61–71.
Roth, R. H., Tam, S. Y., Ida, Y., Yang, J. X., Deutch, A. Y. (1988). Stress and the mesocorticolimbic
dopamine systems. Ann N Y Acad Sci 537: 138–147.
Selemon, L. D., Goldman, Rakic, P., Rajkowska, G. (2002). 2D versus 3D cell counting. Biol
Psychiatry 51: 838–840.
Todtenkopf, M. S., Benes, F. M. (1998). Distribution of glutamate decarboxylase65 immunore-
active puncta on pyramidal and nonpyramidal neurons in hippocampus of schizophrenic
brain. Synapse 29: 323–332.
Van Hoesen, G. W., Morecraft, R. J., Vogt, B. A. (1993). Connections of the monkey cingulate
cortex. In Neurobiology of Cingulate Cortex and Limbic Thalamus, ed. M. Gabriel. Boston,
MA: Birkhauser, pp. 249–284.
Verney, C., Berger, B., Adrien, J., Vigny, A., Gay, M. (1982). Development of the dopaminergic
innervation of the rat cerebral cortex. A light microscopic immunocytochemical study using
anti-tyrosine hydroxylase antibodies. Brain Res 281: 41–52.
Vincent, S. L., Adamec, E., Sorensen, I., Benes, F. M. (1994). The effects of chronic haloperidol
administration on GABA-immunoreactive axon terminals in rat medial prefrontal cortex.
Synapse 17: 26–35.
Vincent, S. L., Pabreza, L., Benes, F. M. (1995). Postnatal maturation of GABA-immunoreactive
neurons of rat medial prefrontal cortex. J Comp Neurol 355: 81–92.
Volk, D. W., Austin, M. C., Pierri, J. N., Sampson, A. R., Lewis, D. A. (2000). Decreased glu-
tamic acid decarboxylase67 messenger RNA expression in a subset of prefrontal cortical
gamma-aminobutyric acid neurons in subjects with schizophrenia. Arch Gen Psychiatry 57:
237–245.
309 “Mis-wired” limbic lobe circuitry
Schizophrenia is clearly a multithetic disorder in which the diverse signs and symp-
toms of the illness arise as a result of dysfunction in a number of brain regions.
Of these clinical features, a growing body of literature indicates that core features
of the illness are represented by disturbances in certain cognitive functions, such
as those mediated by neural circuits that include components of the dorsolateral
prefrontal cortex (DLPFC) and the mediodorsal nucleus (MDN) of the thalamus
(Elvevåg and Goldberg, 2000). The specific causal factors that give rise to these
brain abnormalities remain enigmatic, with a number of genes and a range of envi-
ronmental events identified as potential risk factors for schizophrenia (Lewis and
Lieberman, 2000). In addition, developmental processes appear to play a central
role in the translation of these etiological factors into the appearance of the clinical
features of the illness (Lewis and Levitt, 2002). Consequently, understanding the
structural and functional maturation of the intrinsic and extrinsic circuitry of the
DLPFC and MDN is essential for understanding the pathophysiological basis for
the cognitive deficits in schizophrenia.
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
310
311 Development of thalamocortical circuitry
of the DLPFC when attempting to perform tasks that tap working memory
(Weinberger et al., 1986).
The DLPFC maintains reciprocal excitatory connections with specific subdivi-
sions of the MDN, the principal source of thalamic inputs to the DLPFC (Barbas
et al., 1991; Giguere and Goldman-Rakic, 1988; Goldman-Rakic and Porrino, 1985).
Like the DLPFC, the MDN has been implicated as a site of dysfunction in schizo-
phrenia (Lewis, 2000), and it also appears to play a critical role in working memory
functioning. For example, destruction of some or all of the MDN produces deficits
in working memory performance (Isseroff et al., 1982). Furthermore, during the
delay period of delayed-response tasks, neurons in both the DLPFC (Funahashi
et al., 1989) and the MDN (Fuster and Alexander, 1971) exhibit sustained activity,
which, at least in the DLPFC, has been proposed to be the neural substrate for
working memory (Fuster, 1997; Goldman-Rakic, 1995).
The ability to perform delayed-response tasks first appears between 2 and
4 months of age in monkeys (Goldman-Rakic, 1987) and around 1 year of age in
humans (Diamond, 1985). Performance on these tasks then continues to improve
(as evidenced by the ability to withstand longer delay periods) at a slower rate until
adult functional competence is achieved after puberty in both monkeys (Alexander
and Goldman, 1978) and humans (Casey et al., 2000; Diamond, 2002). Although
the contributions of the MDN are less well studied, studies in humans clearly indi-
cate that changes in the frontal cortex between the ages of 12 and 19 years contribute
to the maturation of cognitive control (Bunge et al., 2002).
Studies in monkeys also suggest that DLPFC neural circuitry is increasingly
involved in the mediation of these improvements in cognitive performance during
postnatal development. That is, the emergence of the ability to perform delayed-
response tasks during infancy does not appear to depend upon the integrity of the
DLPFC but appears to be mediated by subcortical structures like the MDN. For
example, ablation of the DLPFC in infant monkeys does not produce the same
degree of impairment on spatial delayed-response tasks that is observed in adult
animals with such lesions (Alexander and Goldman, 1978). In contrast, during the
first year of life, performance of oculomotor delayed-response tasks do appear to
depend upon the integrity of the MDN (Alexander and Goldman, 1978). Further-
more, during postnatal development, the percentage of DLPFC neurons exhibiting
delay period activity doubles between 12 and 36 months of age (Alexander, 1982).
In contrast, the MDN does not exhibit this type of developmental increase in the
number of delay-activated neurons (Alexander, 1982). Together, these findings sug-
gest that postnatal development is associated with an increasing dependence upon
the DLPFC for the mediation of delayed-response behavior, such that the matu-
ration of the DLPFC enhances the function of brain regions like the MDN, which
appear to subserve these tasks earlier in life (Alexander and Goldman, 1978).
312 D. S. Melchitzky and D. A. Lewis
Fig. 17.1. Connections between the human prefrontal cortex and the mediodorsal nucleus (MDN) of
the thalamus. (A) Schematic view of the lateral surface of the human brain. (B) Coronal
section of the prefrontal cortex, taken at the level of vertical line 1 in (A). (C) Expanded
schematic drawing of the box in (A). Pyramidal neurons in layer 6 send projections to the
MDN as well as to the reticular nucleus of the thalamus (Rt). (D) Coronal section through
the thalamus, taken at the level of vertical line 2 in (A). The individual thalamic nuclei are
outlined. (E) Expanded view of the outline of the thalamus shown in (D). Neurons in the
MDN send axons to the prefrontal cortex that arborize primarily in deep layers 3 and 4
but also extend into layer 1 (C). MDN neurons also send axon collaterals to the inhibitory
neurons of the Rt. Large cortically projecting excitatory neurons in the MDN receive inhibitory
input from smaller GABA neurons located in the MDN and Rt.
313 Development of thalamocortical circuitry
Fig. 17.2. Axon terminals in the dorsolateral prefrontal cortex (DLPFC). (A) Electron micrograph of a
parvalbumin-immunoreactive thalamic axon terminal (PV-T) in layer 4 of the DLPFC forming
an asymmetric, excitatory synapse onto a dendritic spine(s). (B) Electron micrograph of a
biotinylated dextran amine-labeled intrinsic axon terminal (BDA-T) in DLPFC layer 3 forming
an asymmetric synapse onto a parvalbumin-containing dendritic shaft (PVd ). (Adapted from
Melchitzky et al., 1999 (A) and 2001(B).)
315 Development of thalamocortical circuitry
neurons of the reticular thalamic nucleus, which, in turn, provide inhibitory regu-
lation of MDN projection neurons (Fig. 17.1E).
Unfortunately, attempts to explore the functional properties of the circuitry
between the MDN and DLPFC face certain limitations. First, because they lack direct
connections with the periphery, it is very difficult to manipulate the inputs to or
intrinsic components of MDN–DLPFC circuitry; therefore, most of our knowledge
base involves inferences from sensory thalamocortical systems. For example, in con-
trast to the deprivation or manipulation of visual inputs that have been used to study
the functional properties of lateral geniculate thalamus and primary visual cortex
circuitry, such manipulations cannot be made to study the connections between
the MDN and the DLPFC. Second, the majority of studies investigating thalamo-
corticothalamic circuitry have been performed in non-primates, such as rodents
and cats. Given the apparent differences in DLPFC circuitry between rodents and
primates (Lewis, 2002), extrapolation of rodent data to primate systems can only
be made with a number of caveats.
Fig. 17.3. Neuron circuits in the dorsolateral prefrontal cortex (DLPFC). (A, B) Darkfield photomicro-
graphs of biotinylated dextran amine (BDA)-labeled axons resulting from an injection site in
layer 3 of monkey DLPFC. Horizontally oriented, long-range intrinsic axon collaterals of layer
3 pyramidal neurons travel through the gray matter (A: arrows) and form a stripe-like cluster
of labeled axons and terminals confined to layers 1–3 (A). The labeled associational axons
from the same injection site travel through the white matter (B: arrows) and form a dense
cluster that extends across all cortical layers (B). PS indicates principal sulcus. (Scale bar =
100 m.) (C) Schematic diagram illustrating the spatial distribution of the three types of axon
terminal furnished by layer 3 pyramidal neurons in monkey DLPFC. BDA-labeled axons of
pyramidal neurons terminate nearby (local) or after traveling horizontally through the gray
matter (intrinsic) or through the white matter (associational) to form stripes of labeled
terminals. (D) Tangential reconstruction of all stripe-like clusters of retrogradely labeled
neurons arising from a single injection in monkey DLPFC. On this unfolded map, the PS is
oriented as a vertical line, and all labeled structures and other sulci are shown relative to
the PS. The location of the injection site is indicated by asterisk. Intrinsic stripes are shaded
in black, and associational stripes are cross-hatched. Note that the associational projections
form several distinct groups of stripes located at varying distances from the intrinsic stripes.
ASs, superior ramus of arcuate sulcus; CS, cingulate sulcus. (Adapted from Melchitzky et al.,
1998 (A, B) and 2001 (C) and Pucak et al., 1996 (D).)
318 D. S. Melchitzky and D. A. Lewis
development of the connections between MDN and DLPFC has been extrapolated
from data from primary sensory regions in primates and rodents. In primates, the
growth of thalamic axons towards the cortical plate (a dense sheet of immature
neurons that, with differentiation, acquire the characteristic laminated appearance
of the mature cortex) occurs relatively early in the gestation period of 165 days for
macaque monkeys. For example, axons from the lateral geniculate nucleus begin to
approach the area of the primary visual cortex by embryonic day 78 (Rakic, 1977).
Although the thalamic axons begin their journey towards the developing cerebral
cortex early, they appear to “wait” for an extended time in the subplate (composed
of loosely packed neurons located below and born prior to those that form the
cortical plate) before invading the cortical plate. During this “waiting period,”
thalamic axons appear to form transient, synapse-like structures with subplate
neurons, and such contacts appear to be critical for the subsequent formation
of proper connections between thalamic axons and cortical neurons. For example,
following lesions of the subplate, axons from the lateral geniculate nucleus continue
to grow past the visual cortex, suggesting that subplate neurons are required for
these thalamic axons to recognize their appropriate cortical target (Ghosh et al.,
1990). After this “waiting period,” thalamic axons make a second growth spurt
to enter the cortical plate, such that, in the monkey, thalamic axons enter the
visual cortex by embryonic day 124. This foray of thalamic axons into the cortical
plate is somewhat diffuse. For example, in monkey visual cortex, the presence of
ocular dominance columns is not apparent initially and segregation of fibers into
these columns begins to appear 3 weeks before birth (Rakic, 1977). Therefore, the
formation of thalamocorticothalamic circuitry could be affected by adverse events
occurring over a broad period of prenatal development.
During postnatal development, substantial changes transpire in the develop-
mental trajectory for synaptic density in monkey DLPFC observed via electron
microscopy (Bourgeois et al., 1994). The number of asymmetric, excitatory synapses
on dendritic spines increases rapidly between birth and 2 months of age; it then
remains at this relatively high level until adolescence, when the number declines to
adult levels (Fig. 17.4A). A similar process of synaptic overproduction and elimi-
nation also occurs in human DLPFC (Huttenlocher, 1979). In addition, imaging
studies of the DLPFC in children and young adults demonstrate a similar pattern of
initial gray matter growth in childhood followed by gray matter reduction starting
in adolescence (Giedd, 1999; Sowell et al., 2001).
Developmental changes in markers of synaptic inputs to the pyramidal neurons
that participate in thalamocorticothalamic connections occur in a similar fashion.
For example, dendritic spines are the primary sites of excitatory synaptic inputs to
pyramidal cells, and changes in spine number appear to reflect parallel alterations in
excitatory inputs to these neurons (Lund and Holbach, 1991). In monkey DLPFC,
319 Development of thalamocortical circuitry
Fig. 17.4. Age-related changes. (A) Changes during development in the densities of asymmetric exci-
tatory (•) and symmetric inhibitory (◦) synapses in area 46 of monkey dorsolateral prefrontal
cortex (DLPFC). (B) Age-related changes in the mean densities of spines on the dendrites of
layer 3 pyramidal neurons () and of parvalbumin (PV)-containing chandelier neuron axon
cartridges (◦) in layer 3 of monkey DLPFC. Note the parallel time course of developmental
changes in these markers of excitatory and inhibitory inputs to layer 3 pyramidal neurons.
(Adapted from Bourgeois et al., 1994 (A) and Anderson et al., 1995 (B).)
Although the overall density of DLPFC inhibitory synapses does not change
postnatally (Fig. 17.4A) (Bourgeois et al., 1994), functional markers of DLPFC
GABA circuits exhibit substantial postnatal changes. These changes are particularly
prominent in the chandelier class of GABAergic neurons, whose axon terminals are
vertically aligned as morphologically distinct structures (termed “cartridges”) that
form symmetric, inhibitory synapses on the AIS of pyramidal neurons (DeFelipe
et al., 1985). Thus, chandelier neurons provide potent inhibitory regulation of
pyramidal neuron output. Chandelier neurons express the calcium-binding pro-
tein PV (Lewis and Lund, 1990), and PV-containing axon cartridges of chandelier
neurons located in DLPFC layer 3 undergo substantial changes during postnatal
development. As shown in Figure 17.4B, the density of PV-positive cartridges in
monkey DLPFC increases rapidly during the first 3 months of postnatal devel-
opment, undergoes a plateau period that lasts until at least 1.5 years of age, and
then declines during the peripubertal age range to stable adult values (Anderson et
al., 1995). The close temporal match between the developmental changes in spine
density on layer 3 pyramidal neurons and that of PV-containing chandelier neuron
axon cartridges, reflecting a coordination between excitatory and inhibitory inputs,
could be achieved via the synaptic contacts of the axon collaterals of layer 3 pyra-
midal cells onto the dendrites of PV-containing chandelier neurons (Melchitzky
and Lewis, 2003).
Both pre- and postsynaptic markers of GABA neurotransmission at the AIS of
pyramidal neurons also undergo postnatal developmental changes. Like PV, anti-
bodies against the GABA membrane transporter (GAT-1) label the axon cartridges
of chandelier cells. The density of GAT-1-positive cartridges rapidly increases from
birth to 3 months of age, when maximal levels are achieved (Cruz et al., 2003).
GAT-1-positive cartridge density is maintained at these high levels until adoles-
cence, after which a rapid decline brings the density to adult levels. In contrast,
immunoreactivity for the GABAA α 2 -receptor at postsynaptic AIS is greatest at
birth, after which the density of GABAA α 2 -receptor-immunoreactive AIS steadily
decreases to adult levels (Cruz et al., 2003).
The specificity of these postnatal developmental changes in chandelier neuron
inputs to pyramidal cells is illustrated by comparison with the development of
the axon terminals of another subclass of PV-expressing GABA cells, the wide-
arbor neuron. For example, the density of PV-positive axon terminals arising from
wide-arbor cells is low at birth in monkeys and then increases 10-fold, reaching
adult levels approximately 3–4 years of age (Erickson and Lewis, 2002). Because
changes in activity level are associated with corresponding changes in PV content
(Heizmann, 1984), these findings suggest that chandelier neurons provide intra-
columnar inhibition at early postnatal ages, whereas wide-arbor cells supply inter-
columnar inhibition later during postnatal development (Erickson and Lewis,
321 Development of thalamocortical circuitry
2002). This gradual shift from intra- to intercolumnar inhibition may be related to
the late emergence of mature functional properties in the DLPFC, such as spatially
tuned delay-period activity between adjacent GABA and pyramidal neurons (Rao
et al., 1999).
Fig. 17.5. Schematic diagram summarizing the findings from postmortem studies showing alterations
in mediodorsal nucleus of the thalamus (MDN) and dorsolateral prefrontal cortex (DLPFC)
connectivity in schizophrenia. (1) The MDN exhibits a 30–35% reduction in neuron number
and (2) axon terminals from the MDN are reduced by 25% in the thalamic recipient layers
in the DLPFC. (3) Dendritic spine density is decreased by 25% on pyramidal neurons in
deep layer 3 of DLPFC, and (4) the somal size of pyramidal neurons is reduced by 10%.
(5) A subpopulation of gamma-aminobutyric acid (GABA)-ergic neurons exhibit decreased
glutamic acid decarboxylase isoform of 67 kDa (GAD67 ), GABA membrane transporter
1 (GAT-1) and parvalbumin (PV) mRNAs, whereas (6) calretinin (CR) mRNA levels are
unchanged. Therefore, the affected subclass of GABA cells is likely to be the PV-containing
chandelier neuron and not the CR-containing double bouquet cell, an interpretation sup-
ported by (7) the decreased density of GAT1-containing axon cartridges and (8) the
increased density of GABAA α 2 -receptor-immunoreactive pyramidal neuron axon initial seg-
ments. (9) In layer 6, the location of the pyramidal neurons that project to MDN, the density
of dopamine (DA)-containing axons is decreased. (Adapted from Lewis and Lieberman,
2000.)
also cause reduced dendritic spine density in the thalamic recipient layers in the
DLPFC. Indeed, in schizophrenia, spine density is decreased on the basilar dendrites
of DLPFC layer 3 pyramidal cells (Glantz and Lewis, 2000). Furthermore, the largest
reduction in spine density was for those pyramidal cells whose basilar dendrites
extend through the thalamic recipient zone. In addition, the somal size of deep layer
3 pyramidal neurons is reduced in the DLPFC of schizophrenia subjects (Fig. 17.5)
(Pierri et al., 2001; Rajkowska et al., 1998), a finding that may reflect deafferentation.
However, only a subpopulation of deep layer 3 pyramidal neurons appear to be
reduced in somal size (Pierri et al., 2003), making it unlikely that a reduction in
thalamic inputs accounts for this observation.
The presence of reciprocal excitatory connections between the DLPFC and MDN
raises the question of whether corticothalamic projections are also compromised.
Although one study reported a decreased neuronal density in layer 6 of the DLPFC
(Benes et al., 1986), the primary site of origin of corticothalamic projections, other
studies of neuronal size and number have not revealed any changes in this layer
(Rajkowska et al., 1998). These findings suggest that abnormalities in DLPFC effer-
ent projection neurons do not contribute significantly to abnormalities in MDN–
DLPFC connectivity. However, the density of dopamine-containing projections to
the DLPFC, which principally target pyramidal cells, is reduced in layer 6 in sub-
jects with schizophrenia (Fig. 17.5) (Akil et al., 1999). Therefore, although layer 6
pyramidal cells appear to be unaffected structurally in schizophrenia, more subtle
disturbances in afferents that could affect the function of these neurons may be
present in schizophrenia.
A number of findings consistent with functional anomalies in DLPFC GABA
interneurons have been reported in schizophrenia, and recent studies have
revealed that a subset of these neurons, the chandelier class, may be preferentially
affected (Volk and Lewis, 2003). Abnormal function of DLPFC chandelier cells in
schizophrenia is suggested by the finding that reductions in the mRNA level of
the 67 kDa isoform of glutamic acid decarboxylase (GAD67 ) (Akbarian et al., 1995;
Guidotti et al., 2000), an enzyme involved in GABA synthesis, are present in a subset
of GABA neurons found in the same cortical layers in which the chandelier cells
are distributed (Fig. 17.5) (Volk et al., 2000, 2001). In addition, decreased GAT-1
mRNA in a subset of GABA neurons is paralleled by an apparent reduction in GAT-1
protein in the axon cartridges of chandelier cells (Fig. 17.5) (Volk et al., 2001).
Finally, GAD67 mRNA appears to be selectively decreased in PV-containing GABA
neurons and not in neurons that contain another calcium-binding protein, calre-
tinin, which is not found in chandelier neurons (Fig. 17.5) (Hashimoto et al., 2003).
If these reductions in the expression of mRNAs and proteins reflect a deficit
in synaptic GABA, one would expect compensatory upregulation of postsynaptic
receptors. As noted above, immunoreactivity for the α 2 -subunit of the GABAA
324 D. S. Melchitzky and D. A. Lewis
REFERENCES
Akbarian, S., Kim, J. J., Potkin, S. G., et al. (1995). Gene expression for glutamic acid decarboxylase
is reduced without loss of neurons in prefrontal cortex of schizophrenics. Arch Gen Psychiatry
52: 258–266.
Akil, M., Pierri, J. N., Whitehead, R. E., et al. (1999). Lamina-specific alteration in the dopamine
innervation of the prefrontal cortex in schizophrenic subjects. Am J Psychiatry 156: 1580–
1589.
Alexander, G. E. (1982). Functional development of frontal association cortex in monkeys:
behavioral and electrophysiological studies. Neurosci Res Prog Bull 20: 471–479.
Alexander, G. E., Goldman, P. S. (1978). Functional development of the dorsolateral prefrontal
cortex: an analysis utilizing reversible cryogenic depression. Brain Res 143: 233–249.
Anderson, S. A., Classey, J. D., Condé, F., Lund, J. S., Lewis, D. A. (1995). Synchronous develop-
ment of pyramidal neuron dendritic spines and parvalbumin-immunoreactive chandelier
neuron axon terminals in layer III of monkey prefrontal cortex. Neuroscience 67: 7–22.
Barbas, H., Haswell Henion, T. H., Dermon, C. R. (1991). Diverse thalamic projections to the
prefrontal cortex in the rhesus monkey. J Comp Neurol 313: 65–94.
Benes, F. M., Davidson, J., Bird, E. D. (1986). Quantitative cytoarchitectural studies of the cerebral
cortex of schizophrenics. Arch Gen Psychiatry 43: 31–35.
Bourgeois, J.-P., Goldman-Rakic, P. S., Rakic, P. (1994). Synaptogenesis in the prefrontal cortex
of rhesus monkeys. Cereb Cortex 4: 78–96.
Bunge, S. A., Dudukovic, N. M., Thomason, M. E., Vaidya, C. J., Gabrieli, J. D. E. (2002). Immature
frontal lobe contributions to cognitive control in children: Evidence from MRI. Neuron 33:
311.
Byne, W., Buchsbaum, M. S., Kemether, E., et al. (2001). Magnetic resonance imaging of the
thalamic mediodorsal nucleus and pulvinar in schizophrenia and schizotypal personality
disorder. Arch Gen Psychiatry 58: 133–140.
Byne, W., Buchsbaum, M. S., Mattiace, L. A., et al. (2002). Postmortem assessment of thalamic
nuclear volumes in subjects with schizophrenia. Am J Psychiatry 159: 59–65.
Casey, B. J., Giedd, J. N., Thomas, K. M. (2000). Structural and functional brain development
and its relation to cognitive development. Biol Psychol 54: 241–257.
Condé, F., Lund, J. S., Jacobowitz, D. M., Baimbridge, K. G., Lewis, D. A. (1994). Local circuit neu-
rons immunoreactive for calretinin, calbindin D-28k, or parvalbumin in monkey prefrontal
cortex: distribution and morphology. J Comp Neurol 341: 95–116.
326 D. S. Melchitzky and D. A. Lewis
Cruz, D. A., Eggan, S. M., Lewis, D. A. (2003). Postnatal development of pre- and post-synaptic
GABA markers at chandelier cell inputs to pyramidal neurons in monkey prefrontal cortex.
J Comp Neurol 465: 385–400.
DeFelipe, J., Jones, E. G. (1991). Parvalbumin immunoreactivity reveals layer IV of monkey
cerebral cortex as a mosaic of microzones of thalamic afferent terminations. Brain Res 562:
39–47.
DeFelipe, J., Hendry, S. H. C., Jones, E. G., Schmechel, D. (1985). Variability in the terminations of
GABAergic chandelier cell axons on initial segments of pyramidal cell axons in the monkey
sensory-motor cortex. J Comp Neurol 231: 364–384.
Diamond, A. (1985). Development of the ability to use recall to guide action, as indicated by
infants’ performances on AB. Child Devel 56: 868–883.
(2002). Normal development of prefrontal cortex from birth to young adulthood: cognitive
functions, anatomy, and biochemistry. In Principles of Frontal Lobe Function, ed. D. T. Stuss,
R. T. Knight. London: Oxford University Press, pp. 466–503.
Elvevåg, B., Goldberg, T. E. (2000). Cognitive impairment in schizophrenia is the core of the
disorder. Crit Rev Neurobiol 14: 1–21.
Erickson, S. L., Lewis, D. A. (2000). Prefrontal cortical inputs to monkey mediodorsal thalamus.
Soc Neurosci Abstr 26: 1237.
(2002). Postnatal development of parvalbumin- and GABA transporter-immunoreactive axon
terminals in monkey prefrontal cortex. J Comp Neurol 448: 186–202.
Funahashi, S., Bruce, C. J., Goldman-Rakic, P. S. (1989). Mnemonic coding of visual space in the
monkey’s dorsolateral prefrontal cortex. J Neurophysiol 61: 331–349.
Fuster, J. M. (1997). The Prefrontal Cortex: Anatomy, Physiology, and Neuropsychology of the Frontal
Lobe. Philadelphia, PA: Lippincott-Raven.
Fuster, J. J., Alexander, G. E. (1971). Neuron activity related to short-term memory. Science 173:
652–654.
Ghosh, A., Antonini, A., McConnell, S. K., Shatz, C. J. (1990). Requirement for subplate neurons
in the formation of thalamocortical connections. Nature 347: 179–181.
Giedd, J. N. (1999). Brain development during childhood and adolescence: a longitudinal MRI
study. Nat Neurosci 2: 861–863.
Giguere, M., Goldman-Rakic, P. S. (1988). Mediodorsal nucleus: areal, laminar, and tangential
distribution of afferents and efferents in the frontal lobe of rhesus monkeys. J Comp Neurol
277: 195–213.
Gilbert, A. R., Rosenberg, D. R., Harenski, K., et al. (2001). Thalamic volumes in patients with
first-episode schizophrenia. Am J Psychiatry 158: 618–624.
Glantz, L. A., Lewis, D. A. (2000). Decreased dendritic spine density on prefrontal cortical pyra-
midal neurons in schizophrenia. Arch Gen Psychiatry 57: 65–73.
Goldman-Rakic, P. S. (1987). Development of cortical circuitry and cognitive function. Child
Devel 58: 601–622.
(1994). Working memory dysfunction in schizophrenia. J Neuropsychiatry 6: 348–357.
(1995). Cellular basis of working memory. Neuron 14: 477–485.
Goldman-Rakic, P. S., Porrino, L. J. (1985). The primate mediodorsal (MD) nucleus and its
projection to the frontal lobe. J Comp Neurol 242: 535–560.
327 Development of thalamocortical circuitry
Grutzendler, J., Kasthuri, N., Gan, W. B. (2002). Long-term dendritic spine stability in the adult
cortex. Nature 420: 812–816.
Guidotti, A., Auta, J., Davis, J. M., et al. (2000). Decrease in reelin and glutamic acid
decarboxylase67 (GAD67 ) expression in schizophrenia and bipolar disorder. Arch Gen
Psychiatry 57: 1061–1069.
Harrison, P. J., Lewis, D. A. (2003). Neuropathology in schizophrenia. In Schizophrenia, 2nd edn,
ed. S. Hirsch, D. R. Weinberger. Oxford: Blackwell Science, Oxford University Press, pp.
310–325.
Hashimoto, T., Volk, D. W., Eggan, S. M., et al. (2003). Gene expression deficits in a subclass
of GABA neurons in the prefrontal cortex of subjects with schizophrenia. J Neurosci 23:
6315–6326.
Heizmann, C. W. (1984). Parvalbumin an intracellular calcium-binding protein. Distribution
properties and possible roles in mammalian cells. Experientia 40: 910–921.
Huttenlocher, P. R. (1979). Synaptic density in human frontal cortex: developmental changes and
effects of aging. Brain Res 163: 195–205.
Isseroff, A., Rosvold, H. E., Galkin, T. W., Goldman-Rakic, P. S. (1982). Spatial memory impair-
ments following damage to the mediodorsal nucleus of the thalamus in rhesus monkeys.
Brain Res 232: 97–113.
Jones, E. G., Hendry, S. H. C., DeFelipe, J., Benson, D. L. (1994). GABA neurons and their role
in activity-dependent plasticity of adult primate visual cortex. In Cerebral Cortex, Vol. 10:
Primary Visual Cortex in Primates, ed. A. Peters, K. S. Rockland. New York: Plenum Press,
pp. 61–140.
Konick, L. C., Friedman, L. (2001). Meta-analysis of thalamic size in schizophrenia. Biol Psychiatry
49: 28–38.
Kuroda, M., Yokofujita, J., Murakami, K. (1998). An ultrastructural study of the neural circuit
between the prefrontal cortex and the mediodorsal nucleus of the thalamus. Prog Neurobiol
54: 417–458.
Levitt, J. B., Lewis, D. A., Yoshioka, T., Lund, J. S. (1993). Topography of pyramidal neuron
intrinsic connections in macaque monkey prefrontal cortex (areas 9 and 46). J Comp Neurol
338: 360–376.
Lewis, D. A. (2000). Is there a neuropathology of schizophrenia? The Neuroscientist 6: 208–218.
(2002). Neural circuitry approaches to understanding the pathophysiology of schizophrenia.
In Neuropsychopharmacology: The Fifth Generation of Progress, ed. K. L. Davis, D. S. Charney,
J. T. Coyle, C. B. Nemeroff. Philadelphia, PA: Lippincott Williams and Wilkins, pp. 729–743.
Lewis, D. A., Levitt, P. (2002). Schizophrenia as a disorder of neurodevelopment. Annu Rev
Neurosci 25: 409–432.
Lewis, D. A., Lieberman, J. A. (2000). Catching up on schizophrenia: natural history and neuro-
biology. Neuron 28: 325–334.
Lewis, D. A., Lund, J. S. (1990). Heterogeneity of chandelier neurons in monkey neocor-
tex: corticotropin-releasing factor and parvalbumin immunoreactive populations. J Comp
Neurol 293: 599–615.
Lewis, D. A., Cruz, D. A., Melchitzky, D. S., Pierri, J. N. (2001). Lamina-specific reductions
in parvalbumin-immunoreactive axon terminals in the prefrontal cortex of subjects with
328 D. S. Melchitzky and D. A. Lewis
schizophrenia: evidence for decreased projections from the thalamus. Am J Psychiatry 158:
1411–1422.
Lund, J. S., Holbach, S. (1991). Postnatal development of thalamic recipient neurons in monkey
striate cortex: I. A comparison of spine acquisition and dendritic growth of layer 4C alpha
and beta spiny stellate neurons. J Comp Neurol 309: 115–128.
McCarley, R. W., Wible, C. G., Frumin, M., et al. (1999). MRI anatomy of schizophrenia. Biol
Psychiatry 45: 1099–1119.
Melchitzky, D. S., Lewis, D. A. (2003). Preferential targeting of parvalbumin interneurons by
local axon terminals of supragranular pyramidal neurons in monkey prefrontal cortex.
Cereb Cortex 13: 452–460.
Melchitzky, D. S., Sesack, S. R., Pucak, M. L., Lewis, D. A. (1998). Synaptic targets of pyramidal
neurons providing intrinsic horizontal connections in monkey prefrontal cortex. J Comp
Neurol 390: 211–224.
Melchitzky, D. S., Sesack, S. R., Lewis, D. A. (1999). Parvalbumin-immunoreactive axon terminals
in monkey and human prefrontal cortex: Laminar, regional and target specificity of type I
and type II synapses. J Comp Neurol 408: 11–22.
Melchitzky, D. S., Gonzalez-Burgos, G., Barrionuevo, G., Lewis, D. A. (2001). Synaptic targets
of the intrinsic axon collaterals of supragranular pyramidal neurons in monkey prefrontal
cortex. J Comp Neurol 430: 209–221.
Pakkenberg, B. (1990). Pronounced reduction of total neuron number in mediodorsal thalamic
nucleus and nucleus accumbens in schizophrenics. Arch Gen Psychiatry 47: 1023–1028.
Park, S., Holzman, P. S. (1992). Schizophrenics show spatial working memory deficits. Arch Gen
Psychiatry 49: 975–982.
Pearlson, G. D., Marsh, L. (1999). Structural brain imaging in schizophrenia: a selective review.
Biol Psychiatry 46: 627–649.
Pierri, J. N., Volk, C. L. E., Auh, S., Sampson, A., Lewis, D. A. (2001). Decreased somal size of
deep layer 3 pyramidal neurons in the prefrontal cortex in subjects with schizophrenia. Arch
Gen Psychiatry 58: 466–473.
(2003). Somal size of prefrontal cortical pyramidal neurons in schizophrenia: differential effects
across neuronal subpopulations. Biol Psychiatry 54: 111–120.
Popken, G. J., Bunney, Jr W. E., Potkin, S. G., Jones, E. G. (2000). Subnucleus-specific loss
of neurons in medial thalamus of schizophrenics. Proc Natl Acad Sci USA 97: 9276–
9280.
Portas, C. M., Goldstein, J. M., Shenton, M. E., et al. (1998). Volumetric evaluation of the
thalamus in schizophrenic male patients using magnetic resonance imaging. Biol Psychiatry
43: 649–659.
Pucak, M. L., Levitt, J. B., Lund, J. S., Lewis, D. A. (1996). Patterns of intrinsic and associational
circuitry in monkey prefrontal cortex. J Comp Neurol 376: 614–630.
Rajkowska, G., Selemon, L. D., Goldman-Rakic, P. S. (1998). Neuronal and glial somal size in
the prefrontal cortex: a postmortem morphometric study of schizophrenia and Huntington
disease. Arch Gen Psychiatry 55: 215–224.
Rakic, P. (1977). Prenatal development of the visual system in rhesus monkey. Philos Trans R Soc
Lond Ser B 278: 245–260.
329 Development of thalamocortical circuitry
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
330
331 X chromosome, estrogen, and brain development
pronounced in women (Lerer et al., 1999). Also there are age-related reductions in
the number of cortical serotonin receptors 5-HT1A , 5-HT1Bδ : and 5-HT2A in the
frontal and occipital lobes and the hippocampus (Meltzer et al., 1998).
Dopaminergic neurons begin to develop by 6–8 weeks of gestation in humans
(Sundstrom et al., 1993), and development begins earlier in females than in
males (Herlenius and Lagercrantz, 2001). Interestingly, with the onset of puberty,
dopaminergic activity (unlike serotoninergic) increases in some cortical regions,
especially the frontal cortex (Spear, 2000). It has been hypothesized that the
increased risk for the onset of psychosis during adolescence/early adulthood may
be linked to the elevation in dopaminergic activity (Walker, 1994).
PET studies of age-related differences in the dopaminergic system reported a
reduction in dopamine D2 receptor concentration of the caudate nuclei and the
frontal lobes in both men and women (Wong et al., 1984). Furthermore, this decline
is more pronounced in women than in men (Wong et al., 1988).
In summary, therefore, there are age and gender differences in the maturation of
brain regions and neurochemical systems implicated in schizophrenia. What then
is the evidence that gender differences in the brain are caused by sex steroids or sex
chromosomes?
circulating α-fetoprotein has been “switched off ” (Fitch and Denenberg, 1998).
In vitro studies suggest that filopodial outgrowth (an important part of neu-
rotrophism because filopodia act as an early support for dendritic spine matu-
ration) occurs within minutes of exposure to 17β-estradiol (Brinton, 1993). The
mechanism by which estrogen initiates filopodial outgrowth is most likely direct
activation of the Rac1B G-protein, a member of the Rho family of GTPases involved
in actin organization (Dumontier et al., 2000). Further development of dendritic
spines appears to be mediated by estrogen activation of a Src tyrosine kinase that
phosphorylates the glutamate N-methyl-d-aspartate (NMDA) receptor (Yu et al.,
1997). For example, in cortical and hippocampal neurons without synaptic con-
tacts, 17β-estradiol induces a significant increase in the outgrowth of both macro-
and micromorphological features, which is blocked by a NMDA receptor antago-
nist. Activation of these intracellular signaling cascades appears to be dependent on
the subtype of estrogen receptor present: neuroblastoma cells with the α-subtype,
for example, are dependent on the Rac1B G-protein, whereas mechanisms appear
independent of Rac1B where the β-subtype occurs (Brinton, 2001).
These findings have been confirmed by in vivo studies. In the adult female rat,
the densities of dendritic spines and synapses on hippocampal CA1 pyramidal cells
fluctuate across the menstrual cycle with natural fluctuations in ovarian steroid
estradiol levels. Furthermore, these estradiol-induced dendritic changes are blocked
by treatment with an NMDA receptor antagonist (Woolley and McEwen, 1994).
In addition to direct effects on neurons, estrogen also acts with neurotrophins
(such as NGF) to stimulate nerve cell growth indirectly. NGF is essential for early
neuronal development and influences neuron differentiation and growth. Other
important neurotrophins are brain-derived neurotrophic factor (BDNF) and neu-
rotrophins 3 and 4/5 (NT-3 and NT-4/5). Binding of NGF and neurotrophins
to their cognate receptors results in the activation of mechanisms necessary for
growth and survival of neurites, as well as in stimulation of functions related
to neurotransmitter production and release. Importantly, receptors for estrogen
and neurotrophins are located on the same neurons in rodent basal forebrain,
hippocampus, and cerebral cortex (Toran-Allerand, 1996). This colocalization of
estrogen and neurotrophin receptors suggests that they act synergistically on the
same neuron to regulate expression of specific genes enhancing neuronal survival,
differentiation, and plasticity (Toran-Allerand, 1996).
5.0
4.0
3.5
3.0
2.5
N
young on off
Fig. 18.1. Mean right hippocampal choline as an indication of neuronal membrane breakdown in
postmenopausal women taking estrogen replacement therapy (“on”) or never using such
therapy (“off”) plus that in premenopausal women (“young”). Bars indicate 95% confidence
internal. “Off” values versus “young,” p < 0.03; “off” values versus “on,” p < 0.04. (From
Robertson D. M. W., et al., Neurology 2001, 57(11): 2114–2117.)
500
AUC PRL (mU × h/l)
400
300
200
100
young on off
X chromosome
It has been suggested that a gene(s) for schizophrenia could be X-linked or X–Y
linked, with homologous loci on both X and Y chromosomes (DeLisi et al., 1994),
but support for this view has been limited and there is currently little evidence
that schizophrenia is an X-linked disorder. Nonetheless, it is clear that cognitive
functions which are usually highly lateralized (e.g. language) and have gender differ-
ences in development are affected in schizophrenia. Also it has been suggested that
schizophrenia arises from abnormalities in development of cerebral asymmetry.
Consequently, it is of interest to investigate if/how the X chromosome affects
hemispheric asymmetry and development/function of brain regions implicated
in schizophrenia.
Normally in women, the second X chromosome is randomly inactivated; how-
ever, some genes escape inactivation and so may have significant effects on brain.
Also, males always receive their X chromosome from their mother whereas “normal”
females inherit one X chromosome from each of their parents. Turner syndrome is
a genetic disorder in which all or part of one X chromosome is deleted (45X). Indi-
viduals with Turner syndrome provide a model where the effect of loss of the second
X chromosome and/or mode of inheritance on the brain can be investigated. We
reported that loss of the second X chromosome affects the development of anatomi-
cal, metabolic, and functional cerebral asymmetry; language; and visuospatial skills
(Murphy et al., 1993, 1994, 1997). We suggested that this most likely arises from
339 X chromosome, estrogen, and brain development
Conclusions
Both estrogen and the X chromosome affect the development of brain regions
and neurochemical systems that are both crucial to higher cognitive function and
implicated in neuropsychiatric disorders such as schizophrenia.
There are gender differences in schizophrenia and because estrogen has puta-
tive antidopaminergic/antipsychotic actions, it has been suggested that estrogen
may be responsible in women for the delayed onset of the first incidence peak
and that the second peak seen in women may result from the decline in estro-
gen levels at menopause. There is evidence that estrogen protects the nigrostriatal
dopaminergic system against the neurotoxic effects of 1-methyl-a-phenyl-2, 2, 3,
6-tetrahydropyridine in rats (Dluzen et al., 1996). An antipsychotic action of estro-
gens is supported by clinical studies reporting that women have increased admission
rates for psychosis around the menses (Hallonquist et al., 1993) and that psychotic
symptomatology varies with the phase of the menstrual cycle (Lindamer et al.,
1997). Also, women with schizophrenia may have reduced estradiol levels compared
with non-schizophrenia controls (Riecher-Rossler et al., 1994). However, there is
currently little evidence to support the therapeutic use of estrogen in schizophre-
nia. For example, when women with schizophrenia are treated with adjunctive
estrogen there is a slight increase in speed of recovery, but no improvement overall
compared with antipsychotic medication alone (Kulkarni et al., 2001). Despite the
lack of clear evidence for the efficacy of estrogen as an antipsychotic treatment,
it remains plausible that estrogen replacement therapy might protect against late-
onset schizophrenia in postmenopausal women by reducing age-related changes in
brain structure and neurochemistry (e.g. in the hippocampus).
We do not propose that estrogen and the X chromosome are crucial to the
development of schizophrenia in most people. Rather, we suggest that they play a
modulatory role in brain maturation, and their mechanism of action needs to be
341 X chromosome, estrogen, and brain development
REFERENCES
Andreasen, N. C., Smith, M. R., Jacoby, C. G., Dennert, J. W., Olsen, S. A. (1982). Ventricular
enlargement in schizophrenia: definition and prevalence. Am J Psychiatry 139: 292–296.
Andreasen, N. C., Nasrallah, H. A., Dunn, et al. (1986). Structural abnormalities in the frontal
system in schizophrenia: a magnetic resonance imaging study. Arch Gen Psychiatry 43: 136–
144.
Andreasen, N. C., Ehrhardt, J. C., Swayze, III, V. W., et al. (1990a). Magnetic resonance imaging of
the brain in schizophrenia. The pathophysiologic significance of structural abnormalities.
Arch Gen Psychiatry 47: 35–44.
Andreasen, N. C., Swayze, III, V. W., Flaum, M., et al. (1990b). Ventricular enlargement in
schizophrenia evaluated with computed tomographic scanning. Effects of gender, age, and
stage of illness. Arch Gen Psychiatry 47: 1008–1015.
Bedard, P. J., Boucher, R., Daigle, M., di Paolo, T. (1984). Similar effect of estradiol and haloperidol
on experimental tardive dyskinesia in monkeys. Psychoneuroendocrinology 9: 375–379.
Benes, F. M. (1998). Brain development, VII. Human brain growth spans decades. Am J Psychiatry
155: 1489.
Benes, F. M., Turtle, M., Khan, Y., Farol, P. (1994). Myelination of a key relay zone in the hippocam-
pal formation occurs in the human brain during childhood, adolescence, and adulthood.
Arch Gen Psychiatry 51: 477–484.
Bennett-Clarke, C. A., Leslie, M. J., Lane, R. D., Rhoades, R. W. (1994). Effect of serotonin
depletion on vibrissa-related patterns of thalamic afferents in the rat’s somatosensory cortex.
J Neurosci 14: 7594–7607.
Bennett-Clarke, C. A., Chiaia, N. L., Rhoades, R. W. (1996). Thalamocortical afferents in rat
transiently express high-affinity serotonin uptake sites. Brain Res 733: 301–306.
Blue, M. E., Erzurumlu, R. S., Jhaveri, S. (1991). A comparison of pattern formation by thalam-
ocortical and serotonergic afferents in the rat barrel field cortex. Cereb Cortex 1: 380–389.
Brinton, R. D. (1993). 17-Estradiol induction of filopodial growth in cultured hippocampal
neurons within minutes of exposure. Mol Cell Neurosci 4: 36–46.
(2001). Cellular and molecular mechanisms of estrogen regulation of memory function and
neuroprotection against Alzheimer’s disease: recent insights and remaining challenges. Learn
Mem 8: 121–133.
Cases, O., Seif, I., Grimsby, J., Gaspar, P., et al. (1995). Aggressive behavior and altered amounts
of brain serotonin and norepinephrine in mice lacking MAOA. Science 268: 1763–1766.
Cases, O., Vitalis, T., Seif, I., et al. (1996). Lack of barrels in the somatosensory cortex of
monoamine oxidase A-deficient mice: role of a serotonin excess during the critical period.
Neuron 16: 297–307.
Castle, D. J., Murray, R. M. (1993). The epidemiology of late-onset schizophrenia. Schizophr Bull
19: 691–700.
342 M. Craig, W. Cutter, R. Norbury, and D. Murphy
Castle, D. J., Wessely, S., Murray, R. M. (1993). Sex and schizophrenia: effects of diag-
nostic stringency, and associations with and premorbid variables. Br J Psychiatry 162:
658–664.
Coger, R. W., Serafetinides, E. A. (1990). Schizophrenia, corpus callosum, and interhemispheric
communication: a review. Psychiatr Res 34: 163–184.
D’Amato, R. J., Blue, M. E., Largent, B. L., et al. (1987). Ontogeny of the serotonergic projection
to rat neocortex: transient expression of a dense innervation to primary sensory areas. Proc
Natl Acad Sci USA 84: 4322–4326.
De Bellis, M. D., Keshavan, M. S., Beers, S. R., et al. (2001). Sex differences in brain maturation
during childhood and adolescence. Cereb Cortex 11: 552–557.
DeLisi, L. E., Devoto, M., Lofthouse, R., et al. (1994). Search for linkage to schizophrenia on the
X and Y chromosomes. Am J Med Genet 54: 113–121.
Dluzen, D. E., McDermott, J. L., Liu, B. (1996). Estrogen alters MPTP-induced neurotoxicity
in female mice: effects on striatal dopamine concentrations and release. J Neurochem 66:
658–666.
Dumontier, M., Hocht, P., Mintert, U., Faix, J. (2000). Rac1 GTPases control filopodia formation,
cell motility, endocytosis, cytokinesis and development in Dictyostelium. J Cell Sci 113:
2253–2265.
Egan, M. F., Goldberg, T. E., Kolachana, B. S., et al. (2001). Effect of COMT Val108/158 Met
genotype on frontal lobe function and risk for schizophrenia. Proc Natl Acad Sci USA 98:
6917–6922.
Fitch, R. H., Denenberg, V. H. (1998). A role for ovarian hormones in sexual differentiation of
the brain. Behav Brain Sci 21: 311–327; discussion 327–352.
Flaum, M., Arndt, S., Andreasen, N. C. (1990). The role of gender in studies of ventricle enlarge-
ment in schizophrenia: a predominantly male effect. Am J Psychiatry 147: 1327–1332.
Foreman, M. M., Porter, J. C. (1980). Effects of catechol estrogens and catecholamines on hypotha-
lamic and corpus striatal tyrosine hydroxylase activity. J Neurochem 34: 1175–1183.
Freund, L. S., Reiss, A. L., Hagerman, R., Vinogradov, S. (1992). Chromosome fragility and
psychopathology in obligate female carriers of the fragile X syndrome. Arch Gen Psychiatry
49: 54–60.
Fryns, J. P. (1986). The female and the fragile X A study of 144 obligate female carriers. Am J Med
Genet 23: 157–169.
Giedd, J. N., Vaituzis, A. C., Hamburger, S. D., et al. (1996). Quantitative MRI of the temporal
lobe, amygdala, and hippocampus in normal human development: ages 4–18 years. J Comp
Neurol 366: 223–230.
Giedd, J. N., Blumenthal, J., Jeffries, N. O., et al. (1999). Brain development during childhood
and adolescence: a longitudinal MRI study. Nat Neurosci 2: 861–863.
Goldman-Rakic, P. S., Brown, R. M. (1982). Postnatal development of monoamine content and
synthesis in the cerebral cortex of rhesus monkeys. Brain Res 256: 339–349.
Gordon, J. H., Borison, R. L., Diamond, B. I. (1980). Modulation of dopamine receptor sensitivity
by estrogen. Biol Psychiatry 15: 389–396.
Gur, R. E., Mozley, P. D., Shtasel, D. L., et al. (1994). Clinical subtypes of schizophrenia: differences
in brain and CSF volume. Am J Psychiatry 151: 343–350.
343 X chromosome, estrogen, and brain development
Haas, G. L., Hien, D. A., Waked, W., et al. (1989). Sex differences in schizophrenia. Schizophr Res
2: 11.
Haas, G. L., Sweeney, J. A., Hien, D. A., Goldman, D., Deck, M. (1991). Gender differences in
schizophrenia. Schizophr Res 4: 277.
Hafner, H., Riecher-Rossler, A., Maurer, K., Fatkenheuer, B., Loffler, W. (1993). Generating and
testing a causal explanation of the gender difference in age at first onset of schizophrenia.
Psychol Med 23: 925–940.
Hafner, H., Maurer, K., Loffler, W., et al. (1994). The epidemiology of early schizophrenia. Influ-
ence of age and gender on onset and early course. Br J Psychiatry Suppl 23: 29–38.
Hafner, H., An der Heiden, W., Behrens, S., et al. (1998a). Causes and consequences of the gender
difference in age at onset of schizophrenia. Schizophr Bull 24: 99–113.
Hafner, H., Maurer, K., Loffler, W., et al. (1998b). The ABC Schizophrenia Study: a preliminary
overview of the results. Soc Psychiatry Psychiatr Epidemiol 33: 380–386.
Hagerman, R. J. (1987). Fragile X syndrome. Curr Probl Pediatr 17: 627–674.
Hallonquist, J. D., Seeman, M. V., Lang, M., Rector, N. A. (1993). Variation in symptom severity
over the menstrual cycle of schizophrenics. Biol Psychiatry 33: 207–209.
Hampson, E. (1990). Variations in sex-related cognitive abilities across the menstrual cycle. Brain
Cogn 14: 26–43.
Hariri, A. R., Mattay, V. S., Tessitore, A., et al. (2002). Serotonin transporter genetic variation and
the response of the human amygdala. Science 297: 400–403.
Harvey, I., Williams, M., Toone, B. K., et al. (1990). The ventricular–brain ratio (VBR) in func-
tional psychoses: the relationship of lateral ventricular and total intracranial area. Psychol
Med 20: 55–62.
Hedner, J., Lundell, K. H., Breese, G. R., Mueller, R. A., Hedner, T. (1986). Developmental
variations in CSF monoamine metabolites during childhood. Biol Neonate 49: 190–197.
Herlenius, E., Lagercrantz, H. (2001). Neurotransmitters and neuromodulators during early
human development. Early Hum Dev 65: 21–37.
Howard, R., Rabins, P. V., Seeman, M. V., Jeste, D. V., et al. (2000). Late-onset schizophrenia and
very-late-onset schizophrenia-like psychosis: an international consensus. Am J Psychiatry
157: 172–178.
Jablensky, A., Sartorius, N., Ernberg, G. (1992). Schizophrenia: manifestations, incidence and
course in different cultures. A World Health Organization Ten-Country Study. Psychol Med
Monograph 20.
Kendell, R. E., Chalmers, J. C., Platz, C. (1987). Epidemiology of puerperal psychoses. Br J
Psychiatry 150: 662–673.
Kulkarni, J., Riedel, A., de Castella, A. R., et al. (2001). Estrogen: a potential treatment for
schizophrenia. Schizophr Res 48: 137–144.
Lauder, J. M., Krebs, H. (1978). Serotonin as a differentiation signal in early neurogenesis. Dev
Neurosci 1: 15–30.
Lauriello, J., Hoff, A., Wieneke, M. H., et al. (1997). Similar extent of brain dysmorphology in
severely ill women and men with schizophrenia. Am J Psychiatry 154: 819–825.
Lebrand, C., Cases, O., Adelbrecht, C., et al. (1996). Transient uptake and storage of serotonin in
developing thalamic neurons. Neuron 17: 823–835.
344 M. Craig, W. Cutter, R. Norbury, and D. Murphy
Lerer, B., Gelfin, Y., Shapira, B. (1999). Neuroendocrine evidence for age-related decline in central
serotonergic function. Neuropsychopharmacology 21: 321–322.
Leung, A., Chue, P. (2000). Sex differences in schizophrenia, a review of the literature. Acta
Psychiatr Scand Suppl 401: 3–38.
Levallois, C., Valence, C., Baldet, P., Privat, A. (1997). Morphological and morphometric anal-
ysis of serotonin-containing neurons in primary dissociated cultures of human rhomben-
cephalon: a study of development. Brain Res Dev Brain Res 99: 243–252.
Lidow, M. S., Goldman-Rakic, P. S., Rakic, P. (1991). Synchronized overproduction of neuro-
transmitter receptors in diverse regions of the primate cerebral cortex. Proc Natl Acad Sci
USA 88: 10218–10221.
Lindamer, L. A., Lohr, J. B., Harris, M. J., Jeste, D. V. (1997). Gender, estrogen, and schizophrenia.
Psychopharmacol Bull 33: 221–228.
Maki, P. M., Resnick, S. M. (2000). Longitudinal effects of estrogen replacement therapy on PET
cerebral blood flow and cognition. Neurobiol Aging 21: 373–383.
(2001). Effects of estrogen on patterns of brain activity at rest and during cognitive activity: A
review of neuroimaging studies. Neuroimage 14: 789–801.
McInnis, M. G., McMahon, F. J., Chase, G. A., et al. (1993). Anticipation in bipolar affective
disorder. Am J Hum Genet 53: 385–390.
Meltzer, C. C., Smith, G., DeKosky, S. T., et al. (1998). Serotonin in aging, late-life depression, and
Alzheimer’s disease: the emerging role of functional imaging. Neuropsychopharmacology 18:
407–430.
Morris, A. G., Gaitonde, E., McKenna, P. J., Mollon, J. D., Hunt, D. M. (1995). CAG repeat
expansions and schizophrenia: association with disease in females and with early age-at-
onset. Hum Mol Genet 4: 1957–1961.
Moses, E. L., Drevets, W. C., Smith, G., et al. (2000). Effects of estradiol and progesterone
administration on human serotonin 2A receptor binding: a PET study. Biol Psychiatry 48:
854–860.
Murphy, D. G., DeCarli, C., Daly, E., et al. (1993). X-chromosome effects on female brain: a
magnetic resonance imaging study of Turner’s syndrome. Lancet 342: 1197–1200.
Murphy, D. G., Allen, G., Haxby, J. V., et al. (1994). The effects of sex steroids, and the X chromo-
some, on female brain function: a study of the neuropsychology of adult Turner syndrome.
Neuropsychologia 32: 1309–1323.
Murphy, D. G., DeCarli, C., McIntosh, A. R., et al. (1996). Sex differences in human brain
morphometry and metabolism: an in vivo quantitative magnetic resonance imaging and
positron emission tomography study on the effect of aging. Arch Gen Psychiatry 53: 585–594.
Murphy, D. G., Mentis, M. J., Pietrini, P., et al. (1997). A PET study of Turner’s syndrome: effects
of sex steroids and the X chromosome on brain. Biol Psychiatry 41: 285–298.
Nasrallah, H. A., Schwarzkopf, S. B., Olson, S. C., Coffman, J. A. (1990). Gender differences in
schizophrenia on MRI brain scans. Schizophr Bull 16: 205–210.
Nopoulos, P., Flaum, M., Andreasen, N. C. (1997). Sex differences in brain morphology in
schizophrenia. Am J Psychiatry 154: 1648–1654.
O’Connell, P., Woodruff, P. W. R., Wright, I., Jones, P., Murray, R. M. (1997). Developmental
insanity or dementia praecox: was the wrong concept adopted? Schizophr Res 23: 97–106.
345 X chromosome, estrogen, and brain development
O’Donovan, M. C., Guy, C., Craddock, N., et al. (1995). Expanded CAG repeats in schizophrenia
and bipolar disorder. Nat Genet 10: 380–381.
Phillips, S. M., Sherwin, B. B. (1992). Effects of estrogen on memory function in surgically
menopausal women. Psychoneuroendocrinology 17: 485–495.
Phoenix, C. H., Goy, R. W., Gerall, A. A., Young, W. C. (1959). Organizing action of prenatally
administered testosterone proprionate on the tissues mediating behavior in the guinea pig.
Endocrinology 65: 369–382.
Riecher-Rossler, A., Hafner, H., Dutsch-Strobel, A., et al. (1994). Further evidence for a specific
role of estradiol in schizophrenia? Biol Psychiatry 36: 492–494.
Robertson, D. M., van Amelsvoort, T., Daly, E., et al. (2001). Effects of estrogen replacement
therapy on human brain aging: an in vivo 1 H MRS study. Neurology 57: 2114–2117.
Salem, J. E., Kring, A. M. (1998). The role of gender differences in the reduction of etiologic
heterogeneity in schizophrenia. Clin Psychol Rev 18: 795–819.
Seeman, M. V., Lang, M. (1990). The role of estrogens in schizophrenia gender differences.
Schizophr Bull 16: 185–194.
Seifert, Jr., W. E., Foxx, J. L., Butler, I. J. (1980). Age effect on dopamine and serotonin metabolite
levels in cerebrospinal fluid. Ann Neurol 8: 38–42.
Shaywitz, S. E., Shaywitz, B. A., Pugh, K. R., et al. (1999). Effect of estrogen on brain activation
patterns in postmenopausal women during working memory tasks. J Am Med Assoc 281:
1197–1202.
Shelton, R. C., Karson, C. N., Doran, A. R., et al. (1988). Cerebral structural pathology in
schizophrenia: evidence for a selective prefrontal cortical defect. Am J Psychiatry 145: 154–
163.
Shenton, M. E., Kikinis, R., Jolesz, F. A., et al. (1992). Abnormalities of the left temporal lobe
and thought disorder in schizophrenia. A quantitative magnetic resonance imaging study.
N Engl J Med 327: 604–612.
Sherwin, B. B. (1988). Estrogen and/or androgen replacement therapy and cognitive functioning
in surgically menopausal women. Psychoneuroendocrinology 13: 345–357.
Skuse, D. H., James, R. S., Bishop, D. V., et al. (1997). Evidence from Turner’s syndrome of an
imprinted X-linked locus affecting cognitive function. Nature 387: 705–708.
Spear, L. P. (2000). The adolescent brain and age-related behavioral manifestations. Neurosci
Biobehav Rev 24: 417–463.
Spitzer, R., Endicott, J., Robins, E. (1978). Research Diagnostic Criteria (RDC): rationale and
reliability. Arch Gen Psychiatry 35: 773–782.
Sundstrom, E., Kolare, S., Souverbie, F., et al. (1993). Neurochemical differentiation of
human bulbospinal monoaminergic neurons during the first trimester. Dev Brain Res 75:
1–12.
Takahashi, H., Nakashima, S., Ohama, E., Takeda, S., Ikuta, F. (1986). Distribution of serotonin-
containing cell bodies in the brainstem of the human fetus determined with immunohisto-
chemistry using antiserotonin serum. Brain Dev 8: 355–365.
Toran-Allerand, C. D. (1996). The estrogen/neurotrophin connection during neural develop-
ment: is co-localization of estrogen receptors with the neurotrophins and their receptors
biologically relevant? Dev Neurosci 18: 36–48.
346 M. Craig, W. Cutter, R. Norbury, and D. Murphy
Travis, M. J., Mulligan, O., Mulligan, R. S., et al. (1999). Preliminary investigation of the effect
of oestradiol treatment on cortical 5–HT2A receptor binding: a single photon emission
tomography (SPET) study using 123 I -5-I-R91150. Neuroimage 9: S672.
van Amelsvoort, T. A. M. J., Daly, E. Robertson, D. M. R., et al. (2001a). Structural brain abnor-
malities associated with deletion of chromosome 22qaa: quantitative neuroimaging study
of adults with velo-cardio-facial syndrome. Br J Psychiatry 178: 412–419.
van Amelsvoort, T. A. M. J., Abel, K. M., Robertson, D. M. R., et al. (2001b). Prolactin response to
d-fenfluramine in postmenopausal women on and off ERT: comparison with young women.
Psychoneuroendocrinology 26: 493–502.
Vazquez-Barquero, J. L., Cuesta Nunez, M. J., Quintana Pando, F., et al. (1995). Structural abnor-
malities of the brain in schizophrenia: sex differences in the Cantabria First Episode of
Schizophrenia Study. Psychol Med 25: 1247–1257.
Walker, E. F. (1994). Developmentally moderated expressions of the neuropathology underlying
schizophrenia. Schizophr Bull 20: 453–480.
Whitaker-Azmitia, P. M. (2001). Serotonin and brain development: role in human developmental
diseases. Brain Res Bull 56: 479–485.
Williams, A. O., Reveley, M. A., Kolakowska, T., Ardern, M., Mandelbrote, B. M. (1985).
Schizophrenia with good and poor outcome. II: Cerebral ventricular size and its clinical
significance. Br J Psychiatry 146: 239–246.
Wong, D. F., Wagner, Jr., H. N., Dannals, R. F., et al. (1984). Effects of age on dopamine and
serotonin receptors measured by positron tomography in the living human brain. Science
226: 1393–1396.
Wong, D. F., Broussolle, E. P., Wand, G., et al. (1988). In vivo measurement of dopamine receptors
in human brain by positron emission tomography. Age and sex differences. Ann N Y Acad
Sci 515: 203–214.
Woolley, C. S., McEwen, B. S. (1994). Estradiol regulates hippocampal dendritic spine density
via an N-methyl-d-aspartate receptor-dependent mechanism. J Neurosci 14: 7680–7687.
Yan, W., Wilson, C. C., Haring, J. H. (1997). Effects of neonatal serotonin depletion on the
development of rat dentate granule cells. Brain Res Dev Brain Res 98: 177–184.
Yu, X.-M., Askalan, R., Keil, II, G. J., Salter, M. W. (1997). NMDA channel regulation by channel-
associated protein tyrosine kinase Src. Science 275: 674–678.
19
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
347
348 S. M. Lawrie
Table 19.1. Systematic reviews of structural brain abnormalities in schizophrenia versus controls
CT, computed tomography; sMRI, structural magnetic resonance imaging; PM, postmortem; ES, effect size;
CSF, cerebrospinal fluid.
349 Premorbid structural abnormalities in schizophrenia
abnormalities may be progressive in the first few years after onset (see Ch. 20).
They are also challenged by the preliminary findings from prospective high-risk
studies.
high-risk subjects with particularly small AHCs (and assuming that those who
develop schizophrenia will ultimately have AHCs that approximate to those of
FES), the reduced AHC volume may be a trait marker. Prefrontal lobe and thalami
volumes, but not those of the AHC, were associated with measures of genetic
liability (Lawrie et al., 2001). The apparent absence of a relationship between AHC
volume and genetic liability was unexpected and may be attributable to a relatively
narrow range of liability in the Edinburgh High Risk Study or that hippocampal
volumes particularly are related to OCs (see below). If, however, there are further
AHC reductions in those who develop schizophrenia, this might reflect genetic
effects on later neurodevelopment (but could also reflect environmental effects or
a peri-onset effect on plasticity).
In those with psychotic symptoms at any point in the first 5 years of the study,
the whole brain volume at study entry was reduced (after controlling for age,
sex, paternal social class, height, and handedness) compared with those did not
have psychotic symptoms over this time (Lawrie et al., 2001). Those with psy-
chotic symptoms and two or more sMRI scans had non-significant reductions in
whole brain and (left) AHC volume over almost 2 years, but did have significant
reductions in the (right) temporal lobes over time (Lawrie et al., 2002). Very pre-
liminary results suggest that relatively large brains (and relatively small thalami in
women) predict the early onset of psychosis in high-risk subjects (Johnstone et al.,
2002).
At the time of writing, the twentieth person in the Edinburgh High Risk Study
has just developed schizophrenia or a related psychosis. We are currently analyzing
the possible sMRI predictors of schizophrenia and changes as the illness develops,
with both ROI and VBM. Future analyses will include relating pre- and perimorbid
brain structure to other risk factors for schizophrenia.
volume reductions in the hippocampus, entorhinal cortex, and inferior frontal and
fusiform gyri during the transition to psychosis (Pantelis et al., 2000).
Phillips et al. (2002) examined the traced whole brain and hippocampal volumes
as possible predictors of psychosis. Of the eligible referrals, 75% were scanned
between 1995 and 1998; 5% of scans were lost through movement artifact, leaving
60 subjects. Twenty (33%) of those with usable scans developed an acute psy-
chosis (defined as an increase of one or more points in psychotic symptom severity,
several times a week, for more than 1 week) within a year. A structured clinical
interview identified diagnoses of schizophrenia spectrum disorder (11), affective
psychosis (3), bipolar disorder (3), and others (3). The UHR groups who did not
develop psychosis included four patients with depression, and 11 with an anxiety
state.
The scans were conducted at two sites and there was a weak tendency for the scan-
ners to deliver different whole brain volumes and for more of those who developed
psychosis (and of one sex) to be scanned at one site (Phillips et al., 2002). Control-
ling for a slightly smaller whole brain, the 60 UHR patients had significantly smaller
hippocampi (by about 11%; ES, 0.9) than the controls but did not differ from the
FEP group. However, the UHR patients were on average 10 years younger and
11 points lower in premorbid intelligence quotient (IQ) scores than the normals.
The 20 who became psychotic were much more closely similar in demographics to
the 40 who remained non-psychotic but, rather counterintuitively, the (left) hip-
pocampus was larger in those who went on to develop a psychosis. These confusing
results may be attributable to scanner effects, selection bias, and/or confounding,
for example by other diagnoses or gender.
Pantelis et al. (2003) have recently reported on their VBM findings in a slightly
extended sample. In 75 people with prodromal symptoms, defined as above, 23
(31%) developed a psychotic disorder (with roughly equal numbers of schizophre-
nia and affective psychoses). Compared with those who did not develop a psy-
chosis, they had less grey matter in (right) medial temporal, lateral temporal,
and inferior frontal cortex, and in bilateral cingulate cortex. Twenty-one sub-
jects had a repeat scan after 1–2 years, 10 of whom had become psychotic (five
with schizophrenia). They showed reductions over time in (left) parahippocam-
pal, fusiform, orbitofrontal, and cerebellar grey matter, while those who did not
become psychotic only exhibited cerebellar reductions. These are intriguing results
that suggest grey matter reductions in the run up to psychosis and immediately
after it. There are, however, some important issues outstanding; in particular,
whether some of those in the UHR group actually had a psychosis before or at
study entry, and why the ROI and VBM studies appear to have such different
findings.
353 Premorbid structural abnormalities in schizophrenia
Summary
These studies illustrate well the difficulties in conducting this type of research:
acquiring a representative sample who will continue to participate in longitudinal
studies, maintaining technical control of the scanner(s) over lengthy time inter-
vals, and distinguishing psychotic symptoms from psychosis. The two recruitment
strategies used so far have different and complementary strengths. Sampling rela-
tives rather than referrals may provide a population who are more committed to the
research, may be more likely to develop schizophrenia than other psychoses, and
gives the potential to study gene–environment interactions. The main limitation
is the time taken to accumulate enough subjects who develop psychosis, which is
likely to be longer than in studies of people presenting with psychotic symptoms.
The latter studies also have the greater ability to compare diagnoses. Regardless, the
two main studies so far both report reduced medial temporal lobe structures as a
trait marker of schizophrenia and suggest further volume losses around the time
of onset. Such prospective studies are the only way to establish temporal patterns
with any certainty, although very important information can also be obtained from
case-control studies that examine the associations between imaging abnormalities
and known risk factors for the disorder.
Family history
Family history is, of course, a proxy for genetic effects and these might only be
expressed later in development. Given, however, the relative importance of genetic
as opposed to environmental causation of schizophrenia, and that brain growth is
maximal prenatally and for 1–2 years thereafter, it is likely that familial structural
abnormalities are at least partly evident at or shortly after birth. The relationship
between an elevated genetic risk of schizophrenia and structural abnormalities has
been examined by scanning relatives (twins, siblings, offspring, parents), comparing
known familial with presumed sporadic cases, and by examining people with genetic
variants such as schizotypal personality disorder (SPD).
354 S. M. Lawrie
Computed tomography
In the first CT study, Weinberger et al. (1981) compared the VBRs of 10 inpatients
with schizophrenia; 12 of their siblings (from seven sibships), and 17 controls.
The VBR appeared to be, at least partially, under genetic control (as did some
local horn enlargements) and differences were evident: largest in the schizophrenia
patients, then in the siblings, and then the controls. Reveley et al. (1982) obtained
a similar pattern of results in discordant monozygotic (MZ) twins and controls, as
have other sibling studies (DeLisi et al., 1986; Mourot et al., 1997; Silverman et al.,
1998). Mourot et al. (1997) also found the same effect for third ventricle width,
and Honer et al. (1995) reported larger temporal sulci and sylvian fissures (but not
temporal horns) in patients compared with unaffected siblings.
The Copenhagen High Risk Project initially examined 34 offspring of mothers
with schizophrenia, 10 of whose fathers also had a spectrum disorder. Widened
fissures and sulci were related to genetic risk, while an increased VBR appeared
to relate to a gene–environment interaction (see below; Cannon et al., 1989). The
results were internally consistent on substantially enlarging the sample (Cannon
et al., 1993). Widening the sample to include offspring with schizophrenia, SPD,
or other psychiatric disorders demonstrated that global effects were evident in
schizophrenia and SPD but only the schizophrenia patients had enlarged ventricles
(Cannon et al., 1994). However, a subsequent comparison of 16 “discordant sibling
pairs” found that the differences were almost as marked for the sulcal area or the
sulcal to brain ratio as for ventricular area or VBR (Zorrilla et al., 1997).
Studies of the VBR in “sporadic versus familial” schizophrenia provide less con-
sensus, as one of the originators of the distinction noted in a review of the early
studies (Lewis, 1990). Patients without a family history of schizophrenia have been
shown repeatedly to have higher VBRs and more marked ventriculomegaly than
patients with a positive family history (e.g. Vita et al., 1994; reviewed by DeQuardo
et al., 1996). Some notable large studies (n > 100) have failed to find an associ-
ation between family history and the VBR (Johnstone et al., 1989; Jones et al.,
1994), yet several studies do report such an association (e.g. Owens et al., 1985),
including those which go some way to supporting the distinction (e.g. Vita et al.,
1994; Silverman et al., 1998). It may be that schizophrenia patients without an
obvious genetic loading are more likely to have had environmental triggers likely
to increase the VBR (see below). However, determining sporadic status is at best
difficult, when many healthy relatives are likely to be carrying the gene(s), and
the limited resolution of CT increases measurement error. Indeed, sMRI studies
that have adopted similar approaches have all reported similar or greater abnor-
malities in familial cases (Dauphinais et al., 1990; Falkai et al., 2002; Harris et al.,
2002; Roy et al., 1994; Sanderson et al., 2001; Schwarzkopf et al., 1991; Seidman
et al., 2002), with the exception of McDonald et al. (2002), and some suggest
355 Premorbid structural abnormalities in schizophrenia
specific genetic effects in frontal and temporal lobes, the ventricles, and the basal
ganglia.
ventriculomegaly (McDonald et al., 2002; Sharma et al., 1998) has not yet been
externally replicated (Steel et al., 2002). The increased VBR seen by CT in rela-
tives may, therefore, primarily reflect a reduction in brain volume, although the
results on sMRI are equivocal. While some studies find the brain is smaller in rel-
atives than in controls (Cannon et al., 1998b; Keshavan et al., 1997, 2002), and
no patient–relative differences (Cannon et al., 1998b; McDonald et al., 2002; Seid-
man et al., 2002), many find no differences (McDonald et al., 2002; Seidman et al.,
1999, 2002; Sharma et al., 1998; Staal et al., 1998) and one small study did actually
find differences between “obligate carriers” and their affected siblings (Steel et al.,
2002).
The evidence from studies of relatives is also inconclusive for most other brain
regions, in some cases because of insufficient studies and in others because of low
power. There are, for example, isolated reports of abnormal cerebral torque (Sharma
et al., 1999), but the most consistent abnormalities in patients (Table 19.1) have
not been found in relatives (Bartley et al., 1993; Frangou et al., 1997); findings
of abnormal sylvian fissure (Honer et al., 1995) and AHC asymmetry (Schreiber
et al., 1999) have yet to be replicated (Bartley et al., 1993). There is, however,
a fair degree of agreement with the twin and automated studies reviewed above
for frontotemporal differences with a pattern of most effect seen in schizophrenia
patients compared with controls and intermediate effects in relatives. The main
support for this from ROI studies in relatives is a trend from a large study using
sophisticated segmentation algorithms (Cannon et al., 1998b), and from studies
contrasting patients with their unaffected relatives (McDonald et al., 2002; Staal
et al., 2000; Steel et al., 2002). Small (Keshavan et al., 1997, 2002; Schreiber et al.,
1999) and large (Lawrie et al., 2001) studies of high-risk offspring have not found
such differences, and neither have medium-sized studies that did not control for
family membership (Sharma et al., 1998). It appears that the relatively small effects
can also be detected with automated approaches and/or control for between-family
variance.
The importance of statistical power is again illustrated in studies of the third ven-
tricle and the thalamus (the increase in the former probably relating to reductions
in the latter and other surrounding structures). The two studies that did not find
changes in the third ventricle (Schreiber et al., 1999) and the thalamus (Keshavan
et al., 1997) were the two smallest were offspring studies (15 and 11 patients, respect-
ively). Keshavan et al. (2002) found thalamus reductions when they increased the
sample size from 11 to 19. Differences have, however, been found with samples as
small as six (Seidman et al., 1997) and all of the available literature suggests that
third ventricle increases and/or thalamus reductions occur in relatives compared
with controls (Keshavan et al., 1997, 2002; Lawrie et al., 2001; McDonald et al.,
2002; Seidman et al., 1997, 1999; Staal et al., 1998, 2000). Very few studies have
358 S. M. Lawrie
looked at patient–relative differences. Staal et al. (1998) found that the thalamus
was smaller in patients than in their siblings, while thalamus reductions were related
to genetic liability but not psychotic symptoms in the Edinburgh High Risk Study
(Lawrie et al., 2001). Third ventricle volumes are consistently reported as trends in
patient–relative differences (Lawrie et al., 2001; McDonald et al., 2002; Staal et al.,
1998), suggesting that environmental influences on third ventricle increases may
be related to disease expression.
The studies of relatives do make clear that reductions in the AHC or its component
structures are both genetic and phenotypic. In terms of relative differences, patients
are generally found to have smaller AHCs than their unaffected relatives, and rela-
tives are generally found to have smaller AHCs than healthy controls (Table 19.2). In
terms of statistical significance, relatives have smaller AHCs than controls (Keshavan
et al., 1997, 2002; Lawrie et al., 1999, 2001; Schreiber et al., 1999; Seidman et al.,
1997, 1999), and schizophrenia patients have smaller AHCs than relatives (Lawrie
et al., 1999, 2001; O’Driscoll et al., 2001; Steel et al., 2002), although there are some
negative studies (Staal et al., 2000). There are suggestions that the reductions may
be more marked anteriorly (Keshavan et al., 2002; O’Driscoll et al., 2001) and on the
left side (Keshavan et al., 2002; Lawrie et al., 2001; but see Schreiber et al., 1999).
There is, however, good evidence for hippocampal differences as well (Seidman
et al., 2002; Waldo et al., 1994; see also van Erp et al., 2002).
Table 19.2. Region of interest studies of the amygdala–hippocampal complex in patients with
schizophrenia, their relatives and/or controls
MZ, monozygotic; DZ, dizygotic; Hipp., hippocampus; Amyg., amygdala; AHC, amygdala–hippocampal
complex; Ant., anterior; Post., posterior.
360 S. M. Lawrie
by our knowledge of the genes that increase the liability to schizophrenia. That
being said, a small number of studies have directly examined the associations of
some putative genes. With probably the greatest justification, Kunugi et al. (1999)
examined the relationship between hippocampal volume and the gene for neu-
rotrophin 3 (NT-3) (A3 allele) in a small number of patients with schizophrenia
and bipolar disorder. NT-3 A3 allelic status was associated with smaller volumes
only in schizophrenia patients: the absence of an effect in those with bipolar disorder
suggesting a possible interaction with other schizophrenia genes or environmen-
tal risk factors. Any association between hippocampal volume and apolipoprotein
E genotype in schizophrenia is at best unreplicated (Fernandez et al., 1999; Hata
et al., 2002). Finally, increased VBR and enlarged CSF spaces have been found to
be associated with a linkage marker on chromosome 5p in one large pedigree.
This is of note as the imaging findings (presumably reflecting generally reduced
brain volumes) were more strongly associated with the gene than with the disor-
der (Shihabuddin et al., 1996). There is, fortunately, rather stronger evidence for
specific environmental effects: particularly with arguably the best replicated risk
factor, namely OCs.
Obstetric complications
There is actually quite good evidence that people with family histories of schizophre-
nia do not suffer a greater number of or any more severe OCs. Rather, they appear
to be more sensitive to their effects – perhaps because they already have abnormal
brains. There is also an extensive literature on the clinical and imaging abnormalities
associated with OCs in neonates; a wide array of adverse events have the potential to
cause ventriculomegaly, periventricular leucomalacia, and/or hippocampal dam-
age, with a range of neurodevelopmental outcomes. What specificity there is in any
relationship between OCs and brain damage in schizophrenia will, therefore, arise
from gene–environment interactions. It is on this basis that a number of studies
have examined the association between OCs and enlarged ventricles or reduced
hippocampi in schizophrenia patients and related populations.
Ventricles
As with family history, studies of the associations between OCs and CT indices in
schizophrenia have produced inconsistent results. The largest CT studies of patients
tended to find no association (Johnstone et al., 1989; Jones et al., 1994; Owens et al.,
1985). Of 10 studies reviewed by Lewis (1990), five reported an association between
OCs and ventriculomegaly and two of these were studies of relatives. Further,
although Reveley et al. (1982) found an apparent environmental effect on the VBR
in discordant MZ twins and controls, which they attributed to common delivery
361 Premorbid structural abnormalities in schizophrenia
complications with more severe perinatal brain damage in the schizophrenia twin,
they actually found a stronger association between OCs and ventricular size in the
controls (Reveley et al., 1984). DeLisi et al. (1986) also suggested part genetic and
part environmental explanations for ventriculomegaly in their sample and found
such an effect for the frontal horns; however, both OCs and a history of head
injury in childhood were related to ventricular body enlargement, with no effect
attributable to a later diagnosis of schizophrenia.
Data from the Copenhagen High Risk Project strongly suggest a gene–
environment interaction. In the 27 high-risk subjects who were scanned initially and
whose midwife records were available, birth weight was inversely correlated (0.6)
with the VBR. There were weaker correlations with duration of labor and length
at birth (Schulsinger et al., 1984). Those with relatively lower weight (and those
with signs of prematurity) were more likely to have VBRs above the group median,
with stronger correlations in those who had subsequently developed schizophrenia
(0.77) than in those who had not (Silverton et al., 1985). On stepwise regression,
birth weight accounted for 22% of the variance in VBR in the high-risk group, but
64% in the UHR group with schizotypal fathers (Silverton et al., 1988a). There
was no apparent effect of genetic risk alone, maternal illness, or maternal neglect
(Silverton et al., 1988b). More sophisticated analysis revealed that an increased
VBR (and third ventricle width) was particularly related to an interaction between
the level of genetic risk and the severity of delivery complications, with a smaller
effect of low birth weight and no significant effect of total pregnancy complications
(Cannon et al., 1989). Increasing the sample to 157 subjects, with none, one, or two
affected parents, replicated these findings (Cannon et al., 1993).
A history of OCs has also been related to enlarged ventricles on sMRI in
schizophrenia patients (Alvir et al., 1999) and in relatives. Following Suddath et al.
(1990), McNeil et al. (2000) examined an extended sample of 22 discordant MZ
twin pairs and found that those with schizophrenia and large (right lateral and
total) ventricle volumes were more likely to have had labor and neonatal problems,
in addition to a prolonged labor, but no more total adverse events throughout
the pregnancy. More recently, Cannon et al. (2002b) compared 64 patients, 51 of
their unaffected siblings, and 54 controls with obstetric records of hypoxia, pre-
maturity, being small for dates and any perinatal complication. Hypoxic compli-
cations interacted with genetic risk to reduce whole-brain gray matter volumes,
especially in the temporal lobe and in schizophrenia patients, with tendencies
to do so for sulcal and ventricular CSF. Prematurity, small for gestational age
status, and prenatal infection had similar effects but were not associated with
adult schizophrenia and mainly rendered the brain more sensitive to the effects of
hypoxia.
362 S. M. Lawrie
Hippocampi
There is also some good evidence that OCs are related to small hippocampi in
schizophrenia, possibly through gene–environment interaction. A small study of
patients with a positive family history found no differences in limbic areas between
those with and without a history of birth complications (DeLisi et al., 1988), while
Stefanis and colleagues (1999) found smaller (left) hippocampi in patients with a
history of pregnancy and birth complications, but not in patients from multiply
affected families without OCs. McNeil et al. (2000) reported that intrapair dif-
ferences in hippocampal volumes between discordant MZ twins were related to
a prolonged labor (rather than neonatal complications). Using original hospital
records, and focusing on hypoxic events and “blue babies,” van Erp et al. (2002)
found that 72 patients had smaller hippocampi than 58 siblings, who, in turn, had
smaller volumes than healthy controls, but only schizophrenia patients with docu-
mented OCs had smaller hippocampi (siblings with OCs did not). The fact that OC
frequency was equal across the groups argues against gene–environment covaria-
tion, and the relatively small effect size for hypoxia in the probands (0.24) suggests a
sensitivity to the effects of OCs in those destined to develop schizophrenia. Further,
the intraclass correlation coefficients for healthy sibling pairs were higher than for
discordant pairs, suggesting that genetic influences on hippocampal volume are
larger in health than in schizophrenia, and that large genetic variation and/or
unique environmental events influence hippocampal volume in schizophrenia
patients.
Summary
There is fairly persuasive evidence that OCs, particularly labor complications and
proven hypoxia in neonates, interact with genetic risk for schizophrenia to produce
larger lateral ventricles and smaller hippocampi. It may be that a genetically small,
abnormal brain and other OCs increase the susceptibility to these effects. These
effects are likely to be observable at birth, although there is no direct evidence
for this. There are, however, indications that animals exposed experimentally to
hypoxemia in utero have these types of abnormality around term (e.g. Rees et al.,
1999). As during early development the enlarging brain drives intracranial vol-
ume expansion, an early developmental disruption should not be associated with
increases in cortical CSF (Woods, 1998); yet cortical CSF is known to be enlarged in
schizophrenia and appears to be related to genetic effects (see above). The implica-
tion is that a small brain in schizophrenia is, at least in part, attributable to postnatal
events.
correlations with structural measures on both CT and sMRI may, however, suggest
that function is more heavily determined by other factors and/or that there are
structural changes nearer to the time of psychosis. As the evidence for the latter
is increasing, one might expect that likely precipitants for schizophrenia – such as
stressful life events and cannabis consumption – have been examined in relation to
these findings. This does not appear to have happened as yet, at least in relation to
schizophrenia. While there are some studies of the structural effects of cannabis in
non-psychotic subjects, they are far from conclusive. There are a number of studies
in anxiety and depression suggesting that predisposing and precipitating stressors
are associated with reduced volumes of the hippocampus (reviewed by Sapolsky,
2000). Such research is now surely a priority, both to build up a coherent picture
of the pathophysiology of schizophrenia and because it may well have therapeutic
implications.
Conclusions
The results of sMRI studies of twins and of relatives of patients with schizophrenia
strongly suggest that at least some of the neuroanatomical features of the disorder
are present before onset and probably for some time. It is likely that reductions in the
volumes of the whole brain and frontal lobes are primarily genetically mediated and
that this partly accounts for reductions in the amygdala and hippocampus. Increases
in ventricular size and further reductions in the hippocampus are likely to occur
around the time of labor complications. This does not preclude later developmental
changes (some of which could be genetic) or degenerative effects. Relatively little is
known about structural brain development in childhood and adolescence, in health
and schizophrenia, but inference from the degrees of abnormality found in the above
studies suggests that there are further changes in frontal and especially temporal
lobes, perhaps particularly in medial structures, shortly before and possibly after
the onset of psychosis.
More of the same studies can be justified on the grounds that no finding is as
yet conclusive. However, ROI studies should probably have minimum group sizes
of about 60 – unless variance is reduced by recruiting within families – if sub-
tle differences in the brain and ventricles are to be examined, and particularly if
gene–environment interactions are to be assessed. More specific regional effects are
probably best assessed with automated approaches. It should be clear that case–
control and prospective cohort studies of those at elevated risk, for both genetic
and other reasons, are complementary. There may also be important information
gained from studies of early-onset schizophrenia and related neurodevelopmen-
tal disorders. Other analyses of sMRI data, such as calculating the gyral folding
365 Premorbid structural abnormalities in schizophrenia
index, and novel uses of MRI, such as proton spectroscopy and diffusion tensor
imaging, could give additional insights (but have not been described in this chap-
ter as there are as yet only interesting suggestions and no replicated findings in
relatives).
Perhaps the greatest gains in knowledge will arise from more novel approaches.
For example, ultrasound images of fetal and neonatal brains could be used to study
the time course of the effects of genetic and environmental risk factors relevant
to schizophrenia: although studying demonstrable effects rather than even well-
documented risks may be most relevant. Levels of psychosocial stress and cannabis
consumption need to be related to brain structure in subjects at high risk. Most
importantly, specific genetic abnormalities in schizophrenia (once clearly identi-
fied) may have regionally and temporally specific effects. The search for these genes
is, of course, informed by the foregoing studies, and both will be informed by
advances in the knowledge of the genetics and imaging of normal neurodevelop-
ment.
It will take some time before these developments lead to a sophisticated under-
standing of the time course of subregional anatomical differences in schizophrenia,
but it is not too early to begin to think about therapeutic implications. There is good
evidence, for example, that hypothermic treatment of hypoxic brains may reduce
the number of adverse neurodevelopmental outcomes. Once specified, structural
predictors of psychosis would provide a means of identifying subjects at very high
risk of conversion. This might justify early interventions to reduce stress or cannabis
use, and perhaps to prescribe antipsychotic or neuroprotective drugs, which might
ameliorate or even prevent the devastating effects of schizophrenia.
REFERENCES
Alvir, J. M. J., Woerner, M. G., Gunduz, H., Degreef, G., Lieberman, J. A. (1999). Obstretric
complications predict treatment response in first-episode schizophrenia. Psychol Med 29:
621–627.
Ananth, H., Popescu, I., Critchley, H. D., et al. (2002). Cortical and subcortical gray matter
abnormalities in schizophrenia determined through structural magnetic resonance imaging
with optimized volumetric voxel-based morphometry. Am J Psychiatry 159: 1497–1505.
Baare, W. F., van Oel, C. J., Hulshoff Pol, H. E., et al. (2001). Volumes of brain structures in twins
discordant for schizophrenia. Arch Gen Psychiatry 58: 33–40.
Bartley, A. J., Jones, D. W., Torrey, E. F., Zigun, J. R., Weinberger, D. R. (1993). Sylvian fissure
asymmetries in monozygotic twins: a test of laterality in schizophrenia. Biol Psychiatry 34:
853–863.
366 S. M. Lawrie
Bridle, N., Pantelis, C., Wood, S. J., et al. (2002). Thalamic and caudate volumes in monozygotic
twins discordant for schizophrenia. Aust N Z J Psychiatry 36: 347–354.
Buchsbaum, M. S., Yang, S., Hazlett, E., et al. (1997). Ventricular volume and asymmetry in
schizotypal personality disorder and schizophrenia assessed with magnetic resonance imag-
ing. Schizophr Res 27: 45–53.
Cannon, M., Jones, P. (1996). Schizophrenia. J Neurol Neurosurg Psychiatry 61: 604–613.
Cannon, T. D., Mednick, S. A., Parnas, J. (1989). Genetic and perinatal determinants of structural
brain deficits in schizophrenia. Arch Gen Psychiatry 46: 883–889.
Cannon, T. D., Mednick, S. A., Parnas, J., et al. (1993). Developmental brain abnormalities in the
offspring of schizophrenic mothers. I. Contributions of genetic and perinatal factors. Arch
Gen Psychiatry 50: 551–564.
(1994). Developmental brain abnormalities in the offspring of schizophrenic mothers. II.
Structural brain characteristics of schizophrenia and schizotypal personality disorder. Arch
Gen Psychiatry 51: 955–962.
Cannon, T. D., Kaprio, J., Lönnqvist, J., Huttunen, M., Koskenvuo, M. (1998a). The genetic
epidemiology of schizophrenia in a Finnish twin cohort. Arch Gen Psychiatry 55: 67–74.
Cannon, T. D., van Erp, T. G., Huttunen, M., et al. (1998b). Regional gray matter, white matter,
and cerebrospinal fluid distributions in schizophrenic patients, their siblings, and controls.
Arch Gen Psychiatry 55: 1084–1091.
Cannon, T. D., Thompson, P. M., van Erp, T. G., et al. (2002a). Cortex mapping reveals regionally
specific patterns of genetic and disease-specific gray-matter deficits in twins discordant for
schizophrenia. Proc Natl Acad Sci USA 99: 3228–3233.
Cannon, T. D., van Erp, T. G., Rosso, I. M., et al. (2002b). Fetal hypoxia and structural brain
abnormalities in schizophrenic patients, their siblings, and controls. Arch Gen Psychiatry 59:
35–41.
Chemerinski, E., Nopoulos, P. C., Crespo-Facorro, B., Andreasen, N. C., Magnotta, V. (2002).
Morphology of the ventral frontal cortex in schizophrenia: relationship with social dysfunc-
tion. Biol Psychiatry 52: 1–8.
Copolov, D., Velakoulis, D., McGorry, P., et al. (2000). Neurobiological findings in early phase
schizophrenia. Brain Res Rev 31: 157–165.
Dauphinais, I. D., DeLisi, L. E., Crow, T. J., et al. (1990). Reduction in temporal lobe size in siblings
with schizophrenia: a magnetic resonance imaging study. Psychiatry Res 35: 137–147.
DeLisi, L. E., Goldin, L. R., Hamovit, J. R., et al. (1986). A family study of the association of
increased ventricular size with schizophrenia. Arch Gen Psychiatry 43: 148–153.
DeLisi, L. E., Dauphinais, I. D., Gershon, E. S. (1988). Perinatal complications and reduced size
of brain limbic structures in familial schizophrenia. Schizophr Bull 14: 185–191.
DeQuardo, J. R., Goldman, M., Tandon, R. (1996). VBR in schizophrenia: relationship to family
history of psychosis and season of birth. Schizophr Res 20: 275–285.
Dickey, C. C., McCarley, R. W., Voglmaier, M. M., et al. (1999). Schizotypal personality disorder
and MRI abnormalities of temporal lobe gray matter. Biol Psychiatry 45: 1393–1402.
Dickey, C. C., Shenton, M. E., Hirayasu, Y., et al. (2000). Large CSF volume not attributable to
ventricular volume in schizotypal personality disorder. Am J Psychiatry 157: 48–54.
367 Premorbid structural abnormalities in schizophrenia
Downhill, J. E., Buchsbaum, M. S., Hazlett, E. A., et al. (2001). Temporal lobe volume deter-
mined by magnetic resonance imaging in schizotypal personality disorder and schizophre-
nia. Schizophr Res 48: 187–199.
Erel, O., Cannon, T. D., Hollister, J. M., Mednick, S. A., Parnas, J. (1991). Ventricular enlargement
and premorbid deficits in school-occupational attainment in a high risk sample. Schizophr
Res 4: 49–52.
Falkai, P., Honer, W. G., Alfter, D., et al. (2002). The temporal lobe in schizophrenia from uni-
and multiply affected families. Neurosci Lett 325: 25–28.
Fannon, D., Tennakoon, L., Sumich, A., et al. (2000). Third ventricle enlargement and devel-
opmental delay in first-episode psychosis: preliminary findings. Br J Psychiatry 177: 354–
359.
Fernandez, T., Yan, W. L., Hamburger, S., et al. (1999). Apolipoprotine E alleles in childhood-onset
schizophrenia. Am J Med Genet 88: 211–213.
Frangou, S., Sharma, T., Sigmudsson, T., et al. (1997). The Maudsley Family Study 4. Nor-
mal planum temporale asymmetry in familial schizophrenia. Br J Psychiatry 170: 328–
333.
Gur, R. E., Turetsky, B. I., Bilker, W. B., Gur, R. C. (1999). Reduced gray matter volume in
schizophrenia. Arch Gen Psychiatry 56: 905–911.
Gur, R. E., Turetsky, B. I., Cowell, P. E., et al. (2000). Temporolimbic volume reductions in
schizophrenia. Arch Gen Psychiatry 57: 769–775.
Harris, J. G., Young, D. A., Rojas, D. C., et al. (2002). Increased hippocampal volume
in schizophrenics’ parents with ancestral history of schizophrenia. Schizophr Res 55:
11–17.
Hata, T., Kunugi, H., Nanko, S., Fukuda, R., Kaminaga, T. (2002). Possible effect of the APOE
e4 allele on the hippocampal volume and asymmetry in schizophrenia. Am J Med Genet
Neuropsychiatr Genet 114: 641–642.
Honer, W. G., Bassett, A. S., Squires-Wheeler, E., et al. (1995). The temporal lobes, reversed
asymmetry and the genetics of schizophrenia. Neuroreport 7: 221–224.
Hulshoff Pol, H. E., Hoek, H. W., Susser, E., et al. (2000). Prenatal exposure to famine and brain
morphology in schizophrenia. Am J Psychiatry 157: 1170–1172.
Job, D. E., Whalley, H. C., McConnell, S., et al. (2002). Structural gray matter differences between
first-episode schizophrenics and normal controls on voxel-based morphometry. Neuroimage
17: 880–889.
Job, D. E., Whalley, H. C., Glabus, M., Johnstone E. C., Lawrie, S. M. (2003). Voxel-based
morphometry of grey matter reductions in subjects at high risk of schizophrenia. Schizophr
Res 64: 1–13.
Johnstone, E. C., Owens, D. G., Bydder, G. M., et al. (1989). The spectrum of structural brain
changes in schizophrenia: age of onset as a predictor of cognitive and clinical impairments
and their cerebral correlates. Psychol Med 19: 91–103.
Johnstone, E. C., Cosway, R., Lawrie, S. M. (2002). Distinguishing characteristics of subjects
with good and poor early outcome in the Edinburgh High Risk Study. Br J Psychiatry 181:
S26–S29.
368 S. M. Lawrie
Jones, P. B., Harvey, I., Lewis, S. W., et al. (1994). Cerebral ventricle dimensions as risk factors
for schizophrenia and affective psychosis: an epidemiological approach to analysis. Psychol
Med 24: 995–1011.
Keshavan, M. S., Montrose, D. M., Pierri, J. N., et al. (1997). Magnetic resonance imaging and
spectroscopy in offspring at risk for schizophrenia: preliminary studies. Prog Neuropsy-
chopharmacol Biol Psychiatry 21: 1285–1295.
Keshavan, M. S., Dick, E., Mankowski, I., et al. (2002). Decreased left amygddala and hippocampal
volumes in young offspring at risk for schizophrenia. Schizophr Res 58: 173–183.
Konick, L. C., Friedman, L. (2001). Meta-analysis of thalamic size in schizophrenia. Biol Psychiatry
49: 28–38.
Kunugi, H., Hattori, M., Nanko, S., et al. (1999). Dinucleotide repeat polymorphism in the
neurotrophin-3 gene and hippocampal volume in psychoses. Schizophr Res 37: 271–
273.
Lawrie, S. M., Abukmeil, S. S. (1998). Brain abnormality in schizophrenia. A systematic and
quantitative review of volumetric magnetic resonance imaging studies. Br J Psychiatry 172:
110–120.
Lawrie, S. M., Whalley, H., Kestelman, J. N., et al. (1999). Magnetic resonance imaging of brain
in people at high risk of developing schizophrenia. Lancet 353: 30–33.
Lawrie, S. M., Adams, C. E., Thornley, B., Joy, C. (2000). Comprehensiveness of systematic review:
update. Br J Psychiatry 176: 396–401.
Lawrie, S. M., Whalley, H. C., Abukmeil, S. S., et al. (2001). Brain structure, genetic liability and
psychotic symptoms in subjects at high risk of developing schizophrenia. Biol Psychiatry 49:
811–823.
(2002). Temporal lobe volume changes in subjects at high risk of schizophrenia with psychotic
symptoms. Br J Psychiatry 181: 138–143.
Levitt, J. J., McCarley, R. W., Dickey, C. C., et al. (2002). MRI study of caudate nucleus volume and
its cognitive correlates in neuroleptic-naive patients with schizotypal personality disorder.
Am J Psychiatry 159: 1190–1197.
Lewis, S. W. (1990). Computerised tomography in schizophrenia 15 years on. Br J Psychiatry 157:
16–24.
Lim, K. O., Beal, D. M., Harvey, Jr., R. L., et al. (1995). Brain dysmorphology in adults with
congenital rubella plus schizophrenia-like symptoms. Biol Psychiatry 37: 764–776.
McDonald, C., Grech, A., Toulopoulou, T., et al. (2002). Brain volumes in familial and non-familial
schizophrenic probands and their unaffected relatives. Am J Med Genet (Neuropsychiatric
Genetics) 114: 616–625.
McGuffin, P., Asherson, P., Owen, M., Farmer, A. (1994). The strength of the genetic effect. Is
there room for an environmental influence in the aetiology of schizophrenia? Br J Psychiatry
164: 593–599.
McNeil, T. F., Cantor-Graae, E., Weinberger, D. R. (2000). Relationship of obstetric complica-
tions and differences in size of brain structures in monozygotic twin pairs discordant for
schizophrenia. Am J Psychiatry 157: 203–212.
369 Premorbid structural abnormalities in schizophrenia
Meyer-Lindenberg, A., Japee, S., Verchinski, B., et al. (2001). Structural MRI abnormalities in
schizophrenic patients and their unaffected siblings: voxel-based morphometry. Neuroimage
13: S217.
Mourot, A., d’Amato, T., Rochet, T., et al. (1997). Cerebral investigation of healthy siblings of
schizophrenics. Eur Psychiatry 12: 273–278.
Narr, K. L., van Erp, T. G., Cannon, T. D., et al. (2002). A twin study of genetic contributions to
hippocampal morphology in schizophrenia. Neurobiol Dis 11: 83–95.
Nelson, M. D., Saykin, A. J., Flashman, L. A., Riordan, H. J. (1998). Hippocampal volume reduc-
tion in schizophrenia as assessed by magnetic resonance imaging: a meta-analytic study.
Arch Gen Psychiatry 55: 433–440.
Noga, J. T., Bartley, A. J., Jones, D. W., Torrey, E. F., Weinberger, D. R. (1996). Cortical gyral
anatomy and gross brain dimensions in monozygotic twins discordant for schizophrenia.
Schizophr Res 22: 27–40.
O’Driscoll, G. A., Florencio, P. S., Gagnon, D., et al. (2001). Amygdala–hippocampal volume and
verbal memory in first-degree relatives of schizophrenic patients. Psychiatry Res Neuroimag-
ing 107: 75–85.
Owens, D. G., Johnstone, E. C., Crow, T. J., et al. (1985). Lateral ventricular size in schizophrenia:
relationship to the disease process and its clinical manifestations. Psychol Med 15: 27–41.
Paillere-Martinot, M., Caclin, A., Artiges, E., et al. (2001). Cerebral gray and white matter reduc-
tions and clinical correlates in patients with early onset schizophrenia. Schizophr Res 50:
19–26.
Pantelis, C., Velakoulis, D., Suckling, J., et al. (2000). Left medial temporal volume reduction
occurs during the transition from high-risk to first-episode psychosis. Schizophr Res 41: 35.
Pantelis, C., Velakoulis, D., McGorry, P. D., et al. (2003). Neuroanatomical abnormalities before
and after onset of psychosis: a cross-sectional and longitudinal MRI comparison. Lancet
361: 281–288.
Phillips, L. J., Velakoulis, D., Pantelis, C., et al. (2002). Non-reduction in hippocampal volume is
associated with higher risk of psychosis. Schizophr Res 58: 145–158.
Raz, S., Raz, N. (1990). Structural brain abnormalities in the major psychoses: a quantitative
review of the evidence from computerized imaging. Psychol Bull 108: 93–108.
Rees, S., Breen, S., Loeliger, M., McCrabb, G., Harding, R. (1999). Hypoxemia near mid-gestation
has long-term effects on fetal brain development. J Neuropathol Exp Neurol 58: 932–945.
Reveley, A. M., Reveley, M. A., Clifford, C. A., Murray, R. M. (1982). Cerebral ventricular size in
twins discordant for schizophrenia. Lancet i: 540–541.
Reveley, A. M., Reveley, M. A., Murray, R. M. (1984). Cerebral ventricular enlargement in non-
genetic schizophrenia: a controlled twin study. Br J Psychiatry 144: 89–93.
Roy, M. A., Flaum, M. A., Arndt, S. V., Crowe, R. R., Andreasen, N. C. (1994). Magnetic resonance
imaging in familial versus sporadic cases of schizophrenia. Psychiatry Res 54: 25–36.
Sacchetti, E., Calzeroni, A., Vita, A., et al. (1992). The brain damage hypothesis of the season-
ality of births in schizophrenia and major affective disorders: evidence from computerised
tomography. Br J Psychiatry 160: 390–397.
370 S. M. Lawrie
Sanderson, T. L., Doody, G. A., Best, J., Owens, D. G. C., Johnstone, E. C. (2001). Correla-
tions between clinical and historical variables and cerebral structural variables in peo-
ple with mild intellectual disability and schizophrenia. J Intellect Disabil Res 45: 89–
98.
Sapolsky, R. M. (2000). Glucocorticoids and hippocampal atrophy in neuropsychiatric disorders.
Arch Gen Psychiatry 57: 925–935.
Schreiber, H., Baur-Seack, K., Kornhuber, H. H., et al. (1999). Brain morphology in adolescents
at genetic risk for schizophrenia assessed by qualitative and quantitative magnetic resonance
imaging. Schizophr Res 40: 81–84.
Schulsinger, F., Parnas, J., Petersen, E. T., et al. (1984). Cerebral ventricular size in the offspring
of schizophrenic mothers. Arch Gen Psychiatry 41: 602–606.
Schwarzkopf, S. B., Nasrallah, H. A., Olson, S. C., Bogerts, B., McLaughlin, J. A., Mitra, T. (1991).
Family history and brain morphology in schizophrenia: an MRI study. Psychiatry Res 40:
49–60.
Seidman, L. J., Faraone, S. V., Goldstein, J. M., et al. (1997). Reduced subcortical brain volumes in
nonpsychotic siblings of schizophrenic patients: a pilot magnetic resonance imaging study.
Am J Med Genet 74: 507–514.
(1999). Thalamic and amygdala-hippocampal volume reductions in first-degree relatives of
patients with schizophrenia: an MRI-based morphometric analysis. Biol Psychiatry 46: 941–
954.
(2002). Left hippocampal volume as a vulnerability indicator for schizophrenia. Arch Gen
Psychiatry 59: 839–849.
Shapleske, J., Rossell, S. L., Woodruff, P. W. R., David, A. S. (1999). The planum temporale: a
systematic, quantitative review of its structural, functional and clinical significance. Brain
Res Rev 29: 26–49.
Sharma, T., Lancaster, E., Lee, D., et al. (1998). Brain changes in schizophrenia. Volumetric MRI
study of families multiply affected with schizophrenia: the Maudsley Family Study 5. Br J
Psychiatry 173: 132–138.
Sharma, T., Lancaster, E., Sigmundsson, T., et al. (1999). Lack of normal pattern of cerebral
asymmetry in familial schizophrenic patients and their relatives: The Maudsley Family
Study. Schizophr Res 40: 111–120.
Shihabuddin, L., Silverman, J. M., Buchsbaum, M. S., et al. (1996). Ventricular enlargement
associated with linkage marker for schizophrenia-related disorders in one pedigree. Mol
Psychiatry 1: 215–222.
Siever, L. J., Rotter, M., Losonczy, M., et al. (1995). Lateral ventricular enlargement in schizotypal
personality disorder. Psychiatry Res 57: 109–118.
Sigmundsson, T., Suckling, J., Maier, M., et al. (2001). Structural abnormalities in frontal, tem-
poral, and limbic regions and interconnecting white matter tracts in schizophrenic patients
with prominent negative symptoms. Am J Psychiatry 158: 234–243.
Silverman, J. M., Smith, C. J., Guo, S. L., et al. (1998). Lateral ventricular enlargement in
schizophrenic probands and their siblings with schizophrenia-related disorders. Biol Psy-
chiatry 43: 97–106.
371 Premorbid structural abnormalities in schizophrenia
Silverton, L., Finello, K. M., Schulsinger, F., Mednick, S. A. (1985). Low birth weight and ven-
tricular enlargement in a high-risk sample. J Abnorm Psychol 94: 405–409.
Silverton, L., Mednick, S. A., Schulsinger, F., Parnas, J., Harrington, M. E. (1988a). Genetic risk
for schizophrenia, birthweight, and cerebral ventricular enlargement. J Abnorm Psychol 97:
496–498.
Silverton, L., Mednick, S. A., Harrington, M. E. (1988b). Birthweight, schizophrenia and ventric-
ular enlargement in a high-risk sample. Psychiatry 51: 272–280.
Sommer, I., Aleman, A., Ramsey, N., Bouma, A., Kahn, R. (2001). Handedness, language later-
alisation and anatomical asymmetry in schizophrenia. Meta-analysis. Br J Psychiatry 178:
344–351.
Staal, W. G., Hulshoff, Pol, H. E., Schnack, H., van der Schot, A. C., Kahn, R. S. (1998). Partial
volume decrease of the thalamus in relatives of patients with schizophrenia. Am J Psychiatry
155: 1784–1786.
Staal, W. G., Hulshoff Pol, H. E., Schnack, H. G., et al. (2000). Structural brain abnormalities in
patients with schizophrenia and their healthy siblings. Am J Psychiatry 157: 416–421.
Steel, R., Whalley, H., Miller, P., et al. (2002). Structural MRI of the brain in presumed carriers of
genes for schizophrenia, their affected and unaffected siblings. J Neurol Neurosurg Psychiatry
72: 455–458.
Stefanis, N., Frangou, S., Yakeley, J., et al. (1999). Hippocampal volume reduction in schizophre-
nia: effects of genetic risk and pregnancy and birth complications. Biol Psychiatry 46: 697–
702.
Suddath, R. L., Christison, G. W., Torrey, E. F., Casanova, M. F., Weinberger, D. R. (1990).
Anatomical abnormalities in the brains of monozygotic twins discordant for schizophrenia.
N Engl J Med 322: 789–794.
van Erp, T. G. M., Saleh, P. A., Rosso, I. M., et al. (2002). Contributions of genetic risk and
fetal hypoxia to hippocampal volume in patients with schizophrenia or schizoaffective dis-
order, their unaffected siblings, and healthy unrelated volunteers. Am J Psychiatry 159:
1514–1520.
van Horn, J. D., McManus, I. C. (1992). Ventricular enlargement in schizophrenia: a meta-analysis
of studies of the ventricle: brain ratio (VBR). Br J Psychiatry 160: 687–697.
Vita, A., Dieci, M., Giobbio, G. M., et al. (1994). A reconsideration of the relationship between
cerebral structural abnormalities and family history of schizophrenia. Psychiatry Res 53:
41–55.
Waldo, M. C., Cawthra, E., Adler, L. E., et al. (1994). Auditory sensory gating, hippocampal
volume, and catecholamine metabolism in schizophrenics and their siblings. Schizophr Res
12: 93–106.
Walker, E. F., Lewine, R. R., Neumann, C. (1996). Childhood behavioral characteristics and adult
brain morphology in schizophrenia. Schizophr Res 22: 93–101.
Ward, K. E., Friedman, L., Wise, A., Schulz, S. C. (1996). Meta-analysis of brain and cranial size
in schizophrenia. Schizophr Res 22: 197–213.
Weinberger, D. R., DeLisi, L. E., Neophytides, A. N., Wyatt, R. J. (1981). Familial aspects of CT
scan abnormalities in chronic schizophrenic patients. Psychiatry Res 4: 65–71.
372 S. M. Lawrie
Woodruff, P. W. R., McManus, I. C., David, A. S. (1995). Meta-analysis of corpus callosum size
in schizophrenia. J Neurol Neurosurg Psychiatry 58: 457–461.
Woods, B. T. (1998). Is schizophrenia a progressive neurodevelopmental disorder? Toward a
unitary pathogenetic mechanism. Am J Psychiatry 155: 1661–1670.
Wright, I. C., Rabe-Hesketh, S., Woodruff, P. W., et al. (2000). Meta-analysis of regional brain
volumes in schizophrenia. Am J Psychiatry 157: 16–25.
Zorrilla, L. T. E., Cannon, T. D., Kronenberg, S., et al. (1997). Structural brain abnormalities in
schizophrenia: a family study. Biol Psychiatry 42: 1080–1986.
20
Neurodegenerative models of
schizophrenia
L. Fredrik Jarskog, John H. Gilmore, and Jeffrey A. Lieberman
University of North Carolina School of Medicine, Chapel Hill, USA
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
373
374 L. F. Jarskog, J. H. Gilmore, and J. A. Lieberman
the onset of psychosis and then reaches a plateau of stability (McGlashan, 1988).
A more recent study suggests that the deterioration is confined to the first year of
illness: Mason et al. (1996) found that social adjustment deteriorated in the first
year of illness, though remained relatively stable over the next 13 years.
While there is general agreement that overall functioning declines in schizophre-
nia, it is not clear what factors contribute to this functional deterioration. The most
parsimonious explanation for this deterioration may be that it is a consequence of
the symptoms of schizophrenia itself: positive and negative symptoms, and cogni-
tive dysfunction. Untreated or partially treated positive symptoms would obviously
interfere with overall functioning and quality of life, and degeneration or progres-
sion of illness would not necessarily be involved. Both Eaton et al. (1995) and
Mason et al. (1996) found that positive symptoms tend to remain stable after the
first year of illness. There is evidence that primary negative symptoms or the deficit
syndrome does progress in the early years of the illness. In a retrospective chart
review study, Fenton and McGlashan (1994) found that primary negative symp-
toms progressed in severity over the first 5 years of the illness and that the deficit
syndrome was associated with poor outcome and long-term disability. Mayerhoff
et al. (1994) found that first-episode patients had lower rates of the deficit syn-
drome than more chronic populations, and Eaton et al. (1995) found that negative
symptoms were stable after the first 2 years of illness. These studies suggest that
progression of negative symptoms may be limited to the first few years of ill-
ness, though clarification of this issue awaits additional prospective longitudinal
studies.
While initial studies reported progressive cognitive impairment in schizophrenia
(Abrahamson, 1983; Bilder et al., 1992; Smith, 1964), most of the more recent studies
do not indicate a progression of cognitive dysfunction (reviewed by Rund, 1998).
While cognitive decline has been described in geriatric, chronically institutionalized
patients (Friedman et al., 2002; Harvey et al., 1999), a study of older outpatients
found no differences in age-related cognitive decline (Zorrilla et al., 2000). A more
recent longitudinal study of neuropsychological deficits in older patients (mean age
47.6 years) with schizophrenia found them to be stable (Heaton et al., 2001). These
studies suggest that, if progressive deterioration of cognitive functioning occurs
in older patients with schizophrenia, this deterioration is limited to a subset of
very severely ill, chronically institutionalized patients. Cross-sectional comparisons
of cognitive functioning in patients with first-episode and chronic schizophrenia
have found similar degrees of impairment, suggesting that cognitive deficits do not
progress (Addington and Addington, 2002; Moritz et al., 2002; Saykin et al., 1994).
Longitudinal studies in patients with first-episode schizophrenia found either no
progression (Hoff et al., 1999) or even improvement of cognitive impairments
(Nopoulos et al., 1994).
376 L. F. Jarskog, J. H. Gilmore, and J. A. Lieberman
Postmortem neuropathology
During the twentieth century, investigations of the postmortem neuropathology
of schizophrenia have been most notable for demonstrating the absence of a clear
377 Neurodegenerative models of schizophrenia
- shorter dendrites
- longer dendrites - less complex branching
- more complex branching
Fig. 20.2. Model of limited neurodegeneration on structural and histological levels. Longitudinal
neuroimaging studies have demonstrated progressive cortical gray matter loss and ven-
tricular enlargement in schizophrenia, especially in childhood-onset and adult new-onset
populations. The limited neurodegeneration model proposes that cytopathological corre-
lates of gray matter loss include progressive neuronal atrophy and neuropil loss.
Neuroimaging studies
Structural studies
Cross-sectional structural neuroimaging studies have demonstrated a number of
consistent brain abnormalities in schizophrenia primarily involving enlargement of
the lateral and third ventricles and reduction in cortical gray matter volume, both
globally (Gur et al., 1998; Hulshoff Pol et al., 2002; Zipursky et al., 1992) and in
cortical subregions including prefrontal (Gur et al., 1998; Hulshoff Pol et al., 2002),
temporal (Gur et al., 1998; Schlaepfer et al., 1994), and parietal (Schlaepfer et al.,
1994) areas. Since neuropil is a primary component of gray matter, these structural
imaging findings provide in vivo support for the postmortem findings of reduced
cortical neuropil (Selemon et al., 1995, 1998).
One of the limitations of cross-sectional studies is that they do not adequately
address whether a change in brain volume is active or antecedent to the scan. For this
reason, a number of longitudinal structural magnetic resonance imaging (sMRI)
studies have been conducted to address this issue. Interestingly, progressive brain
volume changes have been identified in several different patient cohorts, includ-
ing in patients with prodromal signs of psychosis, in childhood-onset schizophre-
nia, in new-onset schizophrenia, and in certain chronic patient populations. In a
single small study of prodromal patients judged at high risk of transition to psy-
chosis, a progressive loss of gray matter was found in certain hippocampal and
frontal regions only in those patients that developed psychosis (Pantelis et al.,
2002). In childhood-onset disease, several progressive volume changes have been
reported compared with normal controls, including ventricular enlargement
(Rapoport et al., 1997), cortical gray matter loss in frontal and temporal areas
(Jacobsen et al., 1998; Rapoport et al., 1999), and cerebellar volume loss (Keller
et al., 2003). In new-onset schizophrenia, progressive ventricular enlargement has
been identified by several groups (Cahn et al., 2002; DeLisi et al., 1997; Lieber-
man et al., 2001a) but not others (Gur et al., 1998). Progressive volume loss has
been reported in first-episode schizophrenia in cerebral hemispheres bilaterally
(DeLisi et al., 1997), total cerebral gray matter (Cahn et al., 2002), frontal cortex
(Gur et al., 1998), and in superior temporal gyrus (Kasai et al., 2003), while Lieber-
man et al. (2001a) did not detect cortical volume differences. In adults with chronic
schizophrenia, progressive ventricular enlargement has been reported in male
patients (Mathalon et al., 2001) and in patients with poor outcome (Davis et al.,
1998), also suggested by Nair et al. (1997). Mathalon et al. (2001) also reported
accelerated progression of frontotemporal cortical gray matter loss in males with
schizophrenia.
Although the interpretation that progressive brain changes reflect a neurodegen-
erative process in schizophrenia has been challenged (Weinberger and McClure,
380 L. F. Jarskog, J. H. Gilmore, and J. A. Lieberman
2002), the increasing number of studies that have identified accelerated corti-
cal tissue loss and ventricular enlargement suggest that the pathophysiology of
schizophrenia is a dynamic and progressive process, particularly around the onset
of psychosis (Lewis and Lieberman, 2000; Fig. 20.2). Longitudinal studies that
employ higher-resolution neuroimaging techniques should help to define this pro-
cess more precisely.
Conclusions
Although the etiology of schizophrenia remains unknown, the current review
highlights the complexity of its underlying pathophysiology, appearing to involve
both neurodevelopmental and neurodegenerative elements. The histopathology
of schizophrenia remains without a defined phenotype; however, increasing evi-
dence is pointing to subtle neuronal atrophy and synaptodendritic reductions in
postmortem cortex, which may reflect a limited neurodegenerative process. This
conclusion is increasingly supported by neuroimaging studies that find progres-
sive neurostructural changes, especially in gray matter content and ventricle size,
and, less convincingly, studies that report limited progression of clinical symptoms
and neurocognitive function. Taken as a whole, progressive clinical, neurocog-
nitive, and neuroimaging findings appear most evident in the prodromal phase
and the first few years of psychosis, although certain subgroups of chronically ill
383 Neurodegenerative models of schizophrenia
Acknowledgements
This work was supported in part by the UNC Schizophrenia Research Center,
an NIMH Silvio O. Conte Center for the Neuroscience of Mental Disorders
(MH064065, JAL), NIMH grant K08 MH-01752 (LFJ), NIMH grant MH-60352
(JHG), and the Foundation of Hope of Raleigh, North Carolina (JAL).
REFERENCES
Arango, C., Kirkpatrick, B., Koenig, J. (2001). At issue: stress, hippocampal neuronal turnover,
and neuropsychiatric disorders. Schizophr Bull 27: 477–480.
Arnold, S. E., Trojanowski, J. Q., Gur, R. E., et al. (1998). Absence of neurodegeneration and
neural injury in the cerebral cortex in a sample of elderly patients with schizophrenia. Arch
Gen Psychiatry 55: 225–232.
Benes, F. M. (1995). Is there a neuroanatomic basis for schizophrenia? An old question revisited.
Neuroscientist 1: 104–115.
Benes, F. M., McSparren, J., Bird, E. D., SanGiovanni, J. P., Vincent, S. L. (1991). Deficits in
small interneurons in prefrontal and cingulate cortices of schizophrenic and schizoaffective
patients. Arch Gen Psychiatry 48: 996–1001.
Benes, F. M., Vincent, S. L., Todtenkopf, M. (2001). The density of pyramidal and nonpyramidal
neurons in anterior cingulate cortex of schizophrenic and bipolar subjects. Biol Psychiatry
50: 395–406.
Benes, F. M., Walsh, J., Bhattacharyya, S., Sheth, A., Berretta, S. (2003). DNA fragmentation
is decreased in schizophrenia but not in bipolar disorder. Arch Gen Psychiatry 60: 359–
364.
Bertolino, A., Callicott, J. H., Nawroz, S., et al. (1998). Reproducibility of proton magnetic
resonance spectroscopic imaging in patients with schizophrenia. Neuropsychopharmacology
18: 1–9.
Bilder, R. M., Lipschutz-Broch, R. G., Geisler, S. H., Mayerhoff, D. I., Lieberman, J. A. (1992).
Intellectual deficits in first-episode schizophrenia: evidence for progressive deterioration.
Schizophr Bull 18: 437–488.
Black, J. E., Klintsova, A. Y., Kodish, I. M., Greenough, W. T., Uranova, N. A. (2002). What is
missing in the reduced neuropil of schizophrenic prefrontal cortex? A combined golgi and
electron microscopy study. Biol Psychiatry 51: S54.
Cahn, W., Hulshoff Pol, H. E., Lems, E. B. T. E., et al. (2002). Brain volume changes in first-episode
schizophrenia. Arch Gen Psychiatry 59: 1002–1010.
Cannon, M., Jones, P., Huttunen, M. O., et al. (1999). School performance in Finnish children
and later development of schizophrenia. Arch Gen Psychiatry 56: 457–463.
Cannon, M., Caspi, A., Moffitt, T. E., et al. (2002). Evidence for early-childhood, pan-
developmental impairment specific to schizophreniform disorder. Results from a longi-
tudinal birth cohort. Arch Gen Psychiatry 59: 449–456.
Cecil, K. M., Lenkinski, R. E., Gur, R. E., Gur, R. C. (1999). Proton magnetic resonance spec-
troscopy in the frontal and temporal lobes of neuroleptic naive patients with schizophrenia.
Neuropsychopharmacology 20: 131–140.
Church, S. M., Cotter, D., Bramon, E., Murray, R. M. (2002). Does schizophrenia result from
developmental or degenerative processes? J Neural Transm Suppl 63: 129–147.
Cotter, D. R., Pariante, C. M., Everall, I. P. (2001). Glial cell abnormalities in major psychiatric
disorders: the evidence and implications. Brain Res Bull 55: 585–595.
Coyle, J. T., Puttfarcken, P. (1993). Oxidative stress, glutamate, and neurodegenerative disorders.
Science 262: 689–695.
Davis, K. L., Buchsbaum, M. S., Shihabuddin, L., et al. (1998). Ventricular enlargement in poor-
outcome schizophrenia. Biol Psychiatry 43: 783–793.
385 Neurodegenerative models of schizophrenia
DeLisi, L. E., Sakuma, M., Tew, W., et al. (1997). Schizophrenia as a chronic active brain process:
a study of progressive brain structural change subsequent to the onset of schizophrenia.
Psychiatry Res Neuroimaging 74: 129–140.
Done, D. J., Crow, T. J., Johnstone, E. C., Sacker, A. (1994). Childhood antecedents of schizophre-
nia and affective illness: social adjustment at ages 7 and 11. Br Med J 309: 699–703.
Eastwood, S. L., Harrison, P. J. (1995). Decreased synaptophysin in the medial temporal lobe
in schizophrenia demonstrated using immunoautoradiography. Neuroscience 69: 339–
343.
Eaton, W. W., Thara, R., Federman, B., Melton, B., Liang, K.-Y. (1995). Structure and course of
positive and negative symptoms in schizophrenia, Arch Gen Psychiatry 52: 127–134.
Fannon, D., Chitnis, X., Doku, V., et al. (2000). Features of structural brain abnormality in
first-episode psychosis. Am J Psychiatry 157: 1829–1834.
Fenton, W. S., McGlashan, T. H. (1994). Antecedents, symptom progression, and long-term
outcome of the deficit syndrome in schizophrenia. Am J Psychiatry 151: 351–356.
Friedman, J. I., Harvey, P. D., McGurk, S. R., et al. (2002). Correlates of change in functional
status of institutionalized geriatric schizophrenic patients: focus on medical comorbidity.
Am J Psychiatry 159: 1388–1394.
Fuller, R., Nopoulos, P., Arndt, S., et al. (2002). Longitudinal assessment of premorbid cognitive
functioning in patients with schizophrenia through examination of standardized scholastic
test performance. Am J Psychiatry 159: 1183–1189.
Garey, L. J., Ong, W. Y., Patel, T. S., Kanani, M., Davis, A., Mortimer, A. M., Barnes, T. R. E.,
Hirsch, S. R. (1998). Reduced dendritic spine density on cerebral cortical pyramidal neurons
in schizophrenia. J Neurol Neurosurg Psychiatry 65: 446–453.
Glantz, L. A., Lewis, D. A. (1997). Reduction of synaptophysin immunoreactivity in the prefrontal
cortex of subjects with schizophrenia: regional and diagnostic specificity. Arch Gen Psychiatry
54: 943–952.
(2000). Decreased dendritic spine density on prefrontal cortical pyramidal neurons in
schizophrenia. Arch Gen Psychiatry 57: 65–73.
Gur, R. E., Cowell, P., Turetsky, B. I., et al. (1998). A follow-up magnetic resonance imaging study
of schizophrenia. Relationship of neuroanatomical changes to clinical and neurobehavioral
measures. Arch Gen Psychiatry 55: 145–152.
Hambrecht, M., Lammertink, M., Klosterkötter, J., Matuschek, E., Pukrop, R. (2002). Subjec-
tive and objective neuropsychological abnormalities in a psychosis prodrome clinic. Br J
Psychiatry 181(Suppl. 43): S30–S37.
Hardy, J., Gwinn-Hardy, K. (1998). Genetic classification of primary neurodegenerative disease.
Science 282: 1075–1079.
Harrison, P. J. (1999). The neuropathology of schizophrenia: a critical review of the data and
their interpretation. Brain 122: 593–624.
Harrison, P. J., Eastwood, S. L. (2001). Neuropathological studies of synaptic connectivity in the
hippocampal formation in schizophrenia. Hippocampus 11: 508–519.
Harvey, P. D., Silverman, J. M., Mohs, R. C., et al. (1999). Cognitive decline in late-life schizophre-
nia: a longitudinal study of geriatric chronically hospitalized patients. Biol Psychiatry 45:
32–40.
386 L. F. Jarskog, J. H. Gilmore, and J. A. Lieberman
Heaton, R. K., Gladsjo, J. A., Palmer, B. W., et al. (2001). Stability and course of neuropsychological
deficits in schizophrenia. Am J Psychiatry 58: 24–32.
Hinsberger, A. D., Williamson, P. C., Carr, T. J., Stanley, J. A., Drost, D. J., Densmore, M., MacFabe,
G. C., Montemurro, D. G. (1997). Magnetic resonance imaging volumetric and phosphorus
31 magnetic resonance spectroscopy measurements in schizophrenia. J Psychiatry Neurosci
22: 111–117.
Ho, B.-C., Alicata, D., Ward, J., et al. (2003). Untreated initial psychosis: relation to cognitive
deficits and brain morphology in first-episode schizophrenia. Am J Psychiatry 160: 142–
148.
Hoff, A. L., Sakuma, M., Wieneke, M., et al. (1999). Longitudinal neuropsychological follow-up
study of patients with first-episode schizophrenia. Am J Psychiatry 156: 1336–1341.
Hoff, A. L., Sakuma, M., Razi, K., et al. (2000). Lack of association between duration of untreated
illness and severity of cognitive and structural brain deficits at the first episode of schizophre-
nia. Am J Psychiatry 157: 1824–1828.
Huber, G., Gross, G., Shuttler, R., Linz, M. (1980). Longitudinal studies of schizophrenic patients.
Schizophr Bull 6: 592–605.
Hulshoff Pol, H. E., Schnack, H. G., Bertens, M. G. B. C., et al. (2002). Volume changes in gray
matter in patients with schizophrenia. Am J Psychiatry 159: 244–250.
Jacobsen, L. K., Giedd, J., Castellanos, F. X., et al. (1998). Progressive reduction of temporal lobe
structures in childhood-onset schizophrenia. Am J Psychiatry 155: 678–685.
Jarskog, L. F., Gilmore, J. H., Selinger, E. S., Lieberman, J. A. (2000). Cortical Bcl-2 protein
expression and apoptotic regulation in schizophrenia. Biol Psychiatry 48: 641–650.
Jarskog, L. F., Selinger, E. S., Lieberman, J. A., Gilmore, J. H. (2001). Apoptotic regulatory proteins
afford reduced neuroprotection in schizophrenia. Schizophr Res 49: 53S.
Jones, P., Rodgers, B., Murray, R., Marmot, M. (1994). Child development risk factors for adult
schizophrenia in the British 1946 birth cohort. Lancet 344: 1398–1402.
Karson, C. N., Mrak, R. E., Schluterman, K. O., et al. (1999). Alterations in synaptic proteins
and their encoding mRNAs in prefrontal cortex in schizophrenia: a possible neurochemical
basis for “hypofrontality”. Mol Psychiatry 4: 39–45.
Kasai, K., Shenton, M. E., Salisbury, D. F., et al. (2003). Progressive decrease of left superior tem-
poral gyrus gray matter volume in patients with first-episode schizophrenia. Am J Psychiatry
160: 156–164.
Keller, A., Castellanos, F. X., Vaituzis, A. C., et al. (2003). Progressive loss of cerebellar volume in
childhood-onset schizophrenia. Am J Psychiatry 160: 128–133.
Kraeplin E. (1919). Dementia Praecox and Paraphrenia, Edinburgh, E and S Livingstone.
Kremen, W. S., Seidman, L. J., Pepple, J. R., et al. (1994). Neuropsychological risk indicators for
schizophrenia: a review of family studies. Schizophr Bull 20: 103–119.
Lewis, D. A., Lieberman, J. A. (2000). Catching up on schizophrenia: natural history and neuro-
biology. Neuron 28: 325–334.
Lieberman, J. A., Sheitman, B. B., Kinon, B. J. (1997). Neurochemical sensitization in the patho-
physiology of schizophrenia: deficits and dysfunction in neuronal regulation and plasticity.
Neuropsychopharmacology 17: 205–229.
387 Neurodegenerative models of schizophrenia
Lieberman. J. A., Chakos, M., Wu, H., et al. (2001a). Longitudinal study of brain morphology in
first episode schizophrenia. Biol Psychiatry 49: 487–499.
Lieberman, J. A., Perkins, D., Belger, A., et al. (2001b). The early stages of schizophrenia: spec-
ulations on pathogenesis, pathophysiology, and therapeutic approaches. Biol Psychiatry 50:
884–897.
Loebel, A. D., Lieberman, J. A., Alvir, J. M. J., et al. (1992). Duration of psychosis and outcome
in first-episode schizophrenia. Am J Psychiatry 149: 1183–1188.
Margolis, R. L., Chuang, D. M., Post, R. M. (1994). Programmed cell death: implications for
neuropsychiatric disorders. Biol Psychiatry 35: 946–956.
Mason, P., Harrison, G., Glazebrook, C., Medley, I., Croudance, T. (1996). The course of
schizophrenia over 13 years. A report from the International Study on Schizophrenia (IsoS)
coordinated by the World Health Organization. Br J Psychiatry 169: 580–586.
Mathalon, D. H., Sullivan, E. V., Lim, K. O., Pfefferbaum, A. (2001). Progressive brain volume
changes and the clinical course of schizophrenia in men: a longitudinal magnetic resonance
imaging study. Arch Gen Psychiatry 58: 148–157.
Mattson, M. P., Keller, J. N., Begley, J. G. (1998). Evidence for synaptic apoptosis. Exp Neurol 153:
35–48.
Mayerhoff, D. I., Loebel, A. D., Alvir, J. M. J., et al. (1994). The deficit state in first-episode
schizophrenia. Am J Psychiatry 151: 1417–1422.
McGlashan, T. H. (1988). A selective review of recent North American long-term follow-up
studies of schizophrenia. Schizophr Bull 14: 515–542.
Moritz, S., Andresen, B., Perro, C., for the PERSIST Study Group (2002). Neurocognitive perfor-
mance in first-episode and chronic schizophrenic patients. Eur Arch Psychiatry Clin Neurosci
252: 33–37.
Murray, R. M., Lewis, S. W. (1987). Is schizophrenia a neurodevelopmental disorder? Br Med J
296: 681–682.
Nair, T. R., Christensen, J. D., Kingsbury, S. J., et al. (1997). Progression of cerebroventricular
enlargement and the subtyping of schizophrenia. Psychiatry Res Neuroimaging 74: 141–
150.
Nopoulos, P., Flashman, L., Flaum, M., Arndt, S., Andreasen, N. (1994). Stability of cognitive
functioning early in the course of schizophrenia. Schizophr Res 14: 29–37.
Ona, V. O., Li, M., Vonsattel, J. P. G., et al. (1999). Inhibition of caspase-1 slows disease progression
in a mouse model of Huntington’s disease. Nature 399: 263–267.
Onley, J. W., Farber, N. B. (1995). Glutamate receptor dysfunction and schizophrenia. Arch Gen
Psychiatry 52: 998–1007.
Pakkenberg, B. (1990). Pronounced reduction of total neuron number in mediodorsal thalamic
nucleus and nucleus accumbens in schizophrenics. Arch Gen Psychiatry 47: 1023–1028.
(1993). Total nerve cell number in neocortex in chronic schizophrenics and controls estimated
using optical disectors. Biol Psychiatry 34: 768–772.
Pantelis, C., Velakoulis, D., McGorry, P. D., et al. (2002). Neuroanatomical abnormalities before
and after onset of psychosis: a cross-sectional and longitudinal MRI comparison. Lancet
Accessed 10 December 2002: http://image.thelancet.com/extras/01art9092web.pdf.
388 L. F. Jarskog, J. H. Gilmore, and J. A. Lieberman
Pettegrew, J. W., Keshavan, M. S., Panchalingam, K., et al. (1991). Alterations in brain high-
energy phosphate and membrane phospholipid metabolism in first-episode, drug-naive
schizophrenics: a pilot study of the dorsal prefrontal cortex by in vivo phosphorus 31 nuclear
magnetic resonance spectroscopy. Arch Gen Psychiatry 48: 563–568.
Pettegrew, J. W., Keshavan, M. S., Minshew, N. J. (1993). 31 P Nuclear magnetic resonance spec-
troscopy: neurodevelopment and schizophrenia. Schizophr Bull 19: 35–53.
Pfohl, B., Winokur, G. (1982). The evolution of symptoms in institutionialized hebephrenic/
catatonic schizophrenics. Br J Psychiatry 141: 567–572.
Pierri, J. N., Volk, C. L., Auh, S., Sampson, A., Lewis, D. A. (2001). Decreased somal size of deep
layer 3 pyramidal neurons in the prefrontal cortex of subjects with schizophrenia. Arch Gen
Psychiatry 58: 466–473.
Pinals, D. A., Breier, A. (1997). Schizophrenia. In Psychiatry, ed. A. Tasman, J. Kay, J. A. Lieberman.
Philadelphia, PA: W. B. Saunders, pp. 927–965.
Purohit, D. P., Perl, D. P., Haroutunian, V., et al. (1998). Alzheimer disease and related neu-
rodegenerative diseases in elderly patients with schizophrenia. Arch Gen Psychiatry 55: 205–
211.
Rajkowska, G., Selemon, L. D., Goldman-Rakic, P. S. (1998). Neuronal and glial somal size in
the prefrontal cortex: a postmortem morphometric study of schizophrenia and Huntington
disease. Arch Gen Psychiatry 55: 215–224.
Rapoport, J. L., Giedd, J., Kumra, S., et al. (1997). Childhood-onset schizophrenia: progressive
ventricular change during adolescence. Arch Gen Psychiatry 54: 897–903.
Rapoport, J. L., Giedd, J. N., Blumenthal, J., et al. (1999). Progressive cortical change during
adolescence in childhood-onset schizophrenia: a longitudinal magnetic resonance imaging
study. Arch Gen Psychiatry 56: 649–654.
Roberts, G. W., Colter, N., Lofthouse, R., et al. (1986). Gliosis in schizophrenia: a survey. Biol
Psychiatry 21: 1043–1050.
Roberts, G. W., Colter, N., Lofthouse, R., Johnstone, E. C., Crow, T. J. (1987). Is there
gliosis in schizophrenia? Investigation of the temporal lobe. Biol Psychiatry 22: 1459–
1468.
Rund, B. R. (1998). A review of the longitudinal studies of cognitive decline in schizophrenia
patients. Schizophr Bull 24: 425–435.
Saykin, A. J., Shtasel, D. L., Gur, R. E., et al. (1994). Neuropsychological deficits in neuroleptic
naive patients with first episode schizophrenia. Arch Gen Psychiatry 51: 124–131.
Schlaepfer, T. E., Harris, G. J., Tien, A. Y., et al. (1994). Decreased regional cortical gray matter
volume in schizophrenia. Am J Psychiatry 151: 842–848.
Selemon, L. D., Rajkowska, G., Goldman-Rakic, P. S. (1995). Abnormally high neuronal density
in the schizophrenic cortex: a morphometric analysis of prefrontal area 9 and occipital area
17. Arch Gen Psychiatry 52: 805–818.
(1998). Elevated neuronal density in prefrontal area 46 in brains from schizophrenic patients:
application of a three-dimensional, stereologic counting method. J Comp Neurol 392: 402–
412.
Smith, A. (1964). Mental deterioration in chronic schizophrenia. J Nerv Ment Dis 139: 479–487.
389 Neurodegenerative models of schizophrenia
Stanley, J. A., Williamson, P. C., Drost, D. J., et al. (1995). An in vivo study of the prefrontal cortex
of schizophrenic patients at different stages of illness via phosphorus magnetic resonance
spectroscopy. Arch Gen Psychiatry 52: 399–406.
Stevens, J. R. (1982). Neuropathology of schizophrenia. Arch Gen Psychiatry 39: 1131–1139.
Thompson, P. M., Sower, A. C., Perrone-Bizzozero, N. I. (1998). Altered levels of the synaptosomal
associated protein SNAP-25 in schizophrenia. Biol Psychiatry 43: 239–243.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
Weinberger, D. R., McClure, R. K. (2002). Neurotoxicity, neuroplasticity, and magnetic resonance
imaging morphometry: what is happening in the schizophrenic brain? Arch Gen Psychiatry
59: 553–558.
Weinberger, D. R., Berman, K. F., Zec, R. F. (1986). Physiologic dysfunction of dorsolateral pre-
frontal cortex in schizophrenia. I. Regional cerebral blood flow evidence. Arch Gen Psychiatry
43: 114–124.
Woods, B. T. (1998). Is schizophrenia a progressive neurodevelopmental disorder? Toward a
unitary pathogenetic mechanism. Am J Psychiatry 155: 1661–1670.
Wyatt, R. J. (1991). Neuroleptics and the natural course of schizophrenia. Schizophr Bull 17:
325–351.
(1995). Early intervention for schizophrenia: can the course of the illness be altered? Biol
Psychiatry 38: 1–3.
Young, K. A., Manaye, K. F., Liang, C. H., Hicks, P. B., German, D. C. (2000). Reduced number of
mediodorsal and anterior thalamic neurons in schizophrenia. Biol Psychiatry 47: 944–953.
Zipursky, R. B., Lim, K. O., Sullivan, E. V., Brown, B. W., Pfefferbaum, A. (1992). Widespread
cerebral gray matter volume deficits in schizophrenia. Arch Gen Psychiatry 49: 195–205.
Zorrilla, L. T. E., Heaton, R. K, McAdams, L. A., et al. (2000). Cross-sectional study of older
outpatients with schizophrenia and health comparison subjects: no differences in age-related
decline. Am J Psychiatry 157: 1324–1326.
21
The view that major psychiatric disorders such as schizophrenia and bipolar dis-
orders (BPD) are distinct clinical entities is being increasingly challenged (Taylor,
1992). Several authors have proposed the concept of an expanded psychiatric con-
tinuum between affective disorders and schizophrenia and several lines of evidence
support this notion. First, the cross-sectional and longitudinal co-occurrence of
affective and schizophrenic symptoms is well known, leading to that diagnostic
conundrum called schizoaffective disorder (Adler and Strakowski, 2003). Second,
both affective disorder and schizophrenia demonstrate a high degree of genetic sus-
ceptibility; some recent gene mapping studies show common susceptibility genes for
the two disorders (Berrettini, 2000). Third, BPD and schizophrenia also demon-
strate some similarities in neurotransmitter and neurophysiological dysfunction
(Moller, 2003). Finally, many newer atypical antipsychotic agents such as olanzap-
ine and quetiapine, initially approved for the treatment of schizophrenia, are also
being increasingly used in BPD, suggesting a therapeutic continuum (Moller, 2003).
Similar boundary issues exist between schizophrenia and other psychiatric dis-
orders. Although obsessive–compulsive disorder (OCD) and schizophrenia are
generally considered to be separate psychiatric disorders, comorbidities of the
two illnesses are frequent, leading to the occasional diagnosis of schizo-obsessive
disorder (Gross-Isseroff et al., 2003). As will be reviewed below, neuroimaging
studies show overlapping findings in these two illnesses, in particular abnormali-
ties in the frontostriatal circuits. Attention-deficit hyperactivity disorder (ADHD)
also frequently coexists with schizophrenia (Bellak et al., 1987) and BPD (Kim and
Miklowitz, 2002). Many children with ADHD have features of thought disorder
similar to children with schizophrenia (Caplan et al., 2002). Individuals at genetic
risk for schizophrenia (Keshavan et al., 2002a) and BPD (Chang et al., 2000) also
have an increased frequency of ADHD.
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
390
391 Disordered brain development in psychiatric disorders
OCD
Schizo-
ADHD
phrenia
BPD/MDD
Total brain
Corpus callosum
Prefrontal cortex
Hippocampus
Amygdala
Basal ganglia
Thalamus
Cingulate
in unmedicated children with ADHD, compared with both medicated children with
ADHD and controls. Medicated patients with ADHD did not differ from controls
in total white matter volumes. This underscores the importance of controlling for
potentially confounding factors such as medication status and age. Reductions in
total brain volume is similar to that observed in schizophrenia (see Ch. 19).
Frontostriatal circuitry
As the executive, “decision-making” center of the brain, the prefrontal cortex and
its striatal target fields have been considered key regions of interest in ADHD.
Similar to reports of decreased prefrontal lobe activity (Andreasen et al., 1992)
and decreased striatal volumes (Keshavan et al., 1998; Shihabuddin et al., 1998)
in schizophrenia, converging lines of evidence support reduced prefrontal and
striatal volumes and hypoactivation of the prefrontal cortex and striatum in
patients with ADHD (Aylward et al., 1996; Bush et al., 1999; Casey et al., 1997;
Castellanos et al., 1994, 2001, 2002; Filipek et al., 1997; Hynd et al., 1990; Rubia
et al., 1999; Teicher et al., 2000). Reversed asymmetry of the caudate nucleus has
also been found in pediatric patients with ADHD (Hynd et al., 1993). Reduced cau-
date volumes appear to be most pronounced in younger patients with ADHD, and
394 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
Controls ADHD
Mean Mean
95% Confidence Interval 95% Confidence Interval
Males Females
1150
Volume (ml)
1050
950
850
5 10 15 20 5 10 15 20
Age (years) Age (years)
Fig. 21.2. Predicted unadjusted longitudinal growth curves for total cerebral volumes for patients with
attention-deficit hyperactivity disorder (ADHD) and controls. (Taken with permission from
Castellanos, F. X., Lee, P. P., Sharp, W., et al., 2002. Developmental trajectories of brain vol-
ume abnormalities in children and adolescents with attention-deficit/hyperactivity disorder.
JAMA 288: 1740–1748.)
Controls ADHD
Mean Mean
95% Confidence Interval 95% Confidence Interval
11 135
Volume (ml)
10 125
9 115
5 10 15 20 5 10 15 20
Age (years) Age (years)
Fig. 21.3. Predicted unadjusted longitudinal growth curves for total caudate and cerebellar volume for
patients with attention-deficit hyperactivity disorder (ADHD) and controls. (Taken with per-
mission from Castellanos, F. X., Lee, P. P., Sharp, W., et al., 2002. Developmental trajectories of
brain volume abnormalities in children and adolescents with attention-deficit/hyperactivity
disorder. JAMA 288: 1740–1748.)
Obsessive–compulsive disorder
Neurobiological studies in various laboratories using diverse techniques have con-
sistently found alterations in ventral prefrontostriatothalamic function in OCD
associated with symptom severity and response to treatment (Rosenberg et al., 2001;
Fig. 21.4). Recent investigation in treatment-naive pediatric patients with OCD has
suggested a neural network dysplasia, with reduced striatal volumes and increased
anterior cingulate and thalamic volumes. Rosenberg and colleagues (2000a, 2001)
have proposed that a reversible thalamocorticostriatal dysfunction in glutamatergic
396 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
Prefrontal cortex
Volumetric MRI investigation in treatment-naive pediatric patients with OCD
compared with age- and sex-matched controls has demonstrated increased anterior
397 Disordered brain development in psychiatric disorders
5-HT = Serotonin
GABA = Gamma-aminobutyrate
Glutamate
DA = dopamine
Globus Pallidus
Ventral Prefrontal
Cortex Ventral
Striatum
Thalamus
Temporal Lobe
Cortex
Substantia Nigra/
Ventral Tegmentum
Raphe
Nucleus
Fig. 21.5. Schematic diagram showing selected aspects of corticostriatal connections in the neurode-
velopment of obsessive–compulsive disorder. Arrows show the actions of different neuro-
transmitters. (Adapted by permission from D. R. Rosenberg and M. S. Keshavan. Toward
a neurodevelopmental model of obsessive–compulsive disorder. Biol Psychiatry, 43: 623,
1998. Permission to reprint adaptation also from Farchione, T. R., MacMillan, S. N., Rosen-
berg, D. R. 2003. Obsessive–compulsive disorder as a fronto–striatal–thalamic dysfunction.
In Mental and Behavioral Dysfunction in Movement Disorders. Ed. Bedard, M. A., Agid, Y.,
Chouinard, S., et al. Totowa, NJ: Humana Press.)
cingulate volume in the former (Rosenberg and Keshavan, 1998; Fig. 21.6). These
observations contrast with cingulate volume reductions seen in schizophrenia
(Noga et al., 1995; Takahashi et al., 2003). The age-related increase in anterior
cingulate volume in healthy children appears to be absent in children with OCD.
Increased anterior cingulate volumes were positively correlated with increased OCD
symptom severity and inversely correlated with reduced striatal volumes. No sig-
nificant differences were observed between children with OCD and controls in
dorsolateral prefrontal cortex, posterior cingulate, whole temporal lobe, amygdala,
hippocampus, or superior temporal gyrus volumes. By contrast, volumetric reduc-
tions have been seen in these structures in schizophrenia (reviewed by Shenton
et al., 2001). It should be noted that the increases in anterior cingulate volumes seen
in OCD have not been reported in pediatric patients with ADHD. Finally, reduced
NAA levels suggestive of neuronal dysfunction have been reported in the anterior
398 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
2
6 8 10 12 14 16 18 20
Age (years)
Controls
OCD Patients
Fig. 21.6. Anterior cingulate volume as a function of age for pediatric patients with obsessive–
compulsive disorder and normal controls. Opposite neurodevelopmental trajectories in the
patients compared with the healthy subjects may reflect an absence of early neuronal prun-
ing in children with OCD. (Reprinted with permission from Rosenberg, D. R. Keshavan,
M. S. Toward a neurodevelopmental model of obsessive–compulsive disorder. Biol Psychi-
atry 43: 623, 1998.)
cingulate of pediatric patients with OCD (Ebert et al., 1997). Reduced anter-
ior cingulate glutamatergic concentrations have also been reported in pediatric
patients with OCD, which increase to levels not significantly different from controls
after effective treatment with the selective serotonin reuptake inhibitor paroxetine
(Rosenberg et al., 2002b). We have suggested that increased caudate glutamater-
gic concentrations coupled with reduced anterior cingulate glutamatergic concen-
trations in treatment-naive pediatric patients with OCD suggest a tonic–phasic
dysregulation of the glutamatergic system involving the corticostriatal circuits in
OCD (Rosenberg and Keshavan, 1998). Tonic cortical glutamatergic activity has an
inhibitory influence on phasic stress-related glutamate release from the striatum,
particularly the ventral striatum. Therefore, reduced tonic glutamatergic activity in
the anterior cingulate might predispose to phasic glutamatergic overactivity in the
striatum. The fact that caudate glutamatergic increase is reversible with paroxetine
treatment further supports this idea.
399 Disordered brain development in psychiatric disorders
Thalamus
The thalamus serves as the final subcortical relay station to various cortical
structures, including the frontal cortex. There is evidence that thalamic volumes
are reduced in first-episode neuroleptic drug-naive patients with schizophrenia
(Gilbert et al., 2001), whereas thalamic volumes are significantly increased in
treatment-naive pediatric patients with OCD compared with controls. After 12
weeks of monodrug therapy with paroxetine, thalamic volumes decreased signif-
icantly to levels not significantly different from controls. Reduction in thalamic
volumes in pediatric patients with OCD was positively correlated with reduction
in OCD symptom severity, with higher pretreatment thalamic volumes predicting
enhanced response to paroxetine. In contrast, thalamic volumes did not change
significantly in treatment-naive pediatric patients with OCD treated for 12 weeks
with cognitive behavioral therapy (Rosenberg et al., 2000b). Consequently, reduc-
tion in thalamic volumes may be specific to treatment with a selective serotonin
reuptake inhibitor rather than linked to a spontaneous resolution of symptoms or
some more generalized treatment response.
Fitzgerald et al. (2000) used proton MRS to evaluate medial and lateral subregions
of the thalamus since medial thalamic regions have been particularly implicated in
OCD (Modell et al., 1989). Reduced levels of NAA were found in medial, but not
lateral, thalamus in pediatric patients with OCD. Increased thalamic volumes in
pediatric patients with OCD were associated with lower levels of NAA, suggestive
of neuronal dysfunction.
Basal ganglia
Similar to findings in patients with ADHD and schizophrenia, reduced striatal vol-
umes have been observed in child and adolescent patients with OCD compared with
controls, using both quantitative computed tomography (Luxenberg et al., 1988)
and volumetric MRI (Rosenberg et al., 1997b). Reductions in striatal volumes
were associated with increased OCD symptom severity, but not illness duration
(Rosenberg et al., 1997b). Consistent with reduced striatal volumes, increased ven-
tricle to brain ratios and increased third ventricular volumes have been observed in
children and adolescents with OCD compared with healthy controls (Behar et al.,
1984; Rosenberg et al., 1997b).
Also, similar to reports in ADHD, reductions in NAA have been reported in the
striatum in patients with OCD compared with controls (Bartha et al., 1998; Ebert
et al., 1997). Bartha et al. (1998) proposed that this reduction in neuronal viabil-
ity may be mediated by glutamatergic excitotoxicity. Therefore, Rosenberg et al.
(2000a) measured caudate glutamatergic concentrations in treatment-naive pedi-
atric patients with OCD before and after monodrug therapy with paroxetine and
compared these with case-matched pediatric controls. The occipital cortex was also
400 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
Frontal cortex
Reduced frontal lobe volumes have been consistently reported in adult and pedi-
atric patients with MDD (Steingard et al., 2002). These authors also noted reduced
frontal white matter volumes and significantly larger frontal gray matter volumes
in adolescent patients with MDD compared with healthy adolescents. It appears
that patients with ADHD or MDD have reduced frontal white matter volumes com-
pared with controls (Castellannos et al., 2002), but the former also have reduced gray
matter volumes. Striking left, but not right, volumetric reductions in the subgenual
region of the prefrontal cortex, which are associated with reduced regional cerebral
blood flow in this region, have been observed in adult patients with familial MDD
or BPD (at least one first-degree relative with MDD or BPD) compared with that
in patients with non-familial MDD or BPD (no evident family history) (Drevets,
2000). Botteron et al. (2002) also reported comparable left but not right volumetric
reductions of the subgenual region of the prefrontal cortex in 30 young women aged
18–23 years with early-onset MDD and 18 women aged 24–52 years with recur-
rent MDD compared with matched controls. It should be noted that no significant
differences from controls in left or right subgenual prefrontal cortex in patients
with MDD or BPD were seen in other studies (Brambilla et al., 2002; Bremner
et al., 2002). A recent investigation by Nolan et al. (2002) in 22 psychotropic drug-
naive pediatric patients with MDD compared with 22 case-matched controls iden-
tified distinct neuroanatomic differences in left but not right prefrontal cortical vol-
umes in familial but not non-familial MDD. Reduced left prefrontal cortical volume
was correlated with increased depressive symptom severity and longer illness dura-
tion in familial but not non-familial MDD. Left-sided prefrontal cortical volume
reduction in patients with familial MDD may represent a progressive rather than a
static developmental effect and familial/genetic factors may be involved in patho-
genesis given. However, since all investigations to date have been cross-sectional
rather than longitudinal studies, definitive conclusions are premature.
were larger in pediatric patients with MDD than in controls, while hippocampal
volumes tended to be smaller in the MDD patients. Pine et al. (2001) has also found
that excess fear in adolescence is a robust predictor for development of MDD in
early adulthood. Moreover, in a recent fMRI study conducted by Thomas et al.
(2001), measuring amygdala response to fearful faces in children with generalized
anxiety disorder and panic disorder, children with MDD, and healthy controls, the
magnitude of signal change in the amygdala was most associated with the severity
of the child’s anxiety in patients with MDD and generalized anxiety disorder.
When considering whether disordered brain development cuts across diagnostic
boundaries, it is important to point out that increased amygdala volume is one of
the more consistent findings observed in volumetric neuroimaging studies of adult
patients with BPD (Altshuler et al., 2000; Frodl et al., 2002; Strakowski et al., 1999).
Increased amygdala activation in response to fearful facial affect has also been
observed in fMRI studies conducted in patients with BPD (Thomas et al., 2001).
Pediatric MDD carries an increased risk for becoming BPD in adulthood (Geller
et al., 2001; Harrington et al., 1990; Weissman et al., 1999), and increased amygdala
and reduced hippocampal volumes are seen in pediatric patients with MDD and in
adults with first-episode MDD (Frodl et al., 2002; MacMillan et al., 2003). These
findings contrast with those reporting decreased amygdala volumes in patients
with schizophrenia (reviewed by Shenton et al., 2001), and in those at risk for this
disorder (Keshavan et al., 2002b; Ch. 19). Longitudinal MRI studies are necessary
to determine whether anatomic and functional alterations in the amygdala may
serve as potential biomarkers of risk for development of BPD.
Only one available study has examined the corpus callosum in unipolar depres-
sion and this found an increase in size (Wu et al., 1993). The corpus callosum also
appears to be larger in BPD (Hauser et al., 1989), and the striatum appears larger in
first-episode BPD (Strakowski et al., 2002). However, basal ganglia volumes appear
to be reduced in depression (reviewed by Soares and Mann, 1997).
Genetic component
There is a substantial genetic component to both schizophrenia and ADHD.
Early family, twin, and adoptee studies have shown a strong likelihood of genetic
403 Disordered brain development in psychiatric disorders
susceptibility in both ADHD and schizophrenia. Combined reports suggest that the
mean concordance for schizophrenia within monozygotic twins is approximately
46% and within dizygotic pairs is approximately 14% (Gottesman, 1991; Moldin
and Gottesman, 1997). The rate of ADHD in first-degree relatives of children diag-
nosed as having ADHD ranges from 16% to 25% (Biederman et al., 1992, 1991).
Molecular genetic findings suggest that alterations in the dopaminergic system
may underlie both ADHD and schizophrenia. Several groups have reported alter-
ations in the gene encoding the dopamine D4 receptor in children diagnosed with
ADHD (Benjamin et al., 1996; Ebstein et al., 1996; LaHoste et al., 1996). Cook
et al. (1995) and Gill et al. (1997) have both reported an association between
alterations in the gene encoding the dopamine transporter and ADHD. Alter-
ations in the gene encoding catechol-O-methyltransferase (COMT), an enzyme that
degrades dopamine, have been associated with schizophrenia (reviewed by Sawa and
Snyder, 2002) and with BPD (Badner and Gershon, 2002). In animals with a tar-
geted deletion of the COMT gene, decreased levels of dopamine are seen only in
the prefrontal cortex (Gogos et al., 1998). Therefore, changes affecting dopamine
metabolism could lead to a decrease in prefrontal dopamine levels, and such deficits
may underlie the executive function deficits seen in both schizophrenia and BPD.
A heritability component has been suggested in both OCD and BPD. However,
by contrast with schizophrenia where replicable findings are beginning to emerge
(O’Donovan et al., 2003; Ch. 1), few replicated genetic susceptibility loci have been
identified for OCD and BPD. One finding suggests linkage of BPD to a location on
chromosome 18 (Berrettini et al., 1994; Stine et al., 1995). However, the large-scale
National Institute of Mental Health Genetics Bipolar Group study (1997) did not
find an association between BPD and chromosome 18.
Obstetric complications
While accumulating evidence points to the role of OCs in vulnerability for
schizophrenia (Ch. 11), the diagnostic specificity and the etiological significance of
this association remain unclear. Other severe psychiatric disorders, such as ADHD
and psychotic affective disorder, have been linked with OCs; low birth weight,
neonatal complications, and excessive bleeding have been associated with ADHD
(Milberger et al., 1997; Sprich-Buckminster et al., 1993). OCs have been observed
sporadically in BPD (reviewed by Buka and Fan, 1999) but could not be reliably
replicated. Complications within these studies include small sample sizes and data
obtained primarily in retrospect from maternal recall. A recent comprehensive
study showed no excess in OCs in mania (Browne et al., 2000).
Overall, OCs may increase the vulnerability for a range of severe mental disorders
in a relatively non-specific way and may interact with genetic liability and later
environmental risk factors (Verdoux and Sutter, 2002).
404 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
Conclusions
The studies described in this chapter indicate the impressive commonalities in neu-
rodevelopmentally mediated pathophysiological alterations across several major
psychiatric disorders. The similarities in pathophysiology are consistent with
the observations of frequent comorbidity and diagnostic overlap. The similar-
ity between ADHD and schizophrenia in abnormalities of the overall brain vol-
ume, corpus callosum size, and frontostriatal circuits is striking and is consis-
tent with the increasing understanding of attentional impairment as a core feature
of schizophrenia (Ch. 4). Additionally, observations of attentional problems are
among the most robust premorbid predictors of later schizophrenia (Ch. 23). There
is good evidence that ADHD also predicts the later emergence of BPD (Kim and
Miklowitz, 2002). Interestingly, an association has also been proposed between
ADHD and childhood OCD (Geller et al., 2002). Therefore, attentional impair-
ments in childhood may serve as the bridge connecting the commonalities between
diverse psychiatric presentations in adulthood, such as OCD, BPD, and schizophre-
nia (Fig. 21.1). Considering the earlier onset of ADHD compared with the other
disorders, it may even be suggested that attentional impairments, perhaps related
to distributed cortical gray and white matter changes, especially in the prefrontal
cortex, may represent a non-specific precursor for the later development psychiatric
disorders, at least in some patients.
The comparisons between overlapping disorders also reveal some intriguing
differences. For example, while schizophrenia and OCD share similar reductions
in striatal volumes, OCD shows increased cingulate volumes while schizophre-
nia shows decreased volumes. Similarly, patients with BPD and possibly those
with OCD show larger amygdala volumes, whereas smaller amygdala volumes are
observed in schizophrenia. Patients with BPD may also show larger basal gan-
glia, unlike the schizophrenia patients. This suggests that different developmen-
tal trajectories may emerge during late childhood and adolescence from com-
mon neurodevelopmental precursors in childhood. These developmental trajec-
tories may be related to differential pathoplastic responses of the subcortical
brain structures, such as the cingulate and amygdala, and may mediate the emer-
gence of distinct symptomatic presentations. It is useful to recall here that the
amygdala and the cingulate are critically involved in affect regulation and con-
flict monitoring, respectively (Carter et al., 1999; Ledoux, 2003). Consequently,
the amygdala enlargements in BPD might represent a pathological hyperplasia
in that region related to repeated activation in the context of highly valenced
emotional states, such as the manic and depressive episodes. Likewise, the cin-
gulate enlargement in OCD might reflect a hyperresponsive error-monitoring
system. By comparison, failure of optimum functioning of the cingulate and
405 Disordered brain development in psychiatric disorders
REFERENCES
Adler, C. M., Strakowski, S. M. (2003). Boundaries of schizophrenia. Psychiatr Clin North Am 26:
1–23.
Altshuler, L. L., Bartzokis, G., Grieder, T., et al. (2000). An MRI study of temporal lobe structures
in men with bipolar disorder or schizophrenia. Biol Psychiatry 48: 147–162.
Andreasen, N. C., Rezai, K., Alliger, R., et al. (1992). Hypofrontality in neuroleptic-naive patients
and in patients with chronic schizophrenia. Assessment with xenon 133 single-photon
emission computed tomography and the Tower of London. Arch Gen Psychiatry 49: 943–
958.
Aylward, E. H., Reiss, A. L., Reader, M. J., et al. (1996). Basal ganglia volumes in children with
attention-deficit hyperactivity disorder. J Child Neurol 11: 112–115.
Badner, J. A., Gershon, E. S. (2002). Meta-analysis of whole-genome scans of bipolar disorder
and schizophrenia. Mol Psychiatry 7: 405–411.
Bartha, R., Stein, M. B., Williamson, P. C., et al. (1998). A short echo 1 H spectroscopy and
volumetric MRI study of the corpus striatum in patients with obsessive–compulsive disorder
and comparison subjects. Am J Psychiatry 155: 1584–1591.
Behar, D., Rapoport, J. L., Berg, C. J., et al. (1984). Computerized tomography and neuropsy-
chological test measures in adolescents with obsessive–compulsive disorder. Am J Psychiatry
141: 363–369.
Bellak, L., Kay, S. R., Opler, L. A. (1987). Attention deficit disorder psychosis as a diagnostic
category. Psychiatr Dev 5: 239–263.
406 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
Benazon, N. R., Ager, J., Rosenberg, D. R. (2001). Cognitive behavior therapy in treatment-naive
children and adolescents with obsessive–compulsive disorder: an open trial. Behav Res Ther
40: 529–539.
Benjamin, J., Li, L., Patterson, C., et al. (1996). Population and familial association between the
D4 dopamine receptor gene and measures of novelty seeking. Nat Genet 12: 81–84.
Berrettini, W. (2000). Review of bipolar molecular linkage and association studies. Curr Psychiatry
Rep 4: 124–129.
Berrettini, W. H., Ferraro, T. N., Alexander, R. C., Buchberg, A. M., Vogel, W. H. (1994). Quanti-
tative trait loci mapping of three loci controlling morphine preference using inbred mouse
strains. Nat Genet 7: 54–58.
Biederman, J., Faraone, S. V., Keenan, V., Tsuang, M. T. (1991). Evidence of familial association
between attention deficit disorders and major affective disorder. Arch Gen Psychiatry 78:
633–672.
Biederman, J., Faraone, S. V., Keenan, K., et al. (1992). Further evidence for familial-genetic risk
factors in ADHD: patterns of comorbidity in probands and relatives in psychiatrically and
pediatrically referred samples. Arch Gen Psychiatry 49: 728–738.
Birken, D. L., Oldendorf, W. H. (1989). N-Acetyl-L-aspartic acid: a literature review of a com-
pound prominent in 1 H-NMR spectroscopic studies of brain. Neurosci Biobehav Rev 13:
23–31.
Bolton, J., Moore, G. J., MacMillan, S., Stewart, C. M., Rosenberg, D. R. (2001). Case study:
caudate glutamatergic changes with paroxetine persist after medication discontinuation in
pediatric OCD. J Am Acad Child Adolesc Psychiatry 40: 903–906.
Botteron, K. N., Raichle, M. E., Drevets, W. C., Heath, A. C., Todd, R. D. (2002). Volumetric
reduction in left subgenual prefrontal cortex in early onset depression. Biol Psychiatry 51:
342–344.
Brambilla, P., Harenski, K., Nicoletti, M. A., et al. (2001). Anatomical MRI study of basal ganglia
in bipolar disorder patients. Psychiatry Res 106: 65–80.
Brambilla, P., Nicoletti, M. A., Harenski, K., et al. (2002). Anatomical MRI study of subgenual
prefrontal cortex in bipolar and unipolar subjects. Neuropsychopharmacology 27: 792–799.
Bremner, J. D., Vythilingam, M., Vermetten, E., et al. (2002). Reduced volume of orbitofrontal
cortex in major depression. Biol Psychiatry 51: 273–279.
Browne, R., Byrne, M., Mulryan, N., et al. (2000). Labor and delivery complications at birth and
later mania. Br J Psychiatry 176: 369–372.
Buka, S., Fan, A. (1999). Association of prenatal and perinatal complications with subsequent
bipolar disorder and schizophrenia. Schizophr Res 39: 113–119.
Bush, G., Frazier, J. A., Rauch, S. L., et al. (1999). Anterior cingulate cortex dysfunction in
attention-deficit/hyperactivity disorder revealed by fMRI and the Counting Stroop. Biol
Psychiatry 45: 1542–1552.
Caplan, R., Guthrie, D., Tang, B., Nuechterlein, K. H., Asarnow, R. E. (2002). Thought disorder
in attention-deficit hyperactivity disorder. Int Rev Neurobiol 49: 269–284.
Carter, C. S., Botvinick, M. M., Cohen, J. D. (1999). The contribution of the anterior cingulate
cortex to executive process in cognition. Rev Neurosci 10: 49–57.
407 Disordered brain development in psychiatric disorders
Casey, B. J., Castellanos, F. X., Giedd, J. N., et al. (1997). Implication of right frontostriatal circuitry
in response inhibition and attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc
Psychiatry 36: 374–383.
Castellanos, F. X., Giedd, J. N., Eckburg, P., et al. (1994). Quantitative morphology of the caudate
nucleus in attention deficit hyperactivity disorder. Am J Psychiatry 151: 1791–1796.
Castellanos, F. X., Giedd, J. N., Berquin, P. C., et al. (2001). Quantitative brain magnetic resonance
imaging in girls with attention-deficit/hyperactivity disorder. Arch Gen Psychiatry 58: 289–
295.
Castellanos, F. X., Lee, P. P., Sharp, W., et al. (2002). Developmental trajectories of brain volume
abnormalities in children and adolescents with attention-deficit/hyperactivity disorder. J
Am Mod Assoc 288: 1740–1748.
Chang, K. D., Steiner, H., Ketter, T. A. (2000). Psychiatric phenomenology of child and adolescent
bipolar offspring. J Am Acad Child Adolesc Psychiatry 39: 453–460.
Cook, E. H., Sterin, M. A., Krasowski, M. D., et al. (1995). Association of attention-deficit disorder
and the dopamine transporter gene. Am J Hum Genet 56: 993–998.
Drevets, W. C. (2000). Neuroimaging studies of mood disorders. Biol Psychiatry 48: 813–829.
Durston, S., Hulshoff Pol, H. E., Casey, B. J., et al. (2001). Anatomical MRI of the developing
human brain: what have we learned? J Am Acad Child Adolesc Psychiatry 40: 1012–1020.
Ebert, D., Speck, O., Konig, A., et al. (1997). 1 H-magnetic resonance spectroscopy in obsessive–
compulsive disorder: evidence for neuronal loss in the cingulate gyrus and the right striatum.
Psychiatry Res 74: 173–176.
Ebstein, R. P., Novick, O., Umansky, R., et al. (1996). Dopamine D4 receptor (D4DR) exon III
polymorphism associated with the human personality trait of novelty seeking. Nat Genet
12: 78–80.
Ernst, M., Zametkin, A. J., Matochik, J. A., Jons, P. J., Cohen, R. M. (1998). DOPA decarboxylase
activity in attention deficit hyperactivity disorder adults. A [fluorine-18]fluorodopa positron
emission tomographic study. J Neurosci 18: 5901–5907.
Ernst, M., Zametkin, A. J., Matochik, J. A., et al. (1999). High midbrain [18 F]DOPA accumu-
lation in children with attention deficit hyperactivity disorder. Am J Psychiatry 156: 1209–
1215.
Farchione, T. R., MacMillan, S. N., Rosenberg, D. R. (2003). Obsessive–compulsive disorder as
a fronto–striatal–thalamic dysfunction. In Mental and Behavioral Dysfunction in Movement
Disorders, ed. M. A. Bedord, Y. Agid, S. Chouinard, et al. Totowa, NJ: Humana Press, pp.
477–488.
Filipek, P. A., Semrud-Clikeman, M., Steingard, R. J., et al. (1997). Volumetric MRI analysis
comparing attention-deficit hyperactivity disorder and normal controls. Neurology 48: 589–
601.
Fitzgerald, K. D., Moore, G. J., Paulson, L. D., Stewart, C. M., Rosenberg, D. R. (2000). Proton
spectroscopic imaging of the thalamus in treatment-naive pediatric obsessive–compulsive
disorder. Biol Psychiatry 48: 55–72.
Frodl, T., Meisenzahl, E., Zetzsche, T., et al. (2002). Enlargement of the amygdala in patients with
a first episode of major depression. Biol Psychiatry 51: 708–714.
408 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
Geller, B., Zimerman, B., Williams, M., Bolhofner, K., Craney, J. L. (2001). Bipolar disorder
at prospective follow-up of adults who had prepubertal major depressive disorder. Am J
Psychiatry 158: 125–127.
Geller, D. A., Biederman, J., Faraone, S. V., et al. (2002). Attention-deficit/hyperactivity disorder
in children and adolescents with obsessive–compulsive disorder: fact or artifact? J Am Acad
Child Adolesc Psychiatry 41: 52–58.
Gilbert, A. R., Moore, G. J., Keshavan, M. S., et al. (2000). Decrease in thalamic volumes of
pediatric patients with obsessive–compulsive disorder who are taking paroxetine. Arch Gen
Psychiatry 57: 449–456.
Gilbert, A. R., Rosenberg, D. R., Harenski, K., et al. (2001). Thalamic volumes in patients with
first-episode schizophrenia. Am J Psychiatry 158: 618–624.
Gill, M., Daly, G., Heron, S., Hawi, Z., Fitzgerald, M. (1997). Confirmation of association between
attention deficit hyperactivity disorder and a dopamine transporter polymorphism. Biol
Psychiatry 2: 311–313.
Gogos, J. A., Morgan, M., Luine, V., et al. (1998). Catechol-O-methyltransferase-deficient mice
exhibit sexually dimorphic changes in catecholamine levels and behavior. Proc Natl Acad Sci
USA 95: 9991–9996.
Gottesman, I. L. (1991). Schizophrenia Genesis: The Origins of Madness. New York: Freeman.
Gross-Isseroff, R., Hermesh, H., Zohar, J., Weizman, A. (2003). Neuroimaging communality
between schizophrenia and obsessive–compulsive disorder: a putative basis for schizo-
obsessive disorder? World J Biol Psychiatry 4: 129–134.
Harrington, R., Fudge, H., Rutter, M., Pickles, A., Hill, J. (1990). Adult outcomes of childhood
and adolescent depression: psychiatric status. Arch Gen Psychiatry 47: 465–473.
Hauser, P., Dauphinais, I. D., Berrettini, W., et al. (1989). Corpus callosum dimensions mea-
sured by magnetic resonance imaging in bipolar affective disorder and schizophrenia. Biol
Psychiatry 26: 659–668.
Hesslinger, B., Thiel, T., Tebartz van Elst, L., Hennig, J., Ebert, D. (2001). Attention-deficit disorder
in adults with or without hyperactivity: where is the difference? A study in humans using
short echo 1 H-magnetic resonance spectroscopy. Neurosci Lett 304: 117–119.
Hill, D. E., Yeo, R. A., et al. (2003). Magnetic resonance imaging correlates of attention-
deficit/hyperactivity disorder in children. Neuropsychology 17: 496–506.
Hynd, G. W., Semrud-Clikeman, M., Lorys, A. R., Novey, E. S., Eliopulos, D. (1990). Brain
morphology in developmental dyslexia and attention deficit disorder/hyperactivity. Arch
Neurol 47: 919–926.
Hynd, G. W., Hern, K. L., Novey, E. S., et al. (1993). Attention deficit hyperactivity disorder and
asymmetry of the caudate nucleus. J Child Neurol 8: 339–347.
Kayl, A. E., Moore, III, B. D., Slopis, J. M., Jackson, E. F., Leeds, N. E. (2000). Quantitative
morphology of the corpus callosum in children with neurofibromatosis and attention-deficit
hyperactivity disorder. J Child Neurol 15: 90–96.
Keshavan, M. S., Montrose, D. M., Pierri, J. N., et al. (1997). Magnetic resonance imaging and
spectroscopy in offspring at risk for schizophrenia: preliminary studies. Prog Neuropsy-
chopharmacol Biol Psychiatry 21: 1285–1295.
409 Disordered brain development in psychiatric disorders
Keshavan, M. S., Rosenberg, D., Sweeney, J. A., Pettegrew, J. W. (1998). Decreased caudate volume
in neuroleptic-naive psychotic patients. Am J Psychiatry 155: 774–778.
Keshavan, M. S., Sujata, M., Mehra, A., Montrose, D. M., Sweeney, J. A. (2002a). Psychosis
proneness and ADHD in young relatives at risk for schizophrenia. Schizophr Res 59: 85–92.
Keshavan, M. S., Dick, E., Mankowski, I., et al. (2002b). Decreased left amygdala and hippocampal
volumes in young offspring at risk for schizophrenia. Schizophr Res 58: 173–183.
Kellner, C. H., Jolley, R. R., Holgate, R. C., et al. (1991). Brain MRI in obsessive–compulsive
disorder. Psychiatry Res 36: 45–49.
Kim-Cohen, J., Caspi, A., Moffitt, T. E., et al. (2003). Prior juvenile diagnoses in adults with
mental disorder: developmental follow-back of a prospective-longitudinal cohort. Arch Gen
Psychiatry 60: 709–717.
Kim, E. Y., Miklowitz, D. J. (2002). Childhood mania, attention deficit hyperactivity disorder and
conduct disorder: a critical review of diagnostic dilemmas. Bipolar Disord 4: 215–225.
LaHoste, G. J., Swanson, J. M., Wigal, S. B., et al. (1996). Dopamine D4 receptor gene polymor-
phism is associated with attention-deficit hyperactivity disorder. Mol Psychiatry 1: 121–124.
Ledoux, J. (2003). The emotional brain, fear, and the amygdala. Cell Mol Neurobiol 23: 727–738.
Luxenberg, J. S., Swedo, S. E., Flament, M. F., et al. (1988). Neuroanatomical abnormalities in
obsessive–compulsive disorder detected with quantitative X-ray computed tomography. Am
J Psychiatry 145: 1089–1093.
MacMaster, F. P., Carrey, N., Sparkes, S., Khan, S., Kusumakar, V. (2003). Proton spectroscopy in
medication free pediatric attention deficit hyperactivity disorder. Biol Psychiatry 53: 184–
187.
MacMillan, S. N., Szeszko, P. R., Moore, G. J., et al. (2003). Increased amygdala:hippocampal
volumes associated with severity of anxiety in pediatric major depression. J Child Adol
Psychpharm 13: 65–73.
Milberger, S., Biederman, J., Faraone, S. X., Guite, J., Tsuang, M. T. (1997). Pregnancy, deliv-
ery, and infancy complications and attention deficit hyperactivity disorder: issues of gene–
environment interaction. Biol Psychiatry 41: 65–75.
Modell, J. G., Mountz, J. M., Curtis, G. C., Greden, J. F. (1989). Neurophysiologic dysfunction
in basal ganglia/limbic striatal and thalamocortical circuits as a pathogenetic mechanism of
obsessive–compulsive disorder. J Neuropsychiatry Clin Neurosci 1: 27–36.
Moldin, S. O., Gottesman, I. I. (1997). At issue: genes, experiments and chance in schizophrenia-
positioning for the 21st century. Schizoph Bull 23: 547–561.
Moller, H. J. (2003). Bipolar disorder and schizophrenia: distinct illnesses or a continuum? J Clin
Psychiatry 64: 23–27.
National Institute of Mental Health Genetics Initiative Bipolar Group (1997). Genomic survey
of bipolar illness in the NIMH Genetics Initiative pedigrees: a preliminary report. Am J Med
Gen Neuropsychiatr Genet 74: 227–237.
Noga, J. T., Aylward, E., Barta, P. E., Pearlson, G. D. (1995). Cingulate gyrus in schizophrenic
patients and normal volunteers. Psychiatry Res 61: 201–208.
Nolan, C. L., Moore, G. J., Madden, R., et al. (2002). Prefrontal cortical volume in childhood-onset
major depression: preliminary findings. Arch Gen Psychiatry 59: 173–179.
410 C. W. Kreipke, D. R. Rosenberg, and M. S. Keshavan
O’Donovan, M. C., Williams, N. M., Owen, M. J. (2003). Recent advances in the genetics of
schizophrenia. Hum Mol Genet 12(Suppl 2): R125–R133.
Pine, D. S., Cohen, P., Brook, J. (2001). Adolescent fears as predictors of depression. Biol Psychiatry
50: 721–724.
Rosenberg, D. R., Keshavan, M. S., Dick, E. L., et al. (1997a). Corpus callosal morphology in
treatment-naive pediatric obsessive–compulsive disorder. Prog Neuropsychopharmacol Biol
Psychiatry 21: 1269–1283.
Rosenberg, D. R., Keshavan, M. S., O’Hearn, K. M., et al. (1997b). Frontostriatal measurement
in treatment-naive children with obsessive–compulsive disorder. Arch Gen Psychiatry 54:
824–830.
Rosenberg, D. R., Keshavan, M. S. (1998). A. E. Bennett Research Award. Toward a neurodevel-
opmental model of obsessive–compulsive disorder. Biol Psychiatry 43: 623–640.
Rosenberg, D. R., MacMaster, F. P., Keshavan, M. S., et al. (2000a). Decrease in caudate glutamater-
gic concentrations in pediatric obsessive–compulsive disorder patients taking paroxetine. J
Am Acad Child Adolesc Psychiatry 39: 1096–1103.
Rosenberg, D. R., Benazon, N. R., Gilbert, A., Sullivan, A., Moore, G. J. (2000b). Thalamic volume
in pediatric obsessive–compulsive disorder patients before and after cognitive behavioral
therapy. Biol Psychiatry 48: 294–300.
Rosenberg, D. R., MacMillan, S. N., Moore, G. J. (2001). Brain anatomy and chemistry may predict
treatment response in pediatric obsessive–compulsive disorder. Int J Neuropsychopharmacol
4: 179–190.
Rosenberg, D. R., Davanzo, P. A., Gershon, S. (2002a). Pharmacotherapy for Child and Adolescent
Psychiatric Disorders, 2nd edn (revised and expanded). New York: Marcel Dekker.
Rosenberg, D. R., Madden, R., Tang, J., et al. (2002b). Anterior cingulate glutamate and treat-
ment response in pediatric OCD. In Scientific Proceedings of the 49th Annual Meeting of the
American Academy of Child and Adolescent Psychiatry. Washington DC: American Academy
of Child and Adolescent Psychiatry, Vol. D18, p. 118.
Rubia, K., Overmeyer, S., Taylor, E., et al. (1999). Hypofrontality in attention deficit hyperactivity
disorder during higher-order motor control: a study with functional MRI. Am J Psychiatry
156: 891–896.
Sawa, A., Snyder, S. (2002). Schizophrenia: diverse approaches to a complex disease. Science 296:
692–695.
Schweitzer, J. B., Anderson, C., Ernst, M. (2000). Attention-deficit hyperactivity disorder: neu-
roimaging and behavioral/cognitive probes. In Functional Neuroimaging in Child Psychiatry,
ed. M. Ernst, J. Rumsey. Cambridge, UK: Cambridge University Press, pp. 278–297.
Shenton, M. E., Dickey, C. C., Frumin, M., McCarley, R. W. (2001). A review of MRI findings in
schizophrenia. Schizophr Res 49: 1–52.
Shihabuddin, L., Buchsbaum, M. S., Hazlett, E. A., et al. (1998). Dorsal striatal size, shape, and
metabolic rate in never-medicated and previously medicated schizophrenics performing a
verbal learning task. Arch Gen Psychiatry 55: 235–243.
Soares, J. C., Mann, J. J. (1997). The anatomy of mood disorders: review of structural neuroimag-
ing studies. Biol Psychiatry 41: 86–106.
411 Disordered brain development in psychiatric disorders
Sowell, E. R., Toga, A. W., Asarnow, R. (2000). Brain abnormalities observed in childhood-onset
schizophrenia: a review of the structural magnetic resonance imaging literature. Ment Retard
Dev Disabil Res Rev 6: 180–185.
Sprich-Buckminster, S., Biederman, J., Milberger, S., Faraone, S. W., Lehman, B. K. (1993). Are
perinatal complications relevant to the manifestation of ADD? Issues of comorbidity and
familiality. J Am Acad Child Adolesc Psychiatry 32: 1032–1037.
Steingard, R. J., Renshaw, P. F., Hennen, J., et al. (2002). Smaller frontal lobe white matter volumes
in depressed adolescents. Biol Psychiatry 52: 413–417.
Stine, O. C., Xu, J., Koskela, R., et al. (1995). Evidence for linkage of bipolar disorder to chromo-
some 18 with a parent-of-origin effect. Am J Hum Genet 57: 1384–1394.
Strakowski, S. M., DelBello, M. P., Sax, K. W., et al. (1999). Brain magnetic resonance imaging
of structural abnormalities in bipolar disorder. Arch Gen Psychiatry 56: 254–260.
Strakowski, S. M., DelBello, M. P., Zimmerman, M. E., et al. (2002). Ventricular and periventric-
ular structural volumes in first- versus multiple-episode bipolar disorder. Am J Psychiatry
159: 1841–1847.
Takahashi, T., Suzuki, M., Kawasaki, Y., et al. (2003). Perigenual cingulate gyrus volume in patients
with schizophrenia: a magnetic resonance imaging study. Biol Psychiatry 53: 593–600.
Tannock, R. (1998). Attention deficit hyperactivity disorder: advances in cognitive, neurobiolog-
ical, and genetic research. J Child Psychol Psychiatry 39: 65–99.
Taylor, M. A. (1992). Are schizophrenia and affective disorder related? A selective literature review.
Am J Psychiatry 149: 22–32.
Teicher, M. H., Anderson, C. M., Polcari, A., et al. (2000). Functional deficits in basal ganglia
of children with attention-deficit/hyperactivity disorder shown with functional magnetic
resonance imaging relaxometry. Nat Med 6: 470–473.
Thomas, K. M., Drevets, W. C., Dahl, R. E., et al. (2001). Amygdala response to fearful faces in
anxious and depressed children. Arch Gen Psychiatry 58: 1057–1063.
Vaidya, C. J., Austin, G., Kirkorian, G., et al. (1998). Selective effects of methylphenidate in
attention deficit hyperactivity disorder: a functional magnetic resonance study. Proc Natl
Acad Sci USA 95: 14494–14499.
Verdoux, H., Sutter, A. L. (2002). Perinatal risk factors for schizophrenia: diagnostic specificity
and relationships with maternal psychopathology. Am J Med Genet 114: 898–905.
Weissman, M. M., Wolk, S., Wickramaratne, P., et al. (1999). Children with prepubertal-onset
major depressive disorder and anxiety grown up. Arch Gen Psychiatry 56: 794–801.
Wu, J. C., Buchsbaum, M. S., Johnson, J. C., et al. (1993). Magnetic resonance and positron
emission tomography imaging of the corpus callosum: size, shape and metabolic rate in
unipolar depression. J Affect Disord 28: 15–25.
Zametkin, A. J., Nordahl, T. E., Gross, M., et al. (1990). Cerebral glucose metabolism in adults
with hyperactivity of childhood onset. N Engl J Med 323: 1361–1366.
Part IV
Clinical implications
22
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
415
Table 22.1. Predicting schizophrenia: neurointegrative risk factors in population-based, high-risk and
archival–observational studies
Risk factor OR/(RR) SEN (%) SPE (%) PPV (%) NPV (%)
Population-based studies
Delayed developmental milestones at 2 yrs 4.8 7 98 3 99
(Jones et al., 1994)a
Learned to stand at or after 12 months (1.4) – – – –
(Isohanni et al., 2001)b
Excess of neurological signs at 3 yrs 4.6 – – – –
(Cannon et al., 2002)c
Unusual movements at 7 yrs (Rosso et al., 2000)d 4.4 15 96 3 99
Motor coordination dysfunction at 7 yrs (Rosso 2.4 11 96 2 99
et al., 2000)d
High-risk studies
Deviance on a composite index of attentional, – 89 64 22 98
perceptual and motor dysfunctioning at
8–15 yrs (Marcus et al., 1987)e
Gross motor skills at 7–12 yrs – 75 73 33 94
(Erlenmeyer-Kimling et al., 2000)f
Archival-observational studies
Postural hand abnormalities in the first 2 yrs of 7.9g 63 82 – –
life (Walker et al., 1994)g
Choreoathetoid movements in the first 2 yrs of 8.2g 23 96 – –
life (Walker et al., 1994)g
OR, odds ratio; RR, risk ratio (the ratio between the observed and the expected cases); SEN, sensitivity; SPE,
specificity; PPV, positive predictive value; NPV, negative predictive value.
a The British MRC National Survey of Health and Development of 1946 followed prospectively a stratified
random sample (4746 alive at 16 years) of 13 687 births in Britain during the week 3–9 March 1946. Thirty
subjects developed schizophrenia at ages 16–43 and were compared with the remaining 4716 subjects.
b The Northern Finland Birth Cohort Study (Isohanni et al., 1998, 1999, 2001) followed prospectively 96%
of all births in Northern Finland in 1966 (12 058 liveborn children). Psychiatric outcome was established at
16–28 years (58 subjects had developed schizophrenia), and the comparison group included individuals with
no hospital-treated psychiatric outcome (Isohanni et al., 1998, 1999).
c The Dunedin Birth Cohort Study followed prospectively 91% (1037) of all children born between April
1972 and March 1973 in Dunedin (New Zealand). At age 26, 36 had developed schizophreniform disorder,
20 mania, and 278 anxiety/depression. These diagnostic groups were each compared with the remainder of
the cohort (healthy control group).
d The Philadelphia birth cohort study (Bearden et al., 2000; Rosso et al., 2000) followed prospectively over 90%
(9236) of all deliveries at two inner city hospital obstetric wards in Philadelphia between 1959 and 1966. The
cohort included 72 subjects who developed schizophrenia/schizoaffective disorder at ages 19–36, 63 healthy
siblings, and 7941 non-psychiatric controls.
e The National Institute of Mental Health (NIMH) Israeli Kibbutz–City High-Risk Study followed prospectively
50 offspring of schizophrenia parents and 50 offspring of psychiatrically normal parents. Diagnostic outcome
417 Can one identify preschizophrenia children?
preschizophrenia children on the basis of these characteristics? Are there early risk
factors that, in isolation or in combination, flag the preschizophrenia status? These
questions, including the putative neurobiological predictions of the schizophrenia
illness (Ch. 23), are also addressed elsewhere in this volume.
Where they are available from the primary sources or computable through infor-
mation derived from the text, the following association indexes and predictive
parameters will be reported: the odds ratio (OR) is the likelihood of developing
schizophrenia in the children who test positive for the predictor relative to those
who do not. Sensitivity is the percentage of children with adult schizophrenia out-
comes who test positive for the predictor. Specificity is the percentage of children
with normal adult outcomes who test negative for the predictor. Positive predictive
value is the probability of developing adult schizophrenia given the presence of the
predictor in childhood. Negative predictive value is the probability of escaping adult
schizophrenia given the absence of the predictor in childhood.
We will place particular emphasis on prospective population-based, high-risk,
and follow-back studies. All three types of study provide relatively unbiased infor-
mation, as both researchers and informants do not know which subjects will develop
schizophrenia, and the data collected are contemporaneous rather than retrospec-
tive. In addition, general population cohorts are less confounded by the selection
and information biases usually associated with case–control studies.
Neurointegrative predictors
Population-based studies
Unusual movements and coordination problems in mid-childhood were signif-
icantly associated with adult schizophrenia in the Philadelphia 1959–1966 Birth
was established at 26–32 years (nine high-risk subjects had developed schizophrenia-spectrum disorders). The
predictive validity of neurobehavioral deviance was estimated in a subgroup of 46 high-risk and 44 control
offspring.
f The New York High-Risk Project (Cornblatt et al., 1999; Erlenmeyer-Kimling and Cornblatt 1992; Erlenmeyer-
Kimling et al., 2000; Ott et al., 1998) recruited offspring of schizophrenia, affectively ill, and psychiatrically
normal parents in 1971–2 (sample A: 206, mean age 9.5 years) and 1977–9 (sample B: 150, mean age 9.0 years)
and followed them prospectively. Erlenmeyer-Kimling et al. (2000) assessed the predictive validity of gross
motor skills in a sample of 79 offspring of schizophrenia parents. Diagnostic outcome was established at a
mean age of 30.7 years (12 offspring had developed schizophrenia-related psychoses).
g Walker et al. (1994) observed childhood home videos of 30 schizophrenia patients and their 28 healthy siblings,
19 affective disorder patients and their 14 siblings, and 21 subjects from families with no mental illness. The
OR, sensitivity, specificity, positive and negative predictive values reported here refer to the comparison of the
preschizophrenia subjects with their healthy siblings.
418 E. Kravariti, P. Dazzan, P. Fearon, and R. M. Murray
Cohort Study (Rosso et al., 2000). The positive predictive value of these indices
remained low, however (Table 22.1). An excess of motor coordination dysfunction
was also observed in the unaffected siblings of subjects with adult schizophrenia,
suggesting that coordination problems might be related to a genetic vulnerability
to develop the disorder.
Individuals who developed schizophrenia in the 1946 British birth cohort study
(MRC National Survey of Health andDevelopment; Jones et al.,1994)were 4.8times
more likely to have delayed milestones (sitting, standing, walking, and talking) at
age 2 (Table 22.1). However, the probability of developing schizophrenia was only
3% (Table 22.1). The authors indicated that “. . . no child destined to develop
schizophrenia could be singled out as a late walker.” Subjects with a diagnosis of
childhood affective disorder (but not those with an adult-onset affective disorder)
according to an anxiety–depression rating also attained later motor milestones and
had an excess of grimaces and twitches at age 15 (van Os et al., 1997). Therefore,
although there were some similarities between the antecedents of schizophrenia and
those of affective disorders, the effect on the latter was smaller and more evident
in those with an early onset. Age at learning to stand, walk, or become potty-
trained in the Northern Finland 1966 Birth Cohort Study (Isohanni et al., 2001)
showed a linear association not only with schizophrenia (Table 22.1) but also with
other psychoses. By contrast, motor problems (more than 0.3 standard deviation
[SD]) and an excess of neurological signs (OR, 4.6) in the Dunedin study (Cannon
et al., 2002) were present in children who developed adult schizophreniform, but
not manic or anxiety/depression, outcomes. Similarly, children who developed
adult schizophrenia in the 1946 British cohort and in the 1958 cohort (National
Child Development Study of 1958; Done et al., 1994) studies showed more “hand
control and speech problems” than either children who remained well or those who
developed affective psychosis (Crow et al., 1995; Done et al., 1994; Leask et al., 2002).
High-risk studies
Fish (Fish, 1977; Fish et al., 1992) created the term “pandevelopmental retarda-
tion” to refer to abnormalities in physical growth, gross motor development and
visual–motor development that are frequently observed in subjects who develop
schizophrenia (Fish, 1987). Hyperkinesis, poor coordination, motor dysfunction,
and perceptual signs were present in eight of the nine children who later devel-
oped schizophrenia or schizoid personality disorder in the Israeli High Risk Study
(Marcus et al., 1987). Similarly, impairment in gross motor skills in the New York
High Risk Study was predictive of a future diagnosis of schizophrenia with a rea-
sonably high sensitivity and specificity (Erlenmeyer-Kimling et al., 2000). However,
even children with grossly abnormal motor skills only had a 33% probability of
developing schizophrenia (Table 22.1).
419 Can one identify preschizophrenia children?
Archival–observational studies
In a study of sibships filmed from infancy through at least the first 5 years of life, each
including one offspring who later developed schizophrenia and normal offspring,
viewers blind to the subjects’ diagnosis were able to identify the preschizophrenia
siblings on the basis of neuromotor abnormalities (Walker et al., 1994). Those who
would develop schizophrenia were 7.9 times more likely to show postural hand
abnormalities, and 8.2 times more likely to show choreo-athetoid movements than
their control siblings. These group differences were only significant until 2 years of
age, after which they tended to disappear. It is possible that preschizophrenia chil-
dren have an impaired cortical modulation on subcortical structures, as supported
by evidence of disrupted connectivity in patients with schizophrenia (Rosso et al.,
2000).
Such neurodevelopmental deficits could be the consequence of pregnancy and
delivery complications (for example, fetal hypoxia), which have been associated
with both schizophrenia (Geddes and Lawrie, 1995; McGrath and Murray, 1995;
McNeil, 1995) and unusual movements (Rosso et al., 2000). Alternatively, obstetric
complications may just play a non-specific, precipitating, or facilitating role in
subjects with a genetic liability to schizophrenia (Cannon et al., 2000a).
Neuropsychological predictors
Population-based studies
Prospective investigations of population-based samples have reported persistent
intellectual deficits, language pathology, and educational failures in preschizophre-
nia children and adolescents (Bearden et al., 2000; Cannon et al., 1999, 2000b;
Isohanni et al., 1998, 1999; Jones and Done, 1997; Jones et al., 1994), with the
respective functional abnormalities being relatively stable across developmental
periods (Cannon et al., 2000b; Jones and Done, 1997).
Individuals destined to develop schizophrenia in the Philadelphia Birth Cohort
Study showed intellectual abnormalities as early as 4 and 7 years of age (Cannon
et al., 2000b); intelligence quotient (IQ) category at either age emerged as a signif-
icant predictor of adult schizophrenia (Cannon et al., 2000b). These findings were
supplemented by the 1946 and 1958 British birth cohort studies (Jones and Done,
1997), which reported deficits in intellectual capacity, verbal and non-verbal rea-
soning, reading comprehension, and mathematics at various age points between
7 and 16 years of age in preschizophrenia individuals. Repeating a school grade
or receiving special education at age 14 (mainly owing to low IQ) also predicted
future schizophrenia in the Northern Finland Birth Cohort Study (Isohanni et al.,
1998). Similarly, poor intellectual functioning in a national sample of 18-year-
old male conscripts to the Swedish army emerged as a strong predictor of future
420 E. Kravariti, P. Dazzan, P. Fearon, and R. M. Murray
schizophrenia (David et al., 1997), a finding replicated in the national Israeli cohort
of males aged 16–17 years (Davidson et al., 1999).
A common finding in nearly all of the above studies is a linear association between
intellectual functioning and the risk for schizophrenia: the latter appears to be a
function of performance over the entire range of population scores, increasing
progressively as ability declines (Cannon et al., 1999, 2000b; David et al., 1997;
Davidson et al., 1999; Jones and Done, 1997; Jones et al., 1994). The Philadelphia
Birth Cohort Study, for example, reported a 30–60% increase in schizophrenia risk
per unit decrease in ability category (divided into five performance levels), such
that individuals scoring in the deficient range were five to six times more likely
to become schizophrenia than those scoring in the high-average to superior range
(Cannon et al., 1999). Similarly, David et al. (1997) reported a nine-fold increase
in schizophrenia risk among conscripts scoring in the lowest IQ band compared
with those falling within the highest one.
Despite the significant increase in schizophrenia risk in the lower end of the IQ
distribution, in no study did the disorder arise solely from a population subgroup
with low IQ, nor was there evidence of a subgroup with very low scores (Cannon
et al., 1999, 2000b; David et al., 1997; Davidson et al., 1999; Jones and Done, 1997;
Jones et al., 1994). Rather, the IQ distribution of the schizophrenia population
as a whole is shifted downward in a systematic fashion, most likely reflecting an
effect on each individual. This effect is rather subtle: IQ data at age 11 examined
together for the 1946 and 1958 birth cohorts revealed a shift of the schizophrenia
population mean by < 0.5 SD (Jones and Done, 1997). No single child destined to
develop schizophrenia in the former cohort could be singled out as having learning
difficulties (Jones et al., 1994).
Contrary to the prevailing view that schizophrenia risk increases linearly with
decreasing intellectual capacity, the Northern Finland Birth Cohort Study (Isohanni
et al., 1999) failed to confirm linearity in the association between educational
attainment and schizophrenia outcome, raising the possibility that it is distance
from the cognitive norm – in either direction – that increases the odds for the
disorder. Excellent school performance among 16-year-old males in the latter cohort
was associated with nearly a four-fold increase in schizophrenia risk, with 11% of the
preschizophrenia boys compared with only 3% of the comparison group (with no
hospital-treated psychiatric outcome) obtaining excellent school marks. In accord
with this finding, the proportion of preschizophrenia cases falling within the highest
IQ category among the Israeli males aged 16–17 years was six times higher than
that of the comparison subjects (with no hospital-treated psychiatric outcome)
(Davidson et al., 1999). It remains to be seen whether these surprising findings will
be confirmed in other populations.
421 Can one identify preschizophrenia children?
Risk factor OR SEN (%) SPE (%) PPV (%) NPV (%)
OR, odds ratio; SEN, sensitivity; SPE, specificity; PPV, positive predictive value; NPV, negative predictive
value; IQ, intelligence quotient.
a See footnote d in Table 22.1.
b Odds ratio for linear trend, i.e., that associated with moving a category (e.g. tertile or quintile) of score
distribution.
c See footnote a in Table 22.1.
d The National Israeli sample of 16–17-year-old males included 509 subjects who developed schizophrenia
by the age of 26, and 9215 controls who did not appear in the national psychiatric registry by the same age
(Davidson et al., 1999).
e A national sample of 49 968 male conscripts to the Swedish army (David et al., 1997): 195 subjects developed
schizophrenia by the age of 33–34 and were compared with 49 773 individuals with no or other psychotic
outcomes. OR values (unadjusted for confounders) were estimated separately for each IQ band (< 74, 74–81,
82–89, 90–95) by David et al. (1997); SEN, SPE, PPV, and NPV were estimated for all the IQ (< 96) bands
together by the present authors.
f See footnote b, in Table 22.1.
High-risk studies
The “high-risk” strategy reduces the chances of false positives in the face of low
prevalence disorders by studying individuals whose risk for developing schizophre-
nia is high, thus increasing the observed prevalence of the disorder in the sample
studied: children of one schizophrenia parent can be up to 15 times more likely to
develop the disorder than members of the general population. In addition, unlike
population-based studies, which usually study general aspects of development,
research in high-risk groups is a longitudinal quest for variables that predict the
disorder (Erlenmeyer-Kimling et al., 2000). As such, the high-risk paradigm usually
includes measurements of particular relevance to schizophrenia.
Impaired attention is the most extensively investigated and validated neuropsy-
chological predictor in the high-risk literature. For example, the preschizophrenia
offspring of affected parents in the Copenhagen High Risk Study displayed poor
concentration from infancy (Parnas et al., 1982). Similarly, childhood attentional
deficits in the Israeli high-risk sample successfully predicted the development of
schizophrenia-spectrum disorders (Mirsky et al., 1995). In addition, in the New York
High Risk Project, attentional deficits predicted schizophrenia-related psychoses as
opposed to other psychiatric outcomes (Erlenmeyer-Kimling et al., 2000), were
more prevalent among children of schizophrenia parents than offspring of affect-
ively ill patients (Cornblatt and Erlenmeyer-Kimling, 1985; Cornblatt et al., 1992;
Erlenmeyer-Kimling et al., 2000; Freedman et al., 1998), and displayed the greatest
predictive power within the group at genetic risk for schizophrenia (Cornblatt and
Erlenmeyer-Kimling, 1985; Cornblatt et al., 1992). These findings attest both to
the predictive validity and the diagnostic specificity of attentional dysfunction in
high-risk studies (Table 22.3).
Although other cognitive functions have been less extensively investigated, mem-
ory dysfunction, IQ change, and subtest scatter in standard intelligence tests may
provide sensitive indicators of the preschizophrenia state (Erlenmeyer-Kimling
et al., 2000; Ott et al., 1998). The use of intelligence scores from the Revised Wech-
sler Intelligence Scale for Children (WISC-R) or the Revised Adult Scale (WAIS-R)
in the New York High Risk Project achieved one of the best predictive profiles
reported in the high-risk literature to date, with a false-positive rate (equal to
1 minus specificity) of just 2–5% (Ott et al., 1998) (Table 22.3).
However, the focus on single neuropsychological constructs is less promising
than using clusters of cognitive and neurobehavioral variables. Combining an atten-
tional screen with behavioral ratings in the New York High Risk Project enhanced
424 E. Kravariti, P. Dazzan, P. Fearon, and R. M. Murray
Risk factor SEN (%) SPE (%) PPV (%) NPV (%)
SEN, sensitivity; SPE, specificity; PPV, positive predictive value; NPV, negative predictive value.
a Data from the New York High Risk Project (see footnote f, Table 22.1).
b The predictive validity of sustained attention – alone or in conjunction with behavioral ratings – was assessed
in a mixed sample of 21 offspring of schizophrenia parents, 26 children of affectively ill parents, and 40 offspring
of psychiatrically normal parents (total, 87). Diagnostic outcome was last updated when the subjects were in
their early thirties.
c The predictive validity of attention, memory, and motor skills (each alone or in conjunction with the other
predictors) was assessed in a sample of 79 offspring of schizophrenia parents. Diagnostic outcome was estab-
lished at a mean age of 30.7 years (12 offspring had developed schizophrenia-related psychoses).
d The accuracy of a logistic regression model (including variables from Wechsler Intelligence Scales for Children
and Adults) was examined for prediction of schizophrenia-related psychoses (versus no diagnosis) in a mixed
sample of 157 high- and low-risk children (offspring of schizophrenia, affectively ill, and psychiatrically normal
parents). The diagnostic outcome was established at the mean ages of 30.17 years (sample A) and 22.09 years
(sample B). The ranges of SEN, SPE, PPV and NPV resulted from varying the predicted cut-off probability.
e The predictive accuracy of WISC(-R)/WAIS(-R)-derived variables was examined in a sample of 39 offspring
of schizophrenia parents derived from the sample described in footnote d of this table.
measurably the overall accuracy of the attentional model, reducing false positives
by half (Cornblatt et al., 1999). Similarly, combining deficits in attention, verbal
memory, and gross motor skills in the above study achieved higher precision than
any of the contributing variables alone (Erlenmeyer-Kimling et al., 2000).
Table 22.3 presents the predictive profiles of selected neuropsychological predict-
ors used in high-risk studies. As expected from measures specifically selected for
425 Can one identify preschizophrenia children?
their relevance to schizophrenia, and from the increased prevalence of the disorder
in the relatively small samples screened, the examined predictors show moderate-
to-high sensitivity, with 50–85% of future spectrum cases being correctly identi-
fied. Of all subjects predicted to develop schizophrenia-related psychoses, 19–70%
went on to develop the outcome of interest, and, almost invariably, over 90% of
“non-deviant” performers did not develop schizophrenia or related psychoses.
However, the false-positive rates can sometimes be substantial. Although in some
models (Erlenmeyer-Kimling et al., 2000; Ott et al., 1998), only 2–10% of non-cases
are falsely predicted to develop spectrum disorders, about a third of non-cases are
false positives in others (Erlenmeyer-Kimling et al., 2000) (Table 22.3). In addition,
the predictive performance of the reviewed models cannot be extrapolated to the
general population, since only about 10–15% of future schizophrenia patients have
an ill parent, and the normal control samples in the existing high-risk studies are
relatively small (Erlenmeyer-Kimling et al., 2000).
Behavioral predictors
Population-based studies
Teacher-rated behavioral deviance emerged as a potent predictor of adult
schizophrenia in the 1946 and 1958 British birth cohort studies. In the 1946 study,
social anxiety at age 15 showed a significant linear association with schizophre-
nia, such that individuals scoring within the highest tertile were six times more
likely to develop the disorder relative to those scoring in the lowest tertile (Jones
et al., 1994). Teacher-rated anxiety was also a prominent characteristic of the
preschizophrenia 7-year-old males of the 1958 study (Crow et al., 1995), as were
hostility and inconsequential behavior: 50% of the preschizophrenia males com-
pared with only 10% of normal controls received abnormal global ratings of
over-reactive behavior. A clear gender effect was noted in the 1958 study: the
7-year-old girls who later developed schizophrenia were hardly distinguishable
from normal controls. Even though they were finally rated as more withdrawn,
depressed, and likely to dismiss adult values in later childhood (age 11), they
showed no evidence of over-reactive behavior at any time point, with the excep-
tion of a trend towards adult-directed hostility at age 11. By then, depression and
a trend for dismissal of adult values were also added to the behavioral profile
of the preschizophrenia males. In contrast to the preschizophrenia children, the
preaffective children were unremarkable in their profiles, with the exception of
hostility at age 7 (boys) and restlessness at either 7 (boys) or 11 (girls) years of age.
The behavioral abnormalities of the preneurotic individuals were similar to those
of the preschizophrenia group, although they emerged at a slightly later stage of
development and were present in the females rather than the males (Crow et al.,
1995).
426 E. Kravariti, P. Dazzan, P. Fearon, and R. M. Murray
Table 22.4. Predicting schizophrenia: behavioral risk factors in population-based and high-risk studies
Risk factor OR SEN (%) SPE (%) PPV (%) NPV (%)
Population-based studies
Global social maladjustment at 7 yrs 2.54 29 86 2 99
(Bearden et al., 2000)a
Focal behavioral deviance at 4 yrs (Bearden 1.68 33 81 1 99
et al., 2000)a
Focal behavioral deviance at 7 yrs (Bearden 1.65 31 81 2 99
et al., 2000)a
Poor social functioning at 16–17 yrs (males) 4.37 48 88
(Davidson et al., 1999)b
Poor organizational ability at 16–17 yrs 2.03 30 88
(males) (Davidson et al., 1999)b
High-risk studies
Behavior problems at 15 yrs (males) (Olin
and Mednick, 1996)c
a. Disciplinary problems 39 96 88 71
b. Teacher-predicted future emotional or 43 88 67 73
psychotic problems
c. Repeated a grade 29 96 80 73
d. Disturbs class with inappropriate 50 82 64 72
behavior
e. Emotional reaction persists; high-strung 40 85 60 72
f. Lonely and rejected by peers 44 82 58 71
g. Treated by psychologist for problem 36 86 57 73
h. Easily excited or irritated 41 82 58 69
Behavior problems at 15 yrs (females) (Olin
and Mednick, 1996)c
a. Teacher-predicted future emotional or 30 97 75 82
psychotic problems
b. Nervousness 46 82 45 82
OR, odds ratio; SEN, sensitivity; SPE, specificity; PPV, positive predictive value; NPV, negative predictive
value.
a See footnote d in Table 22.1.
c The Copenhagen High Risk Project (Olin and Mednick, 1996) recruited 207 offspring of mothers with severe
schizophrenia and 104 matched control offspring of parents and grandparents with no history of psychiatric
hospitalization (mean age 15.1 yrs). At a mean age of 42 years, 33 subjects had developed schizophrenia,
10 schizophrenia-associated psychosis, and 46 schizotypal or paranoid personality disorder. The predictive
parameters refer to the comparison of the preschizophrenia group with those with non-psychiatric outcomes.
427 Can one identify preschizophrenia children?
High-risk studies
In the seminal study of Barbara Fish (1987), half of the preschizophrenia children
with chronic disturbance from childhood had been abnormally quiet infants with
flat affect, who developed withdrawn or explosive and non-compliant behavior by
the age of 3 years.
Eight of the nine adults who received schizophrenia-spectrum diagnoses in the
National Institute of Mental Health (NIMH) Israeli Kibbutz–City High Risk Study
had shown antisocial and/or withdrawn behavior in childhood (Marcus et al.,
1987). These abnormalities were mainly evident in the males. According to teachers
and parents, females had shown adequate interpersonal adjustment and less overt
dysfunctioning, even though the girls themselves had reported acute feelings of
rejection and not belonging.
Teacher ratings of social dysfunction and behavioral deviance among 15-year-old
males were among the best predictors of future schizophrenia in the Copenhagen
High Risk Study (Olin and Mednick, 1996) (Table 22.4). Compared with normal
controls, preschizophrenia males were more likely to pose discipline problems and
disturb the class, to be emotionally reactive and easily excited, to have been treated
by a school psychologist or repeated a grade, and to be lonely and rejected by
their peers. While both males and females with a schizophrenia outcome were
significantly more likely than normal controls to receive a prediction of future
psychosis by their teachers, the only other item to have predictive value for females
was nervousness. These behavioral indices predicted future schizophrenia with
low to moderate sensitivity, high specificity, and moderate to high positive and
negative predictive values (Table 22.4). Compared with males with other diagnostic
428 E. Kravariti, P. Dazzan, P. Fearon, and R. M. Murray
Archival–observational studies
In a preliminary study of sibships filmed from infancy through at least the first 5
years of life, each including a preschizophrenia child (Walker and Lewine, 1990),
viewers blind to the subjects’ adult psychiatric outcome were able to identify the
future cases at levels well above chance: 25 of their 32 judgements were correct
(P < 0.05). Furthermore, the distinguishing behavioral features of the target chil-
dren compared with their control sibling(s) (i.e. less responsiveness, eye contact,
and positive affect) were apparent at a gross level of analysis, as the viewers were
not instructed to use specific criteria. Similarly, on extending the above study to
a larger sample, Walker et al. (1993) observed lower rates of joy expressions and
increased negative affect in the future cases compared with same-gender siblings.
The emotional blunting of infants, children, and adolescents destined to develop
schizophrenia (Fish, 1987; Walker et al., 1993) is consistent with the lack of
hedonic capacity observed in affected adults, suggesting continuity between some
schizophrenia antecedents and the clinical syndrome. Other evidence in support
of this notion comes from a retrospective study. Baum and Walker (1995) found
positive correlations between withdrawal in childhood and adolescence and psy-
chomotor poverty in adult schizophrenia. Taken together, these findings suggest
that certain primary negative or deficit symptoms may be enduring traits, mani-
fested across the lifespan of schizophrenia patients.
Conclusions
Using a statistical analogy, the null hypothesis that preschizophrenia children as a
group cannot be identified on the basis of early developmental, cognitive, social,
and behavioral characteristics can be rejected. Subtle, yet detectable, abnormalities
in all these functional domains have been reliably reported in preschizophrenia
children, achieving a statistically significant predictive status in epidemiological,
high-risk and archival–observational studies. However, the predictive profile of
these characteristics on a population level advises against their use as schizophre-
nia screens in unselected populations. To date, no single predictor or composite
model has shown sufficient sensitivity, specificity, and positive/negative predictive
values. In the various studies reviewed, 11–93% of individuals with adult-onset
429 Can one identify preschizophrenia children?
REFERENCES
Baum, K. M., Walker, E. F. (1995). Childhood behavioral precursors of adult symptom dimensions
in schizophrenia. Schizophr Res 16: 111–120.
Bearden, C. E., Rosso, I. M., Hollister, J. M., et al. (2000). A prospective cohort study of child-
hood behavioral deviance and language abnormalities as predictors of adult schizophrenia.
Schizophr Bull 26: 395–410.
Cannon, T. D., Rosso, I. M., Bearden, C. E., Sanchez, L. E., Hadley, T. (1999). A prospective cohort
study of neurodevelopmental processes in the genesis and epigenesis of schizophrenia. Dev
Psychopathol 11: 467–485.
Cannon, T. D., Rosso, I. M., Hollister, J. M., et al. (2000a). A prospective cohort study of genetic
and perinatal influences in the etiology of schizophrenia. Schizophr Bull 26: 351–366.
Cannon, T. D., Bearden, C. E., Hollister, J. M., et al. (2000b). Childhood cognitive functioning in
schizophrenia patients and their unaffected siblings: a prospective cohort study. Schizophr
Bull 26: 379–393.
Cannon, M., Caspi, A., Moffitt, T. E., et al. (2002). Evidence for early-childhood, pan-
developmental impairment specific to schizophreniform disorder: results from a longi-
tudinal birth cohort. Arch Gen Psychiatry 59: 449–456.
Cornblatt, B. A., Erlenmeyer-Kimling, L. (1985). Global attentional deviance as a marker of risk
for schizophrenia: specificity and predictive validity. J Abnorm Psychol 94: 470–486.
Cornblatt, B. A., Lenzenweger, M. F., Dworkin, R. H., Erlenmeyer-Kimling, L. (1992). Childhood
attentional dysfunctions predict social deficits in unaffected adults at risk for schizophrenia.
Br J Psychiatry 18(Suppl.): 59–64.
430 E. Kravariti, P. Dazzan, P. Fearon, and R. M. Murray
Cornblatt, B. A., Obuchowski, M., Roberts, S., Pollack, S., Erlenmeyer-Kimling, L. (1999). Cog-
nitive and behavioral precursors of schizophrenia. Dev Psychopathol 11: 487–508.
Crow, T. J., Done, D. J., Sacker, A. (1995). Childhood precursors of psychosis as clues to its
evolutionary origins. Eur Arch Psychiatry Clin Neurosci 245: 61–69.
David, A. S., Malmberg, A., Brandt, L., Allebeck, P., Lewis, G. (1997). I. Q. and risk for schi-
zophrenia: a population-based cohort study. Psychol Med 27: 1311–1323.
Davidson, M., Reichenberg, A., Rabinowitz, J., et al. (1999). Behavioral and intellectual markers
for schizophrenia in apparently healthy male adolescents. Am J Psychiatry 156: 1328–1335.
Done, D. J., Crow, T. J., Johnstone, E. C., Sacker, A. (1994). Childhood antecedents of schizophre-
nia and affective illness: social adjustment at ages 7 and 11. Br Med J 309: 699–703.
Dworkin, R. H., Cornblatt, B. A. (1995). Predicting schizophrenia. Lancet 345: 139–140.
Erlenmeyer-Kimling, L., Cornblatt, B. A. (1992). A summary of attentional findings in the New
York High-Risk Project. J Psychiatr Res 26: 405–426.
Erlenmeyer-Kimling, L., Rock, D., Roberts, S. A., et al. (2000). Attention, memory, and motor
skills as childhood predictors of schizophrenia-related psychoses: the New York High-Risk
Project. Am J Psychiatry 157: 1416–1422.
Fearon, P., Cannon, M., Murray, R. M. (2001). A critique of the idea and science of risk-factor
research in schizophrenia. Int J Ment Health 30: 82–90.
Fish, B. (1977). Neurobiologic antecedents of schizophrenia in children. Evidence for an inherited,
congenital neurointegrative defect. Arch Gen Psychiatry 34: 1297–1313.
(1987). Infant predictors of the longitudinal course of schizophrenia development. Schizophr
Bull 13: 395–409.
Fish, B., Marcus, J., Hans, S. L., Auerbach, J. G., Perdue, S. (1992). Infants at risk for schizophrenia:
sequelae of a genetic neurointegrative defect. A review and replication analysis of pandys-
maturation in the Jerusalem infant development study. Arch Gen Psychiatry 49: 221–235.
Freedman, L. R., Rock, D., Roberts, S. A., Cornblatt, B. A., Erlenmeyer-Kimling, L. (1998). The
New York High-Risk Project: attention, anhedonia and social outcome. Schizophr Res 30:
1–9.
Geddes, J. R., Lawrie, S. M. (1995). Obstetric complications and schizophrenia: a meta-analysis.
Br J Psychiatry 167: 786–793.
Isohanni, I., Jarvelin, M.-R., Nieminen, P., et al. (1998). School performance as a predictor of
psychiatric hospitalization in adult life. A 28-year follow-up in the northern finland 1966
birth cohort. Psychol Med 28: 967–974.
Isohanni, I., Jarvelin, M. R., Jones, P., Jokelainen, J., Isohanni, M. (1999). Can excellent school
performance be a precursor of schizophrenia? A 28-year follow-up in the Northern Finland
1966 birth cohort. Acta Psychiatr Scand 100: 17–26.
Isohanni, M., Jones, P. B., Moilanen, K., et al. (2001). Early developmental milestones in adult
schizophrenia and other psychoses. A 31-year follow-up of the Northern Finland 1966 Birth
Cohort. Schizophr Res 52: 1–19.
Jones, P., Done, D. J. (1997). From birth to onset: a developmental perspective of schizophrenia
in two national birth cohorts. In Neurodevelopment and Adult Psychopathology, ed. M. S.
Keshavan, R. M. Murray. Cambridge, UK: Cambridge University Press, pp. 119–136.
431 Can one identify preschizophrenia children?
Jones, P., Rodgers, B., Murray, R., Marmot, M. (1994). Child developmental risk factors for adult
schizophrenia in the British 1946 birth cohort. Lancet 344: 1398–1402.
Kraepelin, E. (1919). Dementia Praecox and Paraphrenia. Edinburgh: Livingston.
Leask, S. J., Done, D. J., Crow, T. J. (2002). Adult psychosis, common childhood infections and
neurological soft signs in a national birth cohort. Br J Psychiatry 181: 387–392.
Marcus, J., Hans, S. L., Nagler, S., et al. (1987). Review of the NIMH Israeli Kibbutz-City Study
and the Jerusalem Infant Development Study. Schizophr Bull 13: 425–438.
McGrath, J., Murray, R. M. (1995). Risk factors for schizophrenia: from conception to birth. In
Schizophrenia, ed. S. Hirsh, D. Weinberger. Oxford: Blackwell, pp. 187–205.
McNeil, T. F. (1995). Perinatal risk factors and schizophrenia: selective review and methodological
concerns. Epidemiol Rev 17: 107–112.
Mirsky, A. F., Ingraham, L. J., Kugelmass, S. (1995). Neuropsychological assessment of attention
and its pathology in the Israeli cohort. Schizophr Bull 21: 193–204.
Murray, R. M. (1994). Neurodevelopmental schizophrenia: the rediscovery of dementia praecox.
Br J Psychiatry 165: 6–12.
Murray, R. M., Lewis, S. W. (1987). Is schizophrenia a neurodevelopmental disorder? Br Med J
(Clin Res Ed) 295: 681–682.
Olin, S. C., Mednick, S. A. (1996). Risk factors of psychosis: identifying vulnerable populations
premorbidly. Schizophr Bull 22: 223–240.
O’Toole, B. I. (2000). Screening for low prevalence disorders. Aust N Z J Psychiatry 34(Suppl):
S39–S46.
Ott, S. L., Spinelli, S., Rock, D., et al. (1998). The New York High-Risk Project: social and general
intelligence in children at risk for schizophrenia. Schizophr Res 31: 1–11.
Parnas, J., Schulsinger, F., Schulsinger, H., Mednick, S. A., Teasdale, T. W. (1982). Behavioral
precursors of schizophrenia spectrum. A prospective study. Arch Gen Psychiatry 39: 658–
664.
Rosso, I. M., Bearden, C. E., Hollister, J. M., et al. (2000). Childhood neuromotor dysfunction in
schizophrenia patients and their unaffected siblings: a prospective cohort study. Schizophr
Bull 26: 367–378.
van Os, J., Jones, P. B., Lewis, S., Wadsworth, M., Murray, R. M. (1997). Developmental precursors
of affective illness in a general population birth cohort. Arch Gen Psychiatry 54: 625–631.
Walker, E., Lewine, R. J. (1990). Prediction of adult-onset schizophrenia from childhood home
movies of the patients. Am J Psychiatry 147: 1052–1056.
Walker, E. F., Grimes, K. E., Davis, D. M., Smith, A. J. (1993). Childhood precursors of schizophre-
nia: facial expressions of emotion. Am J Psychiatry 150: 1654–1660.
Walker, E. F., Savoie, T., Davis, D. (1994). Neuromotor precursors of schizophrenia. Schizophr
Bull 20: 441–451.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
23
Research on schizophrenia since the 1970s is conspicuous for its “shift to the left,”
emphasizing earlier phases of schizophrenia. This shift toward first-episode studies
replaced earlier cross-sectionally designed neurobiological studies of mostly those
with chronic schizophrenia, thus minimizing the confounds of chronicity and treat-
ment effects (Keshavan and Schooler, 1992). During the 1990s, attention shifted
further left, underscoring the importance of studying the prodromal phase of
schizophrenia with a glimmer of hope for secondary, and possibly primary preven-
tion (McGorry, 1998). Neurodevelopmental models of schizophrenia (Murray and
Lewis, 1987; Weinberger, 1987) have spurred the characterization of the premorbid
phase of schizophrenia. Studies of the premorbid phase help us to identify neu-
rodevelopmentally mediated vulnerability factors, examine the evolution of the
early phase of schizophrenia, unravel the premorbid pathophysiology without the
potential confounds of illness or treatment effects, and facilitate early diagnosis and
preventive intervention (Cornblatt and Obuchowski, 1997; Ch. 24).
This chapter will review the several approaches to investigate premorbid risk
for schizophrenia. What follows will be a critical appraisal of the existing studies
focusing on the populations at risk for schizophrenia, the issues surrounding study
design, predictive and outcome factors identified so far, and the timing of the
studies. Conceptual and methodological issues and future directions will also be
discussed. Specifically, we will address the following questions. Who are the most
likely individuals that, when studied, will reveal useful insight about the nature
of vulnerability to schizophrenia? How should such studies be designed, and what
are the methodological issues that need to be addressed in these studies? What
neurodevelopmental predictors, and/outcome measures, if studied, are likely to
yield the best insights? When (i.e. during which critical periods of development)
is it most fruitful to assess the risk factors? Focused, hypothesis-driven studies
Neurodevelopment and Schizophrenia, ed. Matcheri S. Keshavan et al. Published by Cambridge University
Press.
C Cambridge University Press 2004.
432
433 High-risk studies, brain development, and schizophrenia
mindful of the above questions and deploying state of the art neurobiological tools
of assessment are likely to provide fresh impetus to this important, rather neglected
field of research. They are reviewed here.
At-risk populations
Studies of premorbid risk refer to the investigation of individuals who are consid-
ered more vulnerable to develop schizophrenia than are individuals in the general
population. Studies of risk for schizophrenia can be retrospective or “follow-back”
investigations (Walker et al., 1993) or prospective. Prospective studies could involve
longitudinal investigation of large cohorts of unselected general populations (such
as birth cohorts) or individuals selected for one or other index of high-risk (HR).
These studies have utilized genetic propensity, neurobehavioral markers, or psy-
chopathology to identify the risk status. In this section, we will outline the potential
merits and disadvantages of these strategies, review the lessons learned from the
early first HR studies and present a rationale for more focused next-generation
studies to examine premorbid risk.
of cases, and the number of predictive variables is often inadequate to address the
specific questions. Second, birth cohort studies involve prolonged follow-up per-
iods and are very expensive, leading to a delay of several decades before gaining
predictive knowledge. However, prospective studies of young adult and adolescent
cohorts (e.g. Davidson et al., 1999) may have an advantage of requiring a shorter
follow-up. Third, birth cohort studies are limited by “cohort effects,” which refer
to variation in illness characteristics (such as age of onset) over time among indi-
viduals defined by shared temporal features such as year of birth. This could lead
to variable findings between studies, and bias in interpretation of data (Di Maggio
et al., 2001). Finally, since large populations are involved, and most cohort studies
were not begun with schizophrenia-related research questions in mind, the data
collected at the outset are not fine-grained enough to ascertain neurodevelopmen-
tal antecedents of schizophrenia with any degree of specificity (Jones and Tarrant,
1999).
New York Infant High Risk 12 HR offspring; 12 controls 1952/> 30 years follow-up Pandysmaturation, predicted outcome Scz, 8%; spectrum PD, 50%
Study (Fish et al., 1992) from infancy
Boston Providence High 118 HR Scz; 126 HR Aff 1959/7 years Low IQ at age 7 associated with risk
Risk Study (Goldstein for Scz
et al., 2000)
Copenhagen High Risk 207 HR offspring; 104 1962/30+ years Poor affective control; psychoticism on Psychosis, 20.8%; spectrum
Project (Parnas et al., controls; mean age 15 years MMPI; institutional care; unstable PD, 21.9%
1993) early rearing
Israel High Risk Study 25 HR; 25 controls; all raised 1964/27 years Poor neurobehavioral functioning; Scz, 8%; Aff, 24%
(Mirsky et al., 1995) in kibbutz; age 8–14 years anxiety; undesirable behavior
Rochester High Risk Study HR Scz 20; HR Aff 38 1972/10+ years follow-up Parental psychopathology and family 52% of offspring with
(Wynne et al., 1987) from infancy variables predict school functioning psychopathology
in the offspring
Jerusalem Infant Study HR Scz 29; other 30; 1973/20+ years Poor neurobehavioral functioning 4.1% Scz among HR Scz;
(Marcus et al., 1987) controls 27 0% in controls
New York High Risk Project 63 HR offspring; 100 1971/20+ years Attentional impairment; poor verbal Psychosis, 18.6%; spectrum
(Erlenmeyer-Kimling controls; 43 psychiatric memory; motor abnormalities; PD, 18.1%
et al., 1995) controls behavioral problems; psychoticism on
MMPI
Finnish Adoptive Family 180 offspring of Scz mothers 1974/20+ years Communication deviance in parents Scz, 5.2%; psychosis, 7.8%
Study (Tienari et al., adopted away; 200 associated with schizophrenia risk
1994) offspring of mentally
well mothers
Edinburgh High Risk Study 162 HR Scz; 36 controls 1994/5+ years followed up Childhood behavioral abnormalities; self 18 of 162 (11%) developed Scz
(Johnstone et al., 2003) from adolescence/early and interviewer rated schizotypy; drug
adulthood abuse; major life events; temporal lobe
volume reductions
HR, high risk; Scz, schizophrenia; PD, personality disorder; Aff, affective disorder; MMPI, Minnesota Multiphasic Personality Inventory.
a Includes only studies with published data pertaining to psychopathological outcome at follow-up.
437 High-risk studies, brain development, and schizophrenia
Table 23.2. Relative advantages and limitations of prospective research designs to assess
developmental premorbid risk
Statistical power/cost − ± ± + +
Feasible number of ± ± ± + +
predictors
Generalizability to + − − ± ±
schizophrenia
Specificity to + + ± − +
schizophrenia
risk of manifesting a schizophrenia outcome (Moldin et al., 1991) and that global
attentional deviance was predictive of the subsequent emergence of schizophrenia
among the HR offspring (Cornblatt and Erlenmeyer-Kimling, 1989). Such an
“enhanced HR strategy” suffers from the difficulty that findings from this design
cannot be generalized to all of schizophrenia because a narrowly defined popula-
tion is studied. However, this approach is likely to have more statistical power to
detect differences from controls and can, therefore, be used to conduct the newer
“second-generation” HR studies involving more expensive neurobiological studies
(Table 23.2).
Methodological issues
As discussed above, all the strategies described seeking to elucidate premorbid
neurodevelopmental risk factors have methodological limitations. Lack of statistical
power, increased cost owing to prolonged follow-up and the use of ill-established
and “dated” predictive measures characterize the HR and birth cohort studies;
the problems of recall bias and the inadequacy of retrospective information are
limitations of retrospective studies. Studies of genetic “ultra-HR” populations such
as discordant monozygotic twins and children of two parents with schizophrenia
are limited by the fact that these are rare populations to find. Other methodological
issues that have plagued the previous HR studies are worth considering.
Specificity to schizophrenia
Several of the early HR studies lacked psychiatric control groups (i.e. offspring of
parents with non-schizophrenic disorders) and so failed to examine the issue of
specificity. It is known that individuals at genetic risk for schizophrenia are at an
increased risk for non-schizophrenic psychiatric disorders (Amminger et al., 1999).
It remains unclear whether this is from a transmitted risk for a broad range of psy-
chopathology or whether non-schizophrenic psychopathology is a precursor, or a
milder manifestation, of schizophrenia. Follow-back studies and prospective birth
441 High-risk studies, brain development, and schizophrenia
cohort studies have shown that individuals with schizophrenia in adult life fre-
quently have prior histories of childhood internalizing and externalizing disorders
(Kim-Cohen et al., 2003). These disorders may also precede other psychiatric dis-
orders. The issue of diagnostic specificity can best be addressed in HR studies by
having appropriate psychiatric control groups.
Generalizability to schizophrenia
The HR studies have often suffered from difficulties in generalizability to
schizophrenia. Children of schizophrenia patients are fewer and paternity may often
be uncertain. For these reasons, only children of female schizophrenia patients were
included in many studies (Fish et al., 1992; Parnas et al., 1993; Tienari et al., 1994).
The etiological factors among offspring of schizophrenia mothers may involve a
higher risk of non-genetic factors (such as alcohol/drug use during pregnancy and
increased perinatal complications) interacting with the genetic factors; therefore,
the causative risk factors may differ depending on which parent had the illness
(Verdoux and Sutter, 2002). Some studies excluded offspring because of lack of
intact families (e.g. Erlenmeyer-Kimling et al., 1995). There are practical diffi-
culties in conducting longitudinal studies of offspring from non-intact families.
Recruiting non-representative samples of HR subjects could lead to loss of critical
information on risk factors, leading to the problem of throwing the “baby out with
the bathwater.”
Table 23.3. Relative advantages and disadvantages of putative neurobehavioral and biological markers
for use in prospective high-risk studies
Cognitive
Attentional impairment + ± + + +
Psychophysiological
Smooth pursuit eye movements + ± + ? ?
P300a − − ± ± ?
P50a + ± ? ? ?
Neurobiological
MRI + ± ± + ±
1
H MRS (NAA) + ± + ± ?
31
P MRS (PME) + ± ? ? ?
MRI, magnetic resonance imaging; MRS, magnetic resonance spectroscopy; NAA, N-acetyl aspartate; PME,
phosphomonoesters; +, good; ± , inconsistent; −, unsatisfactory; ?, data unavailable.
a Components of cognitive evoked response potentials.
and symptoms. An example is social cognitive deficits, which may mediate the
relation between cognitive impairment and psychopathology in predisposition to
schizophrenia (Keshavan and Hogarty, 1999). In this review, we confine ourselves
to a discussion of risk markers in the conventional sense. Further details are also
provided on this issue in Ch. 22.
Predictive measures
Cognitive deficits
The strongest evidence of impairment in relatives of schizophrenia patients appears
to be in sustained attention, abstract thinking, and perceptual motor speed (Kremen
et al., 1994). Among the various neuropsychological measures, the continuous per-
formance test appears to be consistently associated with liability to schizophrenia
(Cornblatt and Keilp, 1994). In the NYHRP, attentional impairment in childhood
predicted 58% of the HR subjects who developed schizophrenia spectrum disor-
ders in adulthood (Erlenmeyer-Kimling, 2000). Attentional impairment is trait
related, stable over time, and related to genetic vulnerability (Michie et al., 2000).
Gross motor skills were also abnormal in 75% of offspring, while false-positive
rates were 27%. Short-term verbal memory was impaired in 83% of offspring who
later developed schizophrenia (Erlenmeyer-Kimling, 2000), showing a high sen-
sitivity but with relatively high false-positive rates (28%). By contrast, attentional
impairments had lower sensitivity (58%) and also lower false-positive rates (18%).
443 High-risk studies, brain development, and schizophrenia
Electrophysiological measures
A physiological measure that has received attention in HR studies is eye track-
ing abnormality (Levy et al., 1994), seen in approximately 50% of adult relatives;
studies of SPEMs in adolescent HR subjects have shown significant dysfunction
compared with healthy comparison subjects (Ross, 2003). However, this measure
has not been investigated as a predictor of schizophrenia risk in prospective stud-
ies. Cognitive evoked response potentials have also been proposed as measures of
liability; prolonged latency and reduced amplitude of N100, P300, and P50 compo-
nents have been observed among relatives (Friedman and Squires-Wheeler, 1994).
Abnormal auditory event potentials (Schreiber et al., 1989) and electrodermal hypo-
or hyper-responsiveness (Dykes et al., 1992; Hollister et al., 1994) have also been
demonstrated, albeit less consistently.
Neurobiological measures
The advent of in vivo structural and physiological neuroimaging studies since the
early 1980s has raised the possibility that altered brain structure and function could
be detected in the premorbid phase of schizophrenia. New in vivo approaches to
examine the brain biology of abnormal neurodevelopment are being developed.
Several studies, including our own (Keshavan et al., 2002a; Lawrie et al., 1999;
Schreiber et al., 1999; Seidman et al., 2002), have shown evidence of structural
brain abnormalities in young relatives of schizophrenia parents. In recent years, two
444 M. S. Keshavan
prospective HR follow-up studies have been initiated, the EHRS and the Pittsburgh
Risk Evaluation Program (PREP). The EHRS has made important observations
regarding the neurobehavioral and neurobiological predictors of the emergence
of psychosis among young HR relatives: these include self- and observer-rated
schizotypy, school behavior abnormalities, and medial temporal volume reduc-
tions (reviewed by Johnstone et al., 2003; Miller et al., 2002). Cross-sectional data
from the PREP have provided preliminary magnetic resonance spectroscopy (MRS)
data suggesting that reductions in N-acetylaspartate (an in vivo marker of neu-
ronal integrity) can be detected in offspring at risk for schizophrenia (Keshavan
et al., 1997). Similar reductions have been observed in adult relatives of schizophre-
nia patients (Callicott et al., 1998). Blood oxygenation level-dependent and contrast
functional magnetic resonance imaging enables abnormal regional brain activation
to be studied in adolescent HR subjects. In a preliminary study, reduced activation
in prefrontal brain regions was seen in HR adolescents during a spatial working
memory task (Keshavan et al., 2002b). In vivo phosphorus MRS studies in HR sub-
jects have shown alterations in membrane phospholipid metabolism (decreased
phosphomonoesters, which are precursors of membrane phospholipids) similar to
those seen in first-episode schizophrenia (Keshavan et al., 2003); similar results have
been observed by Klemm et al. (2001). Overall, data regarding the predictive value
of neuroimaging measures for the development of psychotic symptoms are still
sparse, since HR studies incorporating such measures have started only recently,
and only limited follow-up data are available (Johnstone et al., 2003).
Outcome measures
The choice of outcome measures is also critical in designing HR studies. Devel-
opmental pathways can vary widely between individuals; one specific etiological
factor can lead to diverse psychopathological outcomes (multi-finality), and sev-
eral etiological variables and several developmental pathways can lead to a common
clinical outcome (equi-finality) (Von Bertalanffy, 1968). The failure to identify reli-
able predictors of future illness in previous HR studies may have been related to
the use of narrowly defined schizophrenia as the gold standard in assessing out-
come. The likelihood of the adolescent relatives at risk for developing schizophrenia
actually being diagnosed with schizophrenia during the span of follow-up in these
studies was relatively small; in HR follow-up studies, the schizophrenia-related psy-
chotic disorders (schizophrenia, schizoaffective disorders, and schizophreniform
disorders) occurred in 16–18% of offspring of subjects followed up into adult-
hood (Erlenmeyer-Kimling et al., 1995). However, retrospective analyses as well
as follow-up studies of young at-risk individuals suggest that many individuals
develop behavioral and emotional disturbances that may represent features of a
445 High-risk studies, brain development, and schizophrenia
Conclusions
Studies of HR subjects have revealed impaired attentional and neurobehavioral
abnormalities in individuals at genetic risk for schizophrenia. These observations
are consistent with the neurodevelopmental basis of schizophrenia and may have
some predictive value. Further, these findings provide a good foundation for future
studies; the goal of future HR studies is to elucidate the premorbid neurobiolog-
ical risk for schizophrenia and utilize such knowledge for early identification and
intervention in this illness. The following caveats are worth keeping in mind while
designing such studies.
First, previous HR studies were limited by their expense, as they involved pro-
longed follow-up periods with the likelihood of only a small proportion of the
individuals developing the illness. Table 23.2 outlines the merits and disadvantages
of the various approaches to studying premorbid developmental risk. It is proposed
in this review that an “enhanced” HR strategy that involves a genetically at-risk
population and the use of putative neurobehavioral/psychopathological markers
of risk for further selection of participants may increase the likelihood of iden-
tifying premorbid risk indicators in schizophrenia. Using a narrow approach of
identifying only individuals with a genetic predisposition to schizophrenia is likely
to limit generalizability of HR studies to schizophrenia; an alternative is to define
risk broadly, using both vulnerability indicators (family history, biological markers)
as well as prodromal psychopathological indicators (e.g. “subthreshold” psychotic
and negative symptoms) (Yung et al., 1998). Such individuals are difficult to
447 High-risk studies, brain development, and schizophrenia
1997), also has an onset in late adolescence or early adulthood; obsessive compulsive
disorder has also been recently viewed as a neurodevelopmental disorder (Ch. 21).
Recent advances in developmental neurobiology and neuroscience make it rea-
sonable to expect a paradigm shift in research on schizophrenia. Studies of vul-
nerability as well as protective factors and studies of nature as well as nurture are
needed. It is critical that such research is linked meaningfully to our efforts for early
detection and intervention of this debilitating disorder. It is hoped that the third
millennium will usher in a new generation of research studies on HR populations
and move us closer to piecing together the puzzle of schizophrenia.
Acknowledgements
This work was supported in part by NIMH grants MH 01180, MH 64023, and
MH 45156, and by a NARSAD grant.
REFERENCES
American Psychiatric Association (1994). Diagnostic and Statistical Manual of Mental Disorders,
4th edn. Washington, DC: American Psychiatric Press.
Amminger, G. P., Pape, S., Rock, D., et al. (1999). Relationship between childhood behavioral
disturbance and later schizophrenia in the New York High-risk Project. Am J Psychiatry 156:
525–530.
Brown, A. S., Schaefer, C. A., Wyatt, R. J., et al. (2002). Paternal age and risk of schizophrenia in
adult offspring. Am J Psychiatry 159: 1528–1533.
Brown, R. T., Freeman, W. S., Perrin, J. M., et al. (2001). Prevalence and assessment of attention-
deficit/hyperactivity disorder in primary care settings. Pediatrics 107: E43.
Callicott, J. H., Egan, M. F., Bertolino, A., et al. (1998). Hippocampal N-acetyl aspartate in
unaffected siblings of patients with schizophrenia: a possible intermediate neurobiological
phenotype. Biol Psychiatry 44: 941–950. [Erratum in Biol Psychiatry (1999). 45: 244.]
Cantor-Graae, E., Pedersen, C. B., McNeil, T. F., Mortensen, P. B. (2003). Migration as a risk factor
for schizophrenia: a Danish population-based cohort study. Br J Psychiatry 182: 117–122.
Carter, J. W., Parnas, J., Cannon, T. D., Schulsinger, F., Mednick, S. A. (1999). MMPI variables
predictive of schizophrenia in the Copenhagen High-risk Project: a 25-year follow-up. Acta
Psychiatr Scand 99: 432–440.
Chapman, L. J., Chapman, J. P., Raulin, M. L. (1976). Scales for physical and social anhedonia.
J Abnorm Psychol 85: 374–382.
(1978). Body-image aberration in schizophrenia. J Abnorm Psychol 87: 399–407.
Chapman, L. J., Chapman, J. P., Kwapil, T. R., Eckblad, M., Zinser, M. C. (1994). Putatively
psychosis-prone subjects 10 years later. J Abnorm Psychol 103: 171–183.
Cornblatt, B., Erlenmeyer-Kimling, L. (1989). Attention and schizophrenia. Schizophr Res 2: 58.
449 High-risk studies, brain development, and schizophrenia
Cornblatt, B. A., Keilp, J. G. (1994). Impaired attention, genetics, and the pathophysiology of
schizophrenia. Schizophr Bull 20: 31–46.
Cornblatt, B., Obuchowski, M. (1997). Update of high risk research: 1987–1997. Int Rev Psychiatry
9: 437–447.
David, A. S., Malmberg, A., Brandt, L., Allebeck, P., Lewis, G. (1997). IQ and risk for schizophrenia:
a population-based cohort study. Psychol Med 27: 1311–1323.
Davidson, M., Reichenberg, A., Rabinowitz, J., et al. (1999). Behavioral and intellectual markers
for schizophrenia in apparently healthy male adolescents. Am J Psychiatry 156: 1328–1335.
Di Maggio, C., Martinez, M., Menard, J. F., Petit, M., Thibaut, F. (2001). Evidence of a cohort
effect for age at onset of schizophrenia. Am J Psychiatry 158: 489–492.
Done, D. J., Crow, T. J., Johnstone, E. C., Sacker, A. (1994). Childhood antecedents of schizophre-
nia and affective illness: social adjustments at ages 7 and 11. Br Med J 309: 699–703.
Dworkin, R. H., Cornblatt, B. A., Friedmann, R., et al. (1993). Childhood precursors of affective
vs. social deficits in adolescents at risk for schizophrenia. Schizophr Bull 19: 563–577.
Dykes, K. L., Mednick, S. A., Machon, R. A., Praestholm, J., Parnas, J. (1992). Adult third ventricle
width and infant behavioral arousal in groups at high and low risk for schizophrenia.
Schizophr Res 7: 13–18.
Erlenmeyer-Kimling, L. (2000). Neurobehavioral deficits in offspring of schizophrenic parents:
liability indicators and predictors of illness. Am J Med Genet 97: 65–71.
Erlenmeyer-Kimling, L., Squires-Wheeler, E., Hilldoff-Adamo, U. H., et al. (1995). The New York
High-risk Project. Psychoses and cluster A personality disorders in offspring of schizophrenic
parents at 23 years of follow-up. Arch Gen Psychiatry 52: 857–865.
Feinberg, I. (1982). Schizophrenia and late maturational brain changes in man. Psychopharmacol
Bull 18: 29–31.
Fish, B. (1984). Characteristics and sequelae of the neurointegrative disorder in infants at risk for
schizophrenia: 1952–1982. In Children at Risk for Schizophrenia: A Longitudinal Perspective,
ed. N. F. Watt, E. J. Anthony, L. C. Wynne, J. E. Rolf. New York: Cambridge University Press,
pp. 423–439.
Fish, B., Marcus, J., Hans, S. L., Auerbach, J. G., Perdue, S. (1992). Infants at risk for schizophrenia:
sequelae of a genetic neurointegrative defect. A review and replication analysis of pandys-
maturation in the Jerusalem infant development study. Arch Gen Psychiatry 49: 221–
235.
Friedman, D., Squires-Wheeler, E. (1994). Event-related potentials (ERPs) as indicators for risk
for schizophrenia. Schizophr Bull 20: 63–74.
Garver, D. L. (1987). Methodological issues facing the interpretation of high-risk studies:
biological heterogeneity. Schizophr Bull 13: 525–529.
Goldstein, J. M., Seidman, L. J., Buka, S. L., et al. (2000). Impact of genetic vulnerability and
hypoxia on overall intelligence by age 7 in offspring at high risk for schizophrenia compared
with affective psychoses. Schizophr Bull 26: 323–334.
Gooding, D. C., Iacono, W. G. (1995). Schizophrenia through the lens of a developmental
psychopathology perspective. In Developmental Psychopathology: Risk, Disorder, and
Adaptation, Vol. 2, ed. D. Cicchetti, D. J. Cohen. New York: Wiley, pp. 535–580.
Gottesman, I. I. (1991). Schizophrenia Genesis: The Origins of Madness. New York: W. H. Freeman.
450 M. S. Keshavan
Gottesman, I. I., Erlenmeyer-Kimling, L. (2001). Family and twin strategies as a head start in defin-
ing prodromes and endophenotypes for hypothetical early-interventions in schizophrenia.
Schizophr Res 51: 93–102.
Gottesman, I. I., Shields, J. (1982). Schizophrenia: The Epigenetic Puzzle. New York: Cambridge
University Press.
Gunnell, D., Harrison, G., Rasmussen, F., Fouskakis, D., Tynelius, P. (2002). Associations between
premorbid intellectual performance, early-life exposures and early-onset schizophrenia.
Cohort study. Br J Psychiatry 181: 298–305.
Hafner, H. (1990). New perspectives in the epidemiology of schizophrenia. In Search for the
Causes of Schizophrenia, Vol. II, ed. H. Hafner, W. F. Gattaz. Berlin: Springer-Verlag,
pp. 408–431.
Hanson, D. R., Gottesman, I. I., Heston, L. L. (1990). Long-range schizophrenia forecasting: many
a slip twixt cup and lip. In Risk and Protective Factors in the Development of Psychopathology,
ed. J. Rolf, A. Masten, D. Cicchetti, K. Nuechterlein, S. Weintraub. New York: Cambridge
University Press.
Harrison, P. J., Owen, M. J. (2003). Genes for schizophrenia? Recent findings and their patho-
physiological implications. Lancet 361: 417–419.
Hollister, J. M., Mednick, S. A., Brennan, P., Cannon, T. D. (1994). Impaired autonomic ner-
vous system-habituation in those at genetic risk for schizophrenia. Arch Gen Psychiatry 51:
552–558.
Johnstone, E. C., Russell, K. D., Harrison, L. K., Lawrie, S. M. J. (2003). The Edinburgh High
Risk Study: current status and future prospects. World Psychiatry 2: 45–49.
Jones, P. B., Tarrant, C. J. (1999). Specificity of developmental precursors to schizophrenia and
affective disorders. Schizophr Res 39: 121–125.
Jones, P., Rodgers, B., Murray, R., Marmot, M. (1994). Child developmental risk factors for adult
schizophrenia in the British 1946 birth cohort. Lancet 344: 1398–1402.
Josiassen, R. C., Shagass, C., Roemer, R. A., Straumanis, J. J. (1985). Attention-related effects
on somatosensory evoked potentials in college students at high risk for psychopathology.
J Abnorm Psychiatry 94: 507–518.
Kemppainen, L., Veijola, J., Jokelainen, J., et al. (2001). Birth order and risk for schizophrenia:
a 31-year follow-up of the Northern Finland 1966 Birth Cohort. Acta Psychiatr Scand 104:
148–152.
Kendler, K. S. (2002). Hierarchy and heritability: the role of diagnosis and modeling in psychiatric
genetics. Am J Psychiatry 159: 515–518.
Keshavan, M. S., Hogarty, G. E. (1999). Brain maturational processes and delayed onset in
schizophrenia. Dev Psychopathol 11: 525–543.
Keshavan, M. S., Schooler, N. R. (1992). First-episode studies of schizophrenia: criteria and
characterization. Schizophr Bull 18: 491–513.
Keshavan, M. S., Montrose, D. M., Pierri, J. N., et al. (1997). Magnetic resonance imaging and
spectroscopy in offspring at risk for schizophrenia: preliminary studies. Prog Neuropsy-
chopharmacol Biol Psychiatry 21: 1285–1295.
Keshavan, M. S., Dick, E., Mankowski, I., et al. (2002a). Decreased left amygdala and hippocampal
volumes in young offspring at risk for schizophrenia. Schizophr Res 58: 173–183.
451 High-risk studies, brain development, and schizophrenia
Keshavan, M. S., Diwadkar, V. A., Spencer, S. M., et al. (2002b). A preliminary functional magnetic
resonance imaging study in offspring of schizophrenic parents. Prog Neuropsychopharmacol
Biol Psychiatry 26: 1143–1149.
Keshavan, M. S., Stanley, J. A., Montrose, D. M., Minshew, N. J., Pettegrew, J. W. (2003). Pre-
frontal membrane phospholipid metabolism of child and adolescent offspring at risk for
schizophrenia or schizoaffective disorder: an in vivo (31)P MRS study. Mol Psychiatry 8:
316–323.
Kim-Cohen, J., Caspi, A., Moffitt, T. E., et al. (2003). Prior juvenile diagnoses in adults with
mental disorder: developmental follow-back of a prospective-longitudinal cohort. Arch Gen
Psychiatry 60: 709–717.
Klemm, S., Rzanny, R., Riehemann, S., et al. (2001). Cerebral phosphate metabolism in first-
degree relatives of patients with schizophrenia. Am J Psychiatry 158: 958–960.
Klosterkotter, J., Hellmich, M., Steinmeyer, E. M., Schultze-Lutter, F. (2001). Diagnosing
schizophrenia in the initial prodromal phase. Arch Gen Psychiatry 58: 158–164.
Kremen, W. S., Seidman, L. J., Pepple, J. R., et al. (1994). Neuropsychological risk indicators for
schizophrenia: a review of family studies. Schizophr Bull 20: 103–119.
Kwapil, T. R. (1998). Social anhedonia as a predictor of the development of schizophrenia-
spectrum disorders. J Abnorm Psychol 107: 558–565.
Kwapil, T. R., Miller, M. B., Zinser, M. C., Chapman, J., Chapman, L. J. (1997). Magical ideation
and social anhedonia as predictors of psychosis proneness: a partial replication. J Abnorm
Psychol 106: 491–495.
Lawrie, S. M., Whalley, H., Kestelman, J. N., et al. (1999). Magnetic resonance imaging of
brain in people at high risk of developing schizophrenia. [See comments] Lancet 353:
30–33.
Lawrie, S. M., Byrne, M., Miller, P., et al. (2001). Neurodevelopmental indices and the develop-
ment of psychotic symptoms in subjects at high risk of schizophrenia. Br J Psychiatry 178:
524–530.
Leask, S. J., Done, D. J., Crow, T. J. (2002). Adult psychosis, common childhood infections and
neurological soft signs in a national birth cohort. Br J Psychiatry 181: 387–392.
Lencz, T., Raine, A., Scerbo, A., et al. (1993). Impaired eye tracking in undergraduates with
schizotypal personality disorder. Am J Psychiatry 150: 152–154.
Lencz, T., Cornblatt, B., Bilder, R. M. (2001). Neurodevelopmental models of schizophrenia:
pathophysiologic synthesis and directions for intervention research. Psychopharmacol Bull
35: 95–125.
Lenzenweger, M. F. (1994). Psychometric high-risk paradigm, perceptual aberrations, and
schizotypy: an update. Schizophr Bull 20: 121–135.
Levy, D. L., Holzman, P. S., Matthysse, S., Mendell, N. R. (1994). Eye tracking and schizophrenia:
a selective review. Schizophr Bull 20: 47–62.
Maier, W., Franke, P., Hain, C., Kipp, B., Rist, F. (1992). Neuropsychological indicators of
the vulnerability to schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 16: 703–
715.
Marcus, J., Hans, S. L., Nagler, S., et al. (1987). Review of the NIMH Israeli Kibbutz-City Study
and the Jerusalem Infant Development Study. Schizophr Bull 13: 425–438.
452 M. S. Keshavan
Marcus, J., Hans, S. L., Auerbach, J. G., Auerbach, A. G. (1993). Children at risk for schizophrenia:
the Jerusalem Infant Development Study. II. Neurobehavioral deficits at school age. Arch
Gen Psychiatry 50: 797–809.
McGlashan, T. H. (1998). Early detection and intervention of schizophrenia: rationale and
research. Br J Psychiatry 172: 3–6.
McGorry, P. D. (1998). “A stitch in time” . . . the scope for preventive strategies in early psychosis.
Eur Arch Psychiatry Clin Neurosci 248: 22–31.
McGorry, P. D., Yung, A. R., Phillips, L. J., et al. (2002). Randomized controlled trial of inter-
ventions designed to reduce the risk of progression to first-episode psychosis in a clinical
sample with subthreshold symptoms. Arch Gen Psychiatry 59: 921–928.
McGuffin, P., Farmer, A. E., Gottesman, I. I., Murray, R. M., Reveley, A. M. (1984). Twin concor-
dance for operationally defined schizophrenia. Confirmation of familiality and heritability.
Arch Gen Psychiatry 41: 541–545.
McNeil, T. F., Harty, B., Blennow, G., Cantor-Graae, E. (1993). Neuromotor deviation in offspring
of psychotic mothers: a selective developmental deficiency in two groups of children at
heightened psychiatric risk? J Psychiatr Res 27: 39–54.
Mednick, S. A., Parnas, J., Schulsinger, F. (1987). The Copenhagen High-risk Project, 1962–1986.
Schizophr Bull 13: 485–495.
Michie, P. T., Kent, A., Stienstra, R., et al. (2000). Phenotypic markers as risk factors in
schizophrenia: neurocognitive functions. Aust NZ J Psychiatry 34(Suppl): S74–S85.
Miller, P., Byrne, M., Hodges, A., et al. (2002). Schizotypal components in people at high risk of
developing schizophrenia: early findings from the Edinburgh High Risk Study. Br J Psychiatry
180: 179–184.
Mirsky, A. F., Kugelmass, S., Ingraham, L. J., Frenkel, E., Nathan, M. (1995). Overview
and summary: twenty-five year follow-up of high-risk children. Schizophr Bull 21: 227–
239.
Moldin, S. O., Gottesman, I. I., Rice, J., Erlenmeyer-Kimling, L. (1991). Replicated psychometric
correlates of schizophrenia. Am J Psychiatry 148: 762–767.
Murray, R. M., Lewis, S. W. (1987). Is schizophrenia a neurodevelopmental disorder? [Editorial]
Br Med J (Clin Res Ed) 295: 681–682.
Nasrallah, H. A., Tolbert, H. A. (1997). Neurobiology and neuroplasticity in schizophrenia.
Continuity across the life cycle. [Comment] Arch Gen Psychiatry 54: 913–914.
Niemi, L. T., Suvisaari, J. M., Tuulio-Henriksson, A., Lonnqvist, J. K. (2003). Childhood develop-
mental abnormalities in schizophrenia: evidence from high-risk studies. Schizophr Res 60:
239–258.
Nuechterlein, K. H., Dawson, M. E., Gitlin, M., et al. (1992). Developmental processes in
schizophrenic disorders: longitudinal studies of vulnerability and stress. Schizophr Bull 18:
387–425.
Parnas, J., Cannon, T. D., Jacobsen, B., et al. (1993). Lifetime DSM-III-R diagnostic outcomes
in the offspring of schizophrenic mothers. Results from the Copenhagen High-risk Study.
Arch Gen Psychiatry 50: 707–714.
Pearson, J. S., Kley, I. B. (1957). On the application of genetic expectancies as age specific base
rates in the study of human behavior disorders. Psychol Bull 54: 406–420.
453 High-risk studies, brain development, and schizophrenia
Poulton, R. A. C., Moffitt, T. E., Cannon, M., Murray, R., Harrington, H. (2000). Children’s self-
reported psychotic symptoms and adult schizophreniform disorder: a 15-year longitudinal
study. Arch Gen Psychiatry 57: 1053–1058.
Rantakallio, P., Jones, P. B., Moring, J., von Wendt, L. (1997). Association between central nervous
system infections during childhood and adult onset schizophrenia and other psychoses:
a 28-year follow-up. Int J Epidemiol 26: 837–843.
Reveley, A. M., Reveley, M. A., Clifford, C. A., Murray, R. M. (1982). Cerebral ventricular size in
twins discordant for schizophrenia. Lancet i: 540–541.
Ross, R. G. (2003). Early expression of a pathophysiological feature of schizophrenia: saccadic
intrusions into smooth-pursuit eye movements in school-age children vulnerable to
schizophrenia. J Am Acad Child Adolesc Psychiatry 42: 468–476.
Saitoh, O., Niwa, S., Hiramatsu, K., et al. (1984). Abnormalities in late positive components of
event-related potentials may reflect genetic predisposition to schizophrenia. Biol Psychiatry
19: 293–303.
Sarfati, Y., Hardy-Bayle, M. C. (2002). Could cognitive vulnerability identify high-risk subjects
for schizophrenia? Am J Med Genet 114: 893–897.
Schreiber, H., Stolz, G., Rothmeier, J., Kornhuber, H. H., Born, J. (1989). Prolonged latencies of
the N2 and P3 of the auditory event-related potential in children at risk for schizophrenia.
A preliminary report. Eur Arch Psychiatry Neurol Sci 238: 185–188.
Schreiber, H., Baur-Seack, K., Kornhuber, H. H., et al. (1999). Brain morphology in adolescents
at genetic risk for schizophrenia assessed by qualitative and quantitative magnetic resonance
imaging. Schizophr Res 40: 81–84.
Seidman, L. J., Faraone, S. V., Goldstein, J. M., et al. (2002). Left hippocampal volume as a
vulnerability indicator for schizophrenia: a magnetic resonance imaging morphometric
study of nonpsychotic first-degree relatives. Arch Gen Psychiatry 59: 839–849.
Siever, L. J. (1994). Biologic factors in schizotypal personal disorders. Acta Psychiatr Scand 384:
45–50.
Suddath, R. L., Christison, G. W., Torrey, E. F., Casanova, M. F., Weinberger, D. R. (1990).
Anatomical abnormalities in the brains of monozygotic twins discordant for schizophrenia.
N Engl J Med 322: 789–794.
Tienari, P., Wynne, L. C., Moring, J., et al. (1994). The Finnish adoptive family study of
schizophrenia: implications for family research. Br J Psychiatry 164: 20–26.
Venables, P. H., Mednick, S. A., Schulsinger, S. F., et al. (1978). Screening for risk in mental
illness. In Cognitive Defects in the Development of Mental Illness, ed. G. M. Serban. New York:
Brunner/Mazel, pp. 273–303.
Verdoux, H., Sutter, A. L. (2002). Perinatal risk factors for schizophrenia: diagnostic specificity
and relationships with maternal psychopathology. Am J Med Genet 114: 898–905.
Von Bertalanffy, K. (1968). General Systems Theory. New York: Brazilier.
Wahlbeck, K., Forsen, T., Osmond, C., Barker, D. J., Eriksson, J. G. (2001). Association of
schizophrenia with low maternal body mass index, small size at birth, and thinness during
childhood. Arch Gen Psychiatry 58: 48–52.
Walker, E., Lewine, R. (1990). Prediction of adult onset schizophrenia from childhood movies of
patients. Am J Psychiatry 147: 1052–1056.
454 M. S. Keshavan
Walker, E., Grimes, K., Davis, D., Smith, A. (1993). Childhood precursors of schizophrenia: facial
expressions of emotion. Am J Psychiatry 150: 1654–1660.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
Weinberger, D. R., DeLisi, L. E., Neophytides, A. N., Wyatt, R. J. (1981). Familial aspects of CT
scan abnormalities in chronic schizophrenic patients. Psychiatry Res 4: 65–71.
Weiser, M., Reichenberg, A., Rabinowitz, J., et al. (2001). Association between nonpsychotic
psychiatric diagnoses in adolescent males and subsequent onset of schizophrenia. Arch Gen
Psychiatry 58: 959–964.
Wynne, L. C., Cole, R. E., Perkins, P. (1987). University of Rochester child and family study: risk
research in progress. Schizophr Bull 13: 463–476.
Yung, A. R., Phillips, L. J., McGorry, P. D., et al. (1998). Prediction of psychosis. A step towards
indicated prevention of schizophrenia. Br J Psychiatry Suppl 172: 14–20.
Yung, A. R., Phillips, L. J., Yuen, H. P., et al. (2003). Psychosis prediction: 12-month follow up of
a high-risk (“prodromal”) group. Schizophr Res 60: 21–32.
24
455
456 M. S. Keshavan and B. A. Cornblatt
Table 24.1. Phases of early schizophrenia outlining the clinical features, pathophysiology, etiology, and
hypothesis-based preventive interventions
date back to early development (Done et al., 1994; Jones and Cannon, 1998). In
particular, impaired cognition in areas such as attention and working memory
appear to be early enduring core characteristics of schizophrenia, which support
the involvement of pre- or perinatal brain abnormalities (Cornblatt and Malhotra,
2001; Elvevag and Goldberg, 2000; Erlenmeyer-Kimling et al., 1995). These early
deficits have been increasingly viewed as signs of an underlying vulnerability to
illness that is dormant clinically (in terms of psychosis) but, nevertheless, suggests
a fulminating process that will eventually lead to schizophrenia. This is one of
the primary justifications for early intervention, since it is currently believed that
preemptive treatment may stop, or at least reduce, the emergence of psychosis.
Neural network modeling studies also support the view that schizophrenia
may be associated with synaptic or axonal pruning, but not neuronal cell death
(Hoffman and McGlashan, 1997; McGlashan and Hoffman, 2000). Neuropatholog-
ical studies show reduction in the synapse-rich neuropil and a consequent increase
in cortical neuron density (Selemon et al., 1995). Reductions have also been reported
in the expression of synaptophysin, a synaptic marker (Eastwood and Harrison,
1995; Glantz and Lewis, 1997), and in the density of dendritic spines (Garey et al.,
1998; Ch. 17). These observations suggest an overall reduction of cortical synapse-
rich neuropil in schizophrenia. This may lead to reduced neuronal plasticity. The
net effect of reduced neuropil may, therefore, be a compromised neuronal capacity
to modulate the normal academic, familial, and interpersonal demands of adoles-
cence. When a critical threshold of such neuropil loss is exceeded, symptoms and
signs of schizophrenia may appear.
could, therefore, decrease tonic prefrontal dopamine release, and increase phasic
stress-induced mesolimbic dopamine release (Moghaddam, 2002). A dysregula-
tion of the tonic–phasic dopamine system may account for the positive and negative
symptoms of schizophrenia (Grace, 1993).
Excess phasic release of glutamate during recurrent psychotic exacerbations can
cause excitotoxic cell or dendrite loss via NMDA receptor-mediated calcium influx
into cells, or oxidative stress (Goff and Coyle, 2001). This may explain the neurode-
teriorative changes that may occur in some patients with untreated schizophrenic
illness.
It may be seen from the above discussion that glutamate may play a role in
each of the characteristic components of schizophrenic illness: premorbid cogni-
tive deficits, adolescent onset of the illness, and early deterioration. Glutamater-
gic dysfunction may serve as a unitary explanation that links these observations
together. The schizophrenia syndrome may result from the cumulative effect of a
possibly genetically mediated hypoplasia in early and late maturational processes
of brain development interacting with adverse psychosocial factors during adoles-
cence and early adulthood and possible neurotoxic effects of untreated psychosis
early in the schizophrenic illness. The premorbid vulnerability to schizophrenia
may be caused by multiple genetic and environmental factors interacting to affect
early brain development. The adolescent onset of the disorder may be determined
by late brain maturational processes, as well as the stresses unique to adolescence.
It is likely that early brain developmental defects (e.g. neuromigrational errors or
defective neuronal proliferation) may predispose to excessive postnatal synaptic
pruning and/or neuronal apoptosis, leading to the schizophrenic endophenotype.
The development of a deficit state and of persistent psychopathology after illness
onset may be determined by the possible neurotoxic effect of continuing psychotic
illness.
Excitotoxic neuronal loss, interacting with genetic factors, may result from early
brain adversity (viruses, malnutrition, or perinatal trauma) and lead to selective
loss of NMDA receptor-bearing glutamatergic neurons in cortical and subcortical
structures. This would account for the premorbid neurocognitive abnormalities
seen among children and adolescents at risk for schizophrenia. The normative
synaptic pruning process that sets in around late childhood and adolescence may
interact with the state of reduced tonic glutamatergic neurotransmission, perhaps
acting via NMDA receptors, to result in a net excess of synapse elimination in
the cortical and subcortical structures. Reduced activity in the corticostriatal gluta-
matergic neurons would lead to diminished tonic cortical dopamine release because
of reduced mesocortical dopamine release, which comes “on line” during adoles-
cence. Recent data (Scott et al., 2002) have suggested that NMDA agonists may shift
462 M. S. Keshavan and B. A. Cornblatt
the balance of dopamine signaling toward the D1 receptor and away from the D2
receptors, by recruiting D1 receptors to the plasma membrane; this may account for
the therapeutic effects of glutamatergic agonists such as glycine and d-cycloserine
in improving negative symptoms and cognitive deficits in schizophrenia. The inter-
personal and cognitive deficits could lead to an inability to handle the psychosocial
stresses unique to adolescence and early adulthood (Keshavan and Hogarty, 1999).
Such stresses, in turn, may lead to an upregulation of the subcortical dopaminergic
neurons; the behavioral response to stress-induced phasic glutamate and dopamine
release is, therefore, likely to be exaggerated, leading to positive psychotic symptoms.
If untreated, persistent dopaminergic, and consequent phasic glutamatergic, excess
could lead to further excitotoxic brain damage, perhaps by increasing oxidative
stress.
Other similar, integrative models view schizophrenia as a lifetime disorder of
brain development, plasticity, and aging (DeLisi, 1997; Lieberman et al., 1997; Olney
and Farber, 1995). In this chapter, we attempt to extend these models by proposing
that the successive premorbid, morbid, and post-onset pathology of schizophrenia
results from a glutamatergic dysfunction at critical windows of vulnerability during
early and late developmental phases, and the early course of this illness. Each of the
steps of dysfunction in glutamatergic functioning in this model could increase the
risk for the subsequent step, leading to a pathophysiological cascade.
also take their toll. Finally, adolescence is the period when substance use becomes
increasingly prevalent and this may interact with pre-existing risk to lead to psy-
chopathology.
At least a subgroup of schizophrenia patients may have progressive brain deteri-
orative processes following illness onset. Proposed mechanisms for such neurode-
generation include oxidative stress (Coyle, 1996) or neurochemical sensitization
from repeated exposure to neurochemical stressors (Lieberman et al., 1997). How-
ever, one needs also to entertain the possibility that the observed neurobiological
changes during the course of the illness may be plastic adaptations of the nervous
system to being psychotic or cognitively impoverished (Weinberger and McClure,
2002).
Non-antipsychotic medications
Neurodevelopmental models, discussed above, have also suggested therapeutic
trials of drugs with alternative putative (non-dopamine-related) mechanisms of
action. Berger et al. (2004) have been conducting a 1 year trial with low-dose
lithium, the definitive results of which are expected soon. Using predictions
derived from the glutamatergic and membrane models of schizophrenia, respect-
ively, Woods et al. (2002) have conducted a 2 month trial of glycine and a 3 month
study with ethyl eicosopentaenoic acid, an omega-3 fatty acid. Outcome measures
have included psychopathology ratings and rates of conversion into psychosis, cog-
nitive assessments, neuroimaging, and in vitro measures of structural cortical and
cell membrane integrity. The results are awaited.
Psychotherapeutic treatments
It has been suggested (Ch. 20) that cognitive decline has largely occurred in
schizophrenia by the time of the first psychotic presentation. Therefore, a window
of opportunity exists during the early prodromal period, which is characterized
by non-specific behavioral signs and precedes the more schizophrenia-like fea-
tures of the late prodromal phase. The German Schizophrenia Network has been
conducting a 12 month non-pharmacological intervention of up to 26 sessions of
cognitive behavioral therapy over 6 months plus monitoring compared with moni-
toring alone in a group of prodromal patients (Morrison et al., 2002). Bechdolf and
467 Developmental models and early interventions
Acknowledgements
This work was supported in part by NIMH grants MH 45156 and MH 64023.
468 M. S. Keshavan and B. A. Cornblatt
REFERENCES
Akbarian, S., Bunney, W. E., Potkin, S. G., et al. (1993). Altered distribution of nicotamine-
adenine dinucleotide phosphate-diaphorase cells in frontal lobe of schizophrenics implies
disturbances of cortical development. Arch Gen Psychiatry 50: 169–177.
Akbarian, S., Sucher, N., Bradley, D., et al. (1996). Selective alterations in gene expression for
NMDA receptor subunits in prefrontal cortex of schizophrenics. J Neurosci 16: 19–30.
Andreasen, N., Rezai, K., Alliger, R., et al. (1992). Hypofrontality in neuroleptic-naive patients
with chronic schizophrenia. Assessment with xenon 133 single-photon emission computed
tomography and the Tower of London. Arch Gen Psychiatry 49: 943–958.
Arnt, J., Skarsfeldt, T. (1998). Do novel antipsychotics have similar pharmacological characteris-
tics? A review of the evidence. Neuropsychopharmacology 18: 63–101.
Bechdolf, A., Wagner, M. (2003). Cognitive behavioral therapy in the prepsychotic phase: an
exploratory study. Schizophr Res 60(Suppl.): 319.
Behar, T. N., Scott, C. A., Greene, C. L., et al. (1999). Glutamate acting at NMDA receptors
stimulates embryonic cortical neuronal migration. J Neurosci 19: 4449–4461.
Berger, G. E., Profitt, T. M., McConchie, M. A., et al. (2004). Ethyl-eicosapentaenoic acid (E-EPA)
supplementation in early psychosis, Schizophr Res 67: 7.
Cannon, T. D., Huttunen, M. O., Dahlstrom, M., et al. (2002). Antipsychotic drug treatment in
the prodromal phase of schizophrenia. Am J Psychiatry 159: 1230–1232.
Christodolou, G. N. (1991). Prevention of psychopathology with early interventions. Psychother
Psychosom 55: 201–207.
Chugani, H. T., Phelps, M. E., Mazziotta, J. C. (1987). Positron-emission tomography study of
human brain functional development. Ann Neurol 22: 487–497.
Cornblatt, B. A., Malhotra, A. K. (2001). Impaired attention as an endophenotype for molecular
genetic studies of schizophrenia. Am J Med Genet 105: 11–15.
Cornblatt, B., Lencz, T., Correll, C., Author, A., Smith, C. (2002). Treating the prodrome natu-
ralistic findings from the RAP Program. Acta Psychiatr Scand 106(S413): 44.
Cornblatt, B. A., Lencz, T., Smith, C. W., et al. (2003). The schizophrenia program revisited: a
neurodevelopmental perspective. Schizophr Bull 29: 633–651.
Coyle, J. T. (1996). The glutamatergic dysfunction hypothesis for schizophrenia. Harvard Rev
Psychiatry 3: 241–253.
Cuijpers, P. (2003). Examining the effects of prevention programs on the incidence of new cases
of mental disorders: the lack of statistical power. Am J Psychiatry 160: 1385–1391.
DeLisi, L. E. (1995). A prospective follow-up study of brain morphology and cognition in first-
episode schizophrenic patients: preliminary findings. Biol Psychiatry 38: 349–360.
(1997). Is schizophrenia a lifetime disorder of brain plasticity, growth, and aging? Schizophr
Res 23: 119–129.
Deutsch, S. I., Rosse, R. B., Schwartz, B. L., Mastropaolo, J. (2001). A revised excitotoxic hypothesis
of schizophrenia: therapeutic implications. Clin Neuropharmacol 24: 43–49.
Done, D. J., Crow, T. J., Johnstone, E. C., Sacker, A. (1994). Childhood antecedents of
schizophrenia and affective illness: social adjustment at ages 7 and 11. Br Med J 309: 699–
703.
469 Developmental models and early interventions
Eastwood, S. L., Harrison, P. J. (1995). Decreased synaptophysin in the medical temporal lobe in
schizophrenia demonstrated using immunoautoradiography. Neuroscience 69: 339–343.
Eaton, W. W., Thara, R., Federman, B., Melton, B., Liang, K. Y. (1995). Structure and course of
positive and negative symptoms in schizophrenia. Arch Gen Psychiatry 52: 127–134.
Elvevag, B., Goldberg, T. E. (2000). Cognitive impairment in schizophrenia is the core of the
disorder. Crit Rev Neurobiol 14: 1–21.
Erlenmeyer-Kimling, L., Cornblatt, B. A. (1992). A summary of attentional findings in the New
York High-risk Project. J Psychiatr Res 26: 405–426.
Erlenmeyer-Kimling, L., Squires-Wheeler, E., Hilldoff-Adamo, U. H., et al. (1995). The New York
High-risk Project. Psychoses and cluster A personality disorders in offspring of schizophrenic
parents at 23 years of follow-up. Arch Gen Psychiatry 52: 857–865.
Feinberg, I. (1982–83). Schizophrenia: caused by a fault in programmed synaptic elimination
during adolescence? J Psychiatr Res 17: 319–334.
(1990). Cortical pruning and the development of schizophrenia. Schizophr Bull 16: 567–570.
Fenton, W. S., McGlashan, T. H. (1994). Antecedents, symptom progression, and long-term
outcome of the deficit syndrome in schizophrenia. Am J Psychiatry 151: 351–356.
Fish, B. (1987). Infant predictors of the longitudinal course of schizophrenic development.
Schizophr Bull 13: 395–409.
Garey, L. J., Ong, W. Y., Patel, T. S., et al. (1998). Reduced dendritic spines density on cerebral
cortical pyramidal cells in schizophrenia. J Neurol Neurosurg Psychiatry 65: 446–453.
Glantz, L. A., Lewis, D. A. (1997). Reduction of synaptophysin immunoreactivity in the pre-
frontal cortex of subjects with schizophrenia: Regional and diagnostic specificity. Arch Gen
Psychiatry 54: 943–952.
Goff, D. C., Coyle, J. T. (2001). The emerging role of glutamate in the pathophysiology and
treatment of schizophrenia. Am J Psychiatry 158: 1367–1377.
Grace, A. (1993). Cortical regulation of subcortical dopamine systems and its possible relevance
to schizophrenia. J Neur Transm (Gen Sect) 91: 111–134.
Haas, G., Sweeney, J. (1992). Premorbid and onset features of first-episode schizophrenia.
Schizophr Bull 18: 373–386.
Hafner, H., Maurer, K., Loffler, W., Riecher-Rossler, A. (1993). The influence of age and sex on
the early course of schizophrenia. Br J Psychiatry 162: 80–86.
Heresco-Levy, U., Javitt, D. (1998). The role of N-methyl-d-aspartate (NMDA) receptor-mediated
neurotransmission in the pathophysiology and therapeutics of psychiatric syndromes. Eur
Neuropsychopharmacol 8: 141–152.
Hoffman, R., McGlashan, T. (1997). Synaptic elimination, neurodevelopment, and the mecha-
nism of hallucinated “voices” in schizophrenia. Am J Psychiatry 154: 1683–1689.
Jaskiw, G. E., Juliano, D. M., Goldberg,T. E., et al. (1994). Cerebral ventricular enlargement in
schizophreniform disorder does not progress. A seven year follow-up study. Schizophr Res
14: 23–28.
Javitt, D., Zukin, S. (1991). Recent advances in the phencyclidine model of schizophrenia. Am J
Psychiatry 148: 1301–1308.
Jernigan, T. L., Tallal, P. (1990). Late childhood changes in brain morphology observable with
MRI. Dev Med Child Neurol 32: 379–385.
470 M. S. Keshavan and B. A. Cornblatt
Johnstone, E. C., Russell, K. D., Harrison, L. K., Lawrie, S. M. J. (2003). The Edinburgh High
Risk Study: current status and future prospects. World Psychiatry 2: 45–49.
Jones, P., Cannon, M. (1998). The new epidemiology of schizophrenia. Psychiatr Clin North Am
21: 1–25.
Keshavan, M., Anderson, S., Pettegrew, J. (1994). Is schizophrenia due to excesive synaptic
pruning in the prefrontal cortex? The Feinberg hypothesis revisited. J Psychiatr Res 28:
239–265.
Keshavan, M. S. Development, disease and degeneration in schizophrenia: a unitary pathophys-
iological model. (1999). J Psychiatr Res 33: 513–521.
Keshavan, M. S., Hogarty, G. E. (1999). Brain maturational processes and delayed onset in
schizophrenia. Dev Psychopathol 11: 525–543.
Keshavan, M. S., Reynolds, C. F., Miewald, M. J., et al. (1998a). Delta sleep deficit in schizophrenia:
evidence from automated analyses of sleep data. Arch Gen Psychiatry 55: 443–448.
Keshavan, M., Haas, G., Kahn, C., et al. (1998b). Superior temporal gyrus and the course of early
schizophrenia: progressive, static, or reversible? J Psychiatr Res 32: 60–65.
Keshavan, M. S., Stanley, J. A., Montrose, D. M., Minshew, N. J., Pettegrew, J. W. (2003). Pre-
frontal membrane phospholipid metabolism of child and adolescent offspring at risk for
schizophrenia or schizoaffective disorder: an in vivo (31)P MRS study. Mol Psychiatry 8:
316–323.
Kim, J., Kornhuber, H., Schmid-Burgk, W., Holzmuller, B. (1980). Low cerebrospinal fluid glu-
tamate in schizophrenic patients and a new hypothesis on schizophrenia. Neurosci Lett 20:
379–382.
Kovelman, J., Scheibel, A. (1984). A neurohistological correlate of schizophrenia. Biol Psychiatry
19: 1601–1621.
Kraepelin, E. (1919). Dementia Praecox and Paraphrenia. Edinburgh: E. and S. Livingstone.
Lieberman, J. (1993). Prediction of outcome in first-episode schizophrenia. J Clin Psychiatry
54(Suppl.): 13–17.
Lieberman, J., Koreen, A., Chakos, M., et al. (1996). Factors influencing treatment response and
outcome of first-episode schizophrenia: implications for understanding the pathophysiology
of schizophrenia. J Clin Psychiatry 9: 5–9.
Lieberman, J., Sheitman, B., Kinon, B. (1997). Neurochemical sensitization in the pathophys-
iology of schiozphrenia: deficits and dysfunction in neuronal regulation and plasticity.
Neuropsychopharmacology 17: 205–229.
Lieberman, J., Chakos, M., Wu, H., et al. (2001). Longitudinal study of brain morphology in first
episode schizophrenia. Biol Psychiatry 49: 487–499.
Loebel, A. D., Liberman, J. A., Alvir, J. M. J. (1992). Duration of psychosis and outcome in
first-episode schiozphrenia. Am J Psychiatry 149: 1183–1188.
Mathalon, D. H., Ford, J. M., Rosenbloom, M., Pfefferbaum, A. (2000). P300 reduction and
prolongation with illness duration in schizophrenia. Biol Psychiatry 47: 413–427.
McGlashan, T., Fenton, W. (1993). Subtype progression and pathophysiologic deterioration in
early schizophrenia. Schizophr Bull 19: 71–84.
McGlashan, T. H., Hoffman, R. E. (2000). Schizophrenia as a disorder of developmentally reduced
synaptic connectivity. Arch Gen Psychiatry 57: 637–648.
471 Developmental models and early interventions
McGlashan, T. H., Zipursky R. B., Perkins, D., et al. (2003a). The PRIME North America ran-
domized double-blind clinical trial of olanzapine versus placebo in patients at risk of being
prodromally symptomatic for psychosis. I. Study rationale and design. Schizophr Res 61:
7–18.
(2003b). Olanzapine versus placebo treatment of the schizophrenia prodrome: one year results.
Schizophr Res 60(Suppl.): 295.
McGorry, P. D., Yung, A. R., Phillips, L. J., et al. (2002). Randomized controlled trial of inter-
ventions designed to reduce the risk of progression to first-episode psychosis in a clinical
sample with subthreshold symptoms. Arch Gen Psychiatry 59: 921–928.
McGrath, J. (2000). Universal interventions for the primary prevention of schizophrenia. Aust N
Z J Psychiatry 34(Suppl.): S58–S64.
Meltzer, H. (1994). An overview of the mechanism of action of clozapine. J Clin Psychiatry 55:
47–52.
Mednick, S. A., Machon, R. A., Huttunen, M. O. (1988). Adult schizophrenia following prenatal
exposure to an influenza epidemic. Arch Gen Psychiatry 45: 171–176.
Moghaddam, B. (2002). Stress activation of glutamate neurotransmission in the prefrontal cor-
tex: implications for dopamine-associated psychiatric disorders. Biol Psychiatry 51: 775–
787.
Morrison, A. P., Bentall, R. P., French, P., et al. (2002). Randomised controlled trial of early
detection and cognitive therapy for preventing transition to psychosis in high-risk individ-
uals. Study design and interim analysis of transition rate and psychological risk factors. Br
J Psychiatry Suppl 43: S78–S84.
Mrazek, P. J., Haggerty R. J. (1994). Reducing Risks for Mental Disorders: Frontiers for Preventive
Intervention Research. Washington, DC: National Academy Press.
Murray, R. M., Lewis, S. W. (1987). Is schizophrenia a neurodevelopmental disorder? [Editorial]
Br Med J (Clin Res Ed) 295: 681–682.
Olney, J., Farber, N. (1995). Glutamate receptor dysfunction and schizophrenia. Arch Gen Psy-
chiatry 52: 998–1007.
Pettegrew, J. W., Keshavan, M. S., Panchalingam, K., et al. (1991). Alterations in brain high-
energy phosphate and membrane phospholipid metabolism in first-episode, drug-naive
schizophrenics. Arch Gen Psychiatry 48: 563–568.
Pettegrew, J. W., McClure, R., Keshavan, M., et al. (1997). 31 P magnetic resonance spectroscopy
studies of developing brain. In Neurodevelopment and Adult Psychopathology, ed. M. Kesha-
van, R. Murray. New York: Cambridge University Press, pp. 71–92.
Ruhrmann, S., Wagner, M., Wieneke, A., et al. (2002). Intervention in the initial phase of psychosis.
Acta Psychiatr Scand 106(S413): 19.
Scott, L., Kruse, M. S., Forssberg, H., et al. (2002). Selective up-regulation of dopamine D1
receptors in dendritic spines by NMDA receptor activation. Proc Natl Acad Sci USA 99:
1661–1664.
Selemon, J., Rajkowska, G., Goldman-Rakic, P. (1995). Abnormally high neuronal density in the
schizophrenic cortex. Arch Gen Psychiatry, 52: 805–818.
Susser, E. S., Lin, S. P. (1992). Schizophrenia after prenatal exposure to the Dutch Hunger Winter
of 1944–1945. Arch Gen Psychiatry 49: 983–988.
472 M. S. Keshavan and B. A. Cornblatt
Tamminga, C. A. (1998). Schizophrenia and glutamatergic transmission. Crit Rev Neurobiol 12:
21–36.
Thompson, P. M., Vidal, C., Giedd, J. N., et al. (2001). Mapping adolescent brain change reveals
dynamic wave of accelerated gray matter loss in very early-onset schizophrenia. Proc Natl
Acad Sci USA 98: 11650–11655.
Tsai, G., Passani, L., Slusher, B., et al. (1995). Abnormal excitatory neurotransmitter metabolism
in schizophrenia brains. Arch Gen Psychiatry 52: 829–836.
Weinberger, D. R. (1987). Implications of normal brain development for the pathogenesis of
schizophrenia. Arch Gen Psychiatry 44: 660–669.
(1995). From neuropathology to neurodevelopment. Lancet 346: 552–557.
Weinberger, D. R., McClure, R. K. (2002). Neurotoxicity, neuroplasticity, and magnetic resonance
imaging morphometry: what is happening in the schizophrenic brain? Arch Gen Psychiatry
59: 553–558.
Woods, S. W., D’Souza, D. C., Wexler, B. E., Hoffman, R. E., McGlashan, T. H. (2002). Novel
early interventions for prodromal states. Acta Psychiatr Scand 106(S413): 12.
Woods, S. W., Breier, A., Zipursky, R. B., et al. (2003). Randomized trial of olanzapine versus
placebo in the symptomatic acute treatment of the schizophrenic prodrome. Biol Psychiatry
54: 453–464.
Wyatt, R. (1991). Neuroleptics and the natural course of schizophrenia. Schizophr Bull 17: 325–
351.
Zipursky, R. O. (1992). Widespread cerebral gray matter volume deficits in schizophrenia. Arch
Gen Psychiatry 49: 195–205.
Index
473
474 Index