Libro Viro Inmuno
Libro Viro Inmuno
Dynamics
of Immune
Activation in
Viral Diseases
Dynamics of Immune Activation in Viral
Diseases
Pallaval Veera Bramhachari
Editor
Dynamics of Immune
Activation in Viral Diseases
Editor
Pallaval Veera Bramhachari
Department of Biotechnology
Krishna University
Machilipatnam, Andhra Pradesh, India
This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
Preface
Viral infectious diseases signify an essential portion of global public health concern
with millions of deaths annually. Viruses produce disease directly by killing the host
cells they infect or by liberating toxins that can cause tissue damage and functional
derangements in neighboring or distant cells and tissues. In the field of viral immu-
nology, the researchers continue to make noteworthy and exhilarating contributions
to our understanding of the fundamental biology of the immune system. Yet the
practical translational applications of this fascinating and enthralling area of science
are little disappointing with regard to the recurrent viral outbreaks.
The development of an infectious disease in an individual involves complex
interactions between the virus and the host. These interactions are a dynamic inter-
play of host mechanisms aimed at eliminating infections and viral strategies
designed to permit survival in the face of beneficial activation of innate and adaptive
immune responses. Different types of infectious viral agents stimulate distinct types
of immune responses and have evolved unique mechanisms for evading immunity.
Noteworthy, in many instances, virus-induced proteins produced by emerging
viruses can trickily antagonize specific immune recognition mechanisms or circum-
vent cell intrinsic restriction factors, thus influencing the host antiviral immune
responses and complicating the viral replication kinetics. However, there is little
understanding of within-host immunological processes underlying the reservoir
host virus interactions, and this question is hardly addressed in several emerging
viral diseases. Moreover, the viral genome itself plays an imperative role in the
rapid evasion of adaptive immune responses, with the generation of diverse viral
quasi-species that is an inherent consequence of error-prone RNA replication mech-
anisms. Moreover, elite immune controllers do not eliminate the virus from their
system, because of the eventual emergence of virus variants that manage to escape
the determined efforts of the immune system.
From serious pandemics and highly contagious infections to common influenza
episodes, clinical prognosis often relies on early detection of the infectious agent.
The recently developed viral diagnostic methods are reshaping the field of clinical
microbiology and could contribute to reducing the prevalence of serious infectious
diseases. Fortunately, the aspects of modernization that help drive pathogen emer-
gence can also propel scientific innovation and current significant scientific advances
have warranted our skill set to address the challenge of emerging viruses. From
advanced genomic sequencing to novel methods in structural biology, we now have
v
vi Preface
an increasingly classy toolkit with which to facilitate the detection and possible
control of emerging viral diseases. Still, current outbreaks of viral diseases serve as
powerful reminders of our ongoing vulnerability to emerging viral pathogens.
Nonetheless, a number of additional viral diseases are considered for inclusion in
the priority list every year. These events underscore the need for concerted efforts to
develop and implement new interventions while continuing to invest in proven pub-
lic health measures.
The field of viral immunology seeks to understand the mechanisms of virus–host
interaction with a view of applying this knowledge to the design of effective vac-
cines to control viral infections. Whereas several viral infections are still in need of
successful prophylactic vaccines, in many instances we also require therapeutic
vaccines that could boost inadequate immunity. This book primarily emphasizes
several areas of the field that hold substantial promise for translation, but where
further work is critically required to find solutions. We emphasize that our funda-
mental understanding of virus–host relationships is moving in leaps and bounds, but
we lag behind in applying this knowledge to the successful control of many viral
infections. Nevertheless, elucidating the nature of immune responses in individual
natural hosts may inform our understanding of how virus–host equilibria are estab-
lished without substantially impacting host health. Furthermore, this may provide
insights into the mechanisms of disease pathogenesis and immunity in humans.
We strongly believe that this book would provide enough insights into the cur-
rent understanding of adaptive and innate immune response in viral diseases. This
book is an attempt to compile the novel information available on recent advance-
ments on various aspects of differential regulation of each immune cell during
viral pathogenesis and immune evasion. This book aims to revitalize the interac-
tion between fields of virology and immunology in order to advance our under-
standing of dynamics of viral immune pathogenesis, as well as innate and adaptive
immune responses elicited by the host. The book also elucidates a comprehensive
yet representative description of a large number of challenges associated with
immune sensors, namely TLRs, DNA and RNA sensors, and other immune cells
during viral infections, as it is indispensable to possess updated information on
emerging viral diseases. This book could be an essential reading for the novice
and experts in the field of viral immunology, immune interventions, and viral
immunodiagnostics, including latest developments in vaccine research. With
these objectives in mind, the content of this textbook has been arranged in a logi-
cal progression from fundamental to more advanced concepts. Finally, this book
also outlines the most advanced immune techniques used in diagnostics of viral
diseases and also primarily focuses on advancements of vaccine development
research for emerging viral diseases.
We hope that this book stimulates your creativity and wish you success in your
experiments. This book is a stunning reflection of the seriousness with which the
several scientific minds are dedicated to the welfare of the scientific community. I
am extremely thankful to the contributors for paying continuous attention to my
request and showing faith in my capabilities. I shall always remain highly obliged
to all of them forever. These words cannot justify the worthiness of their efforts.
Preface vii
We successfully compiled our creative and thoughtful research work due to gen-
uine concern and painstaking effort of many more well-wishers whose names are
not mentioned, but they are still in our heart. So, the reward is surely worth for their
efforts. I want to dedicate this book to my mother, S. Jayaprada (late). Myself and
the contributing authors hope from the bottom of our hearts that this book will be a
good guidebook and compass for research studies in diagnostic virology and
immunotechnology.
My sincere thanks are extended to all the academicians and scientists who have
contributed the galaxy of topics in the form of chapters and happily agreed to share
their work on Dynamics of Immune Activation in Infectious Viral Diseases in this
volume.
This book is a stunning reflection of the seriousness with which the several sci-
entific minds are dedicated from the immunology and virology scientific commu-
nity. I am extremely thankful to the contributors for paying continuous attention to
my requests and showing obsolete faith in my competencies and capabilities. I shall
always remain highly obliged and indebted to all of them forever. These words can-
not justify the worthiness of their untiring efforts. We appreciate the excellent work
of the authors and coauthors who were invited to contribute diversified chapters in
this book. The credit for making this book a reality goes to them. Me as an editor
and the review team for the chapters especially appreciate sharing expertise with the
contributors. Each chapter is informative and written as a stand-alone chapter, so the
reader can begin anywhere in the book depending upon his or her interests and
needs.
At the same time, I also express my deepest gratitude to my family members,
especially my wife (Ramadevi Ramaswamy) and my kids (Ruthvik and Jayati), for
their kind support which has prompted me to complete the assignment on time. I am
also thankful to the Department of Biotechnology, Krishna University, for the sup-
port. I am equally thankful to the Springer Nature Publishing group for their full
cooperation during the peer review and production of the volume.
I am thankful to my beloved teachers and mentors for their constant support and
motivations at all stages of progress.
ix
About the Book
xi
Contents
xiii
xiv Contents
xv
xvi Editor and Contributors
Contributors
xix
xx Abbreviations
CMV Cytomegalovirus
CNS Central nervous system
CP Classical pathway
CpG Unmethylated deoxycytidylate-phosphate-deoxyguanylate
CPPs Cell-penetrating peptides
CS Complement system
CSF Colony-stimulating factor
CTL Cytotoxic T lymphocytes
DAA Directly acting antivirals
DAI Z-DNA binding protein
DAMPS Damage-associated molecular patterns
DCs Dendritic cells
DDX DExD/H-box
DDX41 DEAD box polypeptide 41
DENV Dengue virus
DHF Dengue hemorrhagic fever
DIV Dog immunodeficiency virus
DNAse Deoxyribonuclease
DSS Dengue shock syndrome
EBOV Ebola virus
EBV Epstein–Barr virus
ECL Electrochemiluminescence
EGF Epidermal growth factor
EPO Leptin and erythropoietin
Fab Fragment antigen binding
Fc Fragment crystalline
FEB Field Effect Biosensing
FGF Fibroblast growth factor
FIV Feline immunodeficiency virus
FO Follicular
FPIA Fluorescence polarization immunoassay
GC Germinal center
G-CSF Granulocyte CSF
GEFs Guanine nucleotide exchange factors
GH Growth hormone
GM-CSF Granulocyte macrophage colony-stimulating factor
HA Hemagglutinin
HBcAg Hepatitis B core antigen
HBsAg Hepatitis B surface antigen
HCC Hepatocellular carcinoma
HCV Hepatitis C
HeV Hendra virus
HFD Hemagglutinin fusion domain
HIN H-inversion
HIV Human immunodeficiency virus
Abbreviations xxi
RNAse Ribonuclease
RT-LAMP Reverse transcription loop-mediated isothermal amplification
RT-RPA Reverse transcription recombinase polymerase amplification
assay
SARM Sterile-alpha and Armadillo motif-containing protein
SARS-CoV Severe Acute Respiratory Syndrome Coronavirus
SCF Stem cell factor
SGPs Small G-protein
SIV Simian immunodeficiency virus
SNPs Single nucleotide polymorphisms
SOCS3 Suppressor of cytokine signaling 3
SPR Surface plasma resonance spectroscopy
ssRNA Single-stranded RNA
STAT1 Signal transducer and activator of transcription 1
STING Stimulator of interferon genes
Syk Spleen tyrosine kinase
TBK1 TANK binding kinase 1
TEM Transmission electron microscopy
TFH cells Typically follicular T helper cells
TGF Transforming growth factor
TIR Toll/IL-1 receptor
TLRs Toll-like receptors
TNF Tumor necrosis factor
TNP T-cell epitope of nucleoprotein
TPR Tetra tricopeptide
TRAF TNF receptor-associated factor
TRAIL TNF-related apoptosis-inducing ligand
Tregs T regulatory cells
TREX1 Three-primer repair endonuclease 1
TRIF TIR domain-containing adaptor protein-inducing IFN-β
TYK2 Tyrosine kinase2
VEEV Venezuelan equine encephalitis virus
VEGF Vascular endothelial growth factor
VHF Viral hemorrhage fever
VHSV Viral hemorrhagic septicemia virus
VSV Vesicular stomatitis virus
WNV West Nile virus
YFV Yellow fever virus
ZIKV Zika virus
Part I
Role of Innate Immunity in Combating Viral
Diseases
Significance and Dynamics of Immune
Responses During Viral Pathogenesis 1
Pallaval Veera Bramhachari
Abstract
Immune system is a homeostatic system which is active against numerous invad-
ing pathogens. Host immune system uses multiple immune responses (innate,
adaptive, and complement system) to eliminate virus/viral particles. Additionally
immune system is also well resourced with sensors that detect invading patho-
gens and direct responses for clearing numerous copies of virus recruits. Modern
technologies help prevent pathogen emergence as well as thrust scientific
improvements in understanding the viral immune responses; moreover, recent
scientific advances in diagnostic virology have undeniably transformed the abil-
ity to address challenges of numerous emerging intricate viruses. The journey of
diagnostic virology has started from serology, nucleic acid sequence-based
amplification techniques, and genomic sequencing techniques to most advanced
innovative methods (e.g., structural biology spectroscopy, NGS, microfluidics,
metagenomics, CRISPR/Cas system, nanotechnology and structural biology),
the world progressed way beyond with several classy diagnostic methods to help
tackle the diagnosis and control of emerging viral diseases. However, the techni-
cal competencies alone are inadequate if not sustained by health promotion strat-
egies to raise awareness of the significance of early detection and diagnosis,
outbreak, and spread of virus. Yet, current outbreaks of viral diseases across the
world dole out authoritative reminders to emerging viral pathogens.
Keywords
Immune system · Viruses · Diagnostic methods · Emerging viral diseases
P. V. Bramhachari (*)
Department of Biotechnology, Krishna University, Machilipatnam, Andhra Pradesh, India
1.1 Introduction
Infectious diseases are reported to account for ~20% of global mortality, of which
one-third of deaths are caused by viral diseases. The viral infections pose major public
health risks and warrant further research and development, including surveillance and
diagnostics. All the lethal viral diseases present an absurdity in mounting the patho-
genesis there by killing their hosts, which is noticeably of no advantage to the virus.
Viruses are infectious agents consisting of nucleic acids coated in a simple protein
casing, infect, replicate in host cells, and cause acute, chronic infections. Since viral
infection is an intricate and highly vibrant process, noticeably affected by the physical
and chemical environment, studies into infectious viral biology ought to preferably
occur in advanced research settings. Viruses replicate by hijacking the host cell’s
machinery and making host cells a huge virus factory and erupt as virus after… virus
after… virus after… virus.
The immune system is also well resourced with sensors that detect invading
pathogens and direct responses for clearing numerous copies of virus recruits. On
the entry of virus, immune system of the host typically elicits both nonspecific
innate and “specific” adaptive immune responses against foreign pathogens.
Activation of varied immune responses, time, and the extent of response rely on
how virus interacts and spreads within host cells. Host immune system uses multi-
ple immune responses (innate, adaptive, and complement system) to eliminate
virus/viral particles. The immune system employs most efficient mechanisms
depending on the distinctiveness of infectious agents. Diverse actions occur during
viral infections, equally toward free viral particles in addition to infected cells.
Race between virus and immune response establishes: whether the intruder will
eliminate or establish a persistent infection. The host cell can also be damaged
unswervingly by virus or by viral immune response. However, balance between
good and bad antiviral immune response relies on the amount of viral load, chronic-
ity of infection, and magnitude of tissues infected (Zinkernagel 1996). Therefore, a
balance exists between immune activation vis-à-vis immune suppression during the
setting of chronic infection. Viruses evolved numerous strategies to escape immune
system to institute a chronic infection. Few viruses endure in definite cell types and
hide from immune system, while some encode specific genes that target infected
cells or immune system. A few viruses limit their replication, thus restricting acces-
sibility of antigen to alert the immune system. However, a few viruses are error
prone in their replication, escalating the possibility of making escape viral mutants.
Nonetheless, these immune evasion strategies of virus are unprejudiced by regula-
tion within the immune system (Finlay and McFadden 2006).
During early stages of infection “innate” response limits virus multiplication, which
implicates synthesis of diffusible proteins called interferons, cytokines, and chemo-
kines and stimulation of “natural killer” and dendritic lymphocytes (French and
Yokoyama 2003). In order to limit viral replication, first line of defense system has
1 Significance and Dynamics of Immune Responses During Viral Pathogenesis 5
Another important second line of antiviral response is adaptive immune system that
includes CD8+ and CD4+ T cells and neutralizing antibodies which act against viral
particles and infected cells in combination. Days to weeks are essential to mount an
adaptive immune response tailored for specific virus. Adaptive immune response
possesses two components: humoral response (utilities virus-specific antibodies by
B lymphocytes) and cell-mediated response (specific cytotoxic T lymphocytes
(CTL) that kill infected cells) (Kim et al. 1999). Both the components of adaptive
defense system ensue production of long-lived “memory cells” that allow much
more quick response for consequent infection with similar virus (Campos and
Godson 2003). Antibodies are most vital mechanisms against viral particles, while
cytotoxic mechanisms are noteworthy against infected cells.
surfaces (e.g., the gastrointestinal and respiratory tracts). Antibody can neutralize
virus by (1) preventing host cell—virus interactions or (2) distinguishing viral anti-
gens on infected cells that results in antibody-dependent cytotoxic cells (ADCC) or
complement-mediated lysis (CML) (Parkin and Cohen 2001; Chaplin 2010).
1.4 Significance
Viruses are intracellular pathogens that invade and infect host cells. Immune system
is a homeostatic system which is active against numerous invading pathogens inside
the body to clear infections. Propitiously, the aspects of recent modernization tech-
nologies that helped prevent pathogen emergence can also impel scientific improve-
ments; moreover, recent scientific advances in diagnostic virology undeniably
transformed the ability to address the challenges of numerous emerging intricate
1 Significance and Dynamics of Immune Responses During Viral Pathogenesis 7
viruses across the globe. Starting from serology, nucleic acid sequence-based ampli-
fication techniques, genomic sequencing techniques to most advanced innovative
methods (structural biology spectroscopy, NGS, microfluidics, metagenomics,
CRISPR/Cas system, nanotechnology, and structural biology), the world has pro-
gressed way beyond with more classy diagnostic methods to help tackle the detec-
tion and control of emerging viral diseases. However, the technical competencies
alone are inadequate if not sustained by health promotion strategies to raise aware-
ness of the significance of early detection, outbreak, and spread of virus. Still, cur-
rent outbreaks of viral diseases across the world dole out powerful reminders of our
ongoing susceptibility to emerging viral pathogens. Reflecting the diversity of
viruses and viral diseases, these are notoriously difficult drug targets since they
modify and adapt themselves quickly to build up resistance and emerge as new
serotypes. Nonetheless, better perceptiveness of host–pathogen interactions, viral
protein, and nucleic acid functions led to additional rational drug designs, resulting
in important therapeutic advances against viral diseases. Furthermore, novel plat-
forms for vaccine design, namely nanoparticles and virus-like particles, have
embarked avenues for development of new vaccine targets. Treatment strategies for
viral infections comprise hindering binding of virus to host cells by inhabiting on
host cell receptor with an additional molecule, or use of vaccine developed for a
particular virus or an analogous target of diverse viruses. Serious actions should
accentuate the call for strenuous efforts to develop and execute novel interventions
in viral disease diagnosis and vaccine development keeping public health perspec-
tive in view.
Dynamics of Immune Activation in Viral Diseases is an authoritative reference
book in virology, which provides the current understanding of adaptive and innate
immune response in viral diseases. This book also illustrates about differential regu-
lation of each immune cells during viral pathogenesis and immune evasion. This
book aims to revitalize the interaction between fields of virology and immunology
in order to advance our understanding of dynamics of viral immune pathogenesis,
as well as innate and adaptive immune responses elicited by host. Recent advance-
ments in immune intervention and viral immunodiagnostics, including latest devel-
opments in vaccine research, will be discussed. It also covers new areas of immune
biology where innate part of immune system helps adaptive part through cross talk
with adaptive immune system. This book primarily emphasizes on the recent chal-
lenges of immune sensors, namely TLRs, DNA and RNA sensors, and other immune
cells during viral infections, as it is indispensable to possess updated information on
emerging viral diseases. Apart from the current understanding of immune response
in human viral diseases, this book also outlines the most advanced immune tech-
niques used in diagnostics of viral diseases and also primarily focuses on advance-
ments of vaccine development research for emerging viral diseases.
Acknowledgement DR. PVBC is grateful to Krishna University, Machilipatnam, for the support
extended.
References
Belardelli F, Ferrantini M (2002) Cytokines as a link between innate and adaptive antitumor immu-
nity. Trends Immunol 23(4):201–208
Campos M, Godson DL (2003) The effectiveness and limitations of immune memory: understand-
ing protective immune responses. Int J Parasitol 33(5–6):655–661
Chaplin DD (2010) Overview of the immune response. J Allergy Clin Immunol 125(2):S3–S23
Finlay BB, McFadden G (2006) Anti-immunology: evasion of the host immune system by bacte-
rial and viral pathogens. Cell 124(4):767–782
French AR, Yokoyama WM (2003) Natural killer cells and viral infections. Curr Opin Immunol
15(1):45–51
Haller O, Kochs G, Weber F (2006) The interferon response circuit: induction and suppression by
pathogenic viruses. Virology 344(1):119–130
Kim JJ, Nottingham LK, Tsai A, Lee DJ, Maguire HC, Oh J, Dentchev T, Manson KH, Wyand MS,
Agadjanyan MG, Ugen KE, Weiner DB (1999) Antigen-specific humoral and cellular immune
responses can be modulated in rhesus macaques through the use of IFN-γ, IL-12, or IL-18 gene
adjuvants. J Med Primatol 28(4–5):214–223
Mogensen TH (2009) Pathogen recognition and inflammatory signaling in innate immune
defenses. Clin Microbiol Rev 22(2):240–273
Ou R, Zhang M, Huang L, Moskophidis D (2008) Control of virus-specific CD8+ T-cell exhaus-
tion and immune-mediated pathology by E3 ubiquitin ligase Cbl-b during chronic viral infec-
tion. J Virol 82(7):3353–3368
Paladino P, Cummings DT, Noyce RS, Mossman KL (2006) The IFN-independent response to
virus particle entry provides a first line of antiviral defense that is independent of TLRs and
retinoic acid-inducible gene I. J Immunol 177(11):8008–8016
Parkin J, Cohen B (2001) An overview of the immune system. Lancet 357(9270):1777–1789
Paul S, Lal G (2017) The molecular mechanism of natural killer cells function and its importance
in cancer immunotherapy. Front Immunol 8:1124
Rosendahl Huber S, van Beek J, de Jonge J, Luytjes W, van Baarle D (2014) T cell responses to
viral infections–opportunities for peptide vaccination. Front Immunol 5:171
Shayakhmetov DM, Di Paolo NC, Mossman KL (2010) Recognition of virus infection and innate
host responses to viral gene therapy vectors. Mol Ther 18(8):1422–1429
Sun JC, Lanier LL (2009) Natural killer cells remember: an evolutionary bridge between innate
and adaptive immunity? Eur J Immunol 39(8):2059–2064
Thimme R, Binder M, Bartenschlager R (2012) Failure of innate and adaptive immune responses
in controlling hepatitis C virus infection. FEMS Microbiol Rev 36(3):663–683
Zinkernagel RM (1996) Immunology taught by viruses. Science 271(5246):173–178
Antibody-Dependent Enhancement
of Viral Infections 2
Ruta Kulkarni
Abstract
Antiviral antibodies constitute an important component of the host immune
response against viral infections and serve to neutralize and reduce infectivity of
the virus. However, these antibodies, intended to protect the host, may some-
times prove beneficial to the virus, by facilitating viral entry and replication in
the target cell. This phenomenon, known as antibody-dependent enhancement
(ADE) of infection, is a result of interaction of virus–antibody immune com-
plexes with Fcγ and/or complement receptors on certain types of host cells and
promotes viral entry into the host cells. The internalized immune complexes then
modulate host immune response so as to enhance viral replication and aggravate
disease severity. The possibility of induction of ADE remains a concern in the
development and implementation of viral vaccines and immunotherapeutics.
Keywords
Viral infection · Neutralization · Antibody-dependent enhancement · Immune
response · Vaccine · Immunotherapeutic
2.1 Introduction
R. Kulkarni (*)
Department of Communicable Diseases, Interactive Research School for Health Affairs
(IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
growth of the pathogen and consequently exacerbates disease in the host. This phe-
nomenon has far-reaching implications for disease control and prevention, as thera-
peutic antibodies deployed to protect the host may aid the pathogen instead.
Similarly, antibodies induced by vaccination may actually increase the risk and/or
severity of disease in subsequent host–pathogen encounters.
This chapter aims to provide insights into the current knowledge on ADE of viral
infections, with a focus on its molecular mechanisms and contribution to viral
pathogenesis and disease, as well as implications for disease control strategies.
Fig. 2.1 Schematic representation of effect of antibody on virus–host cell interaction. (a) In the
absence of virus-specific antibody, virus enters the host cell via interaction with cell surface (virus-
specific) receptor, followed by replication and release of progeny virions. (b) In the presence of
non-neutralizing or sub-neutralizing concentrations of antibody, an additional pathway of entry
into host cell is available to the virus. Virus–antibody immune complexes are internalized via anti-
body interaction with cell surface Fc receptor (FcR), resulting in antibody-dependent enhancement
(ADE) of viral infection and replication. Internalized immune complexes bring about suppression
of cellular innate antiviral immune response further boosting viral replication. (c) In the presence
of neutralizing concentrations of antibody, virus attachment and entry into host cell is blocked
Table 2.1 Summary of key features of antibody-dependent enhancement (ADE) of viral infections
Virus (family) Mechanism of ADE Clinical significance of ADE Implications for vaccines
Dengue virus 1. Antibodies against viral EDI, EDII, prM 1. Association of ADE with severe 1. Increased risk of severe dengue among
(family proteins involved in ADE dengue (DHF/DSS) during secondary seronegative Dengvaxia (CYD-TDV)
Flaviviridae) 2. Extrinsic ADE: FcR-mediated virus heterotypic infections has been vaccine recipients led to WHO
internalization of virus–antibody immune demonstrated recommending adoption of a pre-vaccination
complexes into monocytes, macrophages, 2. Severe disease during primary screening strategy, and vaccine
dendritic cells infections among infants born to administration only to dengue seropositives
3. Intrinsic ADE: Modulation of host dengue-immune mothers has been 2. Identification of stronger correlates of
antiviral response by LILR-B1 co-ligation, attributed to ADE protection, development of more robust
downregulation of TLR-dependent, RIG-I/ assays for estimation of neutralizing/
MDA5 signaling pathways, induction of enhancing ability, long-term follow-up of
IL-10 production vaccine recipients in clinical trials suggested
4. Antibody-mediated enhancement of viral to better predict vaccine-related ADE
fusion suggested 3. Subunit EDIII, NS1 vaccines, chimeric
5. ADE associated with massive release of vaccines with replaced DENV pr gene,
inflammatory cytokines leading to alteration T-cell response-inducing vaccines suggested
of vascular permeability and plasma leakage to reduce ADE, but in vivo protection not yet
which are the hallmarks of severe dengue demonstrated
Human Antibodies against viral gp41 and gp120 Positive correlation of enhancing ADE indicated by observation of higher
immunodeficiency enhance viral entry into T cells, monocytes/ antibody level with plasma viral load infection rate/risk of infection among
virus (family macrophages, and granulocytic cells, by the and negative correlation with CD4 vaccine recipients in AIDSVAX, RV144
Retroviridae) following mechanisms: cell count suggest association of ADE clinical trials, but not yet confirmed
1. Complement-mediated, C-ADE: with accelerated immunosuppression
Interaction of antibody-opsonized virus with and disease progression
complement component C3d,g, cellular
complement receptor CR2 and virus-specific
receptor CD4, co-receptor CXCR4
2. FcR-mediated, FcR-ADE: By CD4-
dependent or independent process
R. Kulkarni
2
Influenza virus 1. Antibodies against viral hemagglutinin Increased risk of medically attended ADE suggested to be responsible for
(family (HA) and neuraminidase (NA) mediate virus illness among individuals with prior vaccine-associated enhanced respiratory
Orthomyxoviridae) uptake via FcRs into macrophages, possibly influenza-like illness during 1918 disease in immunized pigs and ferrets, but
leading to increased antigen presentation and pandemic, and among seasonal mechanism not yet clearly understood
T-cell activation influenza vaccine recipients during
2. Enhancement of viral fusion by anti-HA2 2009 H1N1 pandemic, is suggestive
antibodies suggested of ADE
Respiratory Antibodies against viral glycoproteins G and Clinical relevance remains unclear in Enhanced disease in formalin-inactivated
syncytial virus F mediate virus uptake via FcRs into light of contradictory reports RSV vaccine recipients initially attributed to
(family monocytes, macrophages, dendritic cells regarding association of maternal ADE, but other mechanisms also recently
Paramyxoviridae) leading to immune response modulation antibody-induced ADE with severe suggested
disease in infants
Ebola virus (family Antibodies against viral glycoprotein (GP) Clinical relevance not yet understood Vaccine-related ADE not yet demonstrated,
Filoviridae) promote virus internalization by FcR- but remains a concern. Avoiding induction of
mediated or complement component C1q/ known infectivity-enhancing antibodies,
C1q receptor-mediated process into while retaining T-cell epitopes in vaccines
monocytes/macrophages, endothelial, proposed for ADE mitigation
epithelial cells, and hepatocytes
SARS— Antibodies against viral spike (S) Clinical relevance still debated Impact on vaccine safety not yet understood
Coronavirus glycoprotein mediate virus uptake via FcRs
Antibody-Dependent Enhancement of Viral Infections
as “extrinsic ADE.” However, studies on Ross River Virus suggested that internal-
ized immune complexes further serve to enhance viral replication by suppression of
the cellular innate antiviral immune responses (Lidbury and Mahalingam 2000;
Suhrbier and La Linn 2003). This mechanism is termed as “intrinsic ADE” and has
been subsequently observed in flaviviruses as well (Chareonsirisuthigul et al. 2007;
Ubol et al. 2010). Thus, ADE is a complex phenomenon comprising of extrinsic and
intrinsic components, which together contribute to augmentation of viral infection
and replication. The consequence of this increased virus production is the massive
release of inflammatory and vasoactive mediators by host cells, which ultimately
leads to exacerbation of viral pathogenesis and disease severity.
Prior sensitization of the humoral immune response is a prerequisite for ADE
(Halstead 2003; Taylor et al. 2015). This phenomenon is thus widely observed dur-
ing secondary infection with a heterotypic virus of the same genus, wherein preex-
isting antibodies against the primary (sensitizing) infection bind to the (secondary)
virus, but fail to neutralize it. The pathogenicity and outcome of secondary infection
is influenced by the time interval between primary/secondary infections, with
increasing time being associated with more severe disease, and may be explained by
the waning of broadly neutralizing antibodies over time. Passively acquired anti-
bodies are also capable of inducing ADE, as indicated by the enhancement of viral
disease by preexisting maternal antibody in infants born to dengue-immune mothers
(Kliks et al. 1988; Chau et al. 2008, 2009).
The ADE phenomenon has implications for the use of antiviral immunoglobulins
as therapy against viral infection, due to the associated risk of enhancement of dis-
ease (Taylor et al. 2015). ADE also poses a major challenge for implementation of
vaccination programs, as vaccine-induced antibodies may enhance subsequent viral
infection, thus placing vaccine recipients at increased risk of severe disease. Indeed,
concerns regarding the safety of the world’s first licensed dengue vaccine
“Dengvaxia” have forced authorities to reconsider the mass vaccination strategy
and issue specific recommendations for safe implementation of the vaccine in den-
gue control programs (Wilder-Smith et al. 2019).
ADE has been exploited by a variety of viruses belonging to different virus fami-
lies. The mechanism and clinical significance of ADE of selected viruses will be
reviewed in this chapter and are also summarized in Table 2.1. The ADE phenom-
enon has been most extensively studied in dengue virus and will be considered in
detail here.
and the EDIII (lateral ridge) region involved in cellular attachment. These antibod-
ies are mostly serotype-specific; however, antibodies raised against the quaternary
“envelope dimer epitope” (EDE) have been found to potently neutralize all four
DENV serotypes (Dejnirattisai et al. 2015). Such protective antibodies represent
only a small subset of the antibody repertoire in dengue patients, with the immune
response being dominated by cross-reactive, non-neutralizing antibodies targeting
EDI, EDII (fusion loop) and prM, which have the potential to enhance viral infec-
tion (Smith et al. 2014). Moreover, all neutralizing antibodies are also capable of
enhancing infection when present at sub-neutralizing concentration.
The role of antibodies against prM protein is particularly important because of its
ability to enhance infection of immature virions, which are otherwise noninfectious.
The prM protein, present in the intracellular immature virus particle, covers the
fusion domain of the E protein, thus preventing its fusion with the host cell mem-
brane. Cleavage of the precursor (pr) peptide from the prM protein during the pro-
cess of virus maturation renders the released mature virus particle “infectious” to
host cells for further rounds of replication (Li et al. 2008; Junjhon et al. 2010).
Inefficient processing of the prM protein results in the production of immature or
partially immature virus particles, which are normally noninfectious due to their
inability to fuse with host cell. However, studies using human monoclonal antibod-
ies obtained from persons with secondary dengue have revealed that these immature
virus particles are capable of infecting host cells in the presence of the non-
neutralizing anti-prM antibodies, and are thus infectious during secondary infec-
tions with the possibility of contributing to severe disease (Dejnirattisai et al. 2010;
Schmidt 2010). Similarly, antibodies specific to the fusion loop (FL) in EDII have
also been shown to enhance the infectivity of immature DENV particles (Rodenhuis-
Zybert et al. 2011).
inflammatory cytokines and other chemical mediators at high levels (Pang et al.
2007). ADE of DENV infection and replication results in increased presentation of
viral antigens on the surface of infected cells, leading to activation and proliferation
of T cells sensitized during a prior infection. This results in release of cytokines
such as IFN-γ, TNF-α, IL-2, IL-6, IL1-β, IL-8 and immunoregulators such as
IL-12p70, which directly act upon the vascular endothelial cells resulting in plasma
leakage, the hallmark of severe dengue. Increased levels of such cytokines have
been associated with plasma leakage in severe dengue patients (Chaturvedi et al.
2000), while effect of the cytokine response on alteration of vascular permeability
has been demonstrated in vitro and in mice models, with disruption of the apical
junction complexes in endothelial cells being the suggested mechanism (Dewi et al.
2004; Appanna et al. 2012; Puerta-Guardo et al. 2013).
ADE has also been reported in other viruses of family Flaviviridae including
Murray Valley Encephalitis virus, Japanese encephalitis virus (JEV), West Nile
virus (WNV), yellow fever virus (YFV), and Zika virus (ZIKV) of genus Flavivirus,
as well as hepatitis C virus (HCV), of genus Hepacivirus (Hawkes and Lafferty
1967; Cardosa et al. 1983; Gould and Buckley 1989; Meyer et al. 2008; Bardina
et al. 2017). Further, infection with one Flavivirus has also been demonstrated to
enhance subsequent infection with another virus of the same genus, as observed for
DENV-ZIKV in in vitro studies, mice models, and rhesus macaques (Bardina et al.
2017; George et al. 2017). Such DENV-ZIKV cross-enhancement has also been
observed in primary human PBMCs (Li et al. 2018); however, in the absence of
clinical or epidemiological evidence, the clinical relevance of these findings is
unknown. Interestingly, a beneficial effect of cross-enhancement among flaviviruses
has been reported recently in a clinical trial, with preexisting JEV vaccine-induced
antibodies enhancing immunogenicity of subsequently administered YFV vaccine
(Chan et al. 2016).
ADE of HIV infection was first reported in the 1980s, shortly after the identification
and isolation of HIV, and two different mechanisms, namely, complement-mediated
ADE (C-ADE) (Robinson Jr. et al. 1987, 1988) and FcR-mediated ADE (FcR-ADE)
(Takeda et al. 1988; Homsy et al. 1989), were described. Through either of these
mechanisms, virus-specific antibodies present in sera of HIV-infected individuals
were shown to enhance viral entry and in some cases replication within T cells,
monocytes/macrophages, and granulocytic cells (Beck et al. 2008). Further, both
these mechanisms have also been shown to enhance HIV infection of human syncy-
tiotrophoblast cells, thus suggesting contribution of ADE to materno-fetal transmis-
sion (Tóth et al. 1994). Both these forms of ADE are mediated by antibodies
targeting the viral surface glycoproteins gp41 and gp120 (Trischmann et al. 1995;
Takada and Kawaoka 2003). Such enhancing antibodies have been detected in 72%
of HIV patients (Subbramanian et al. 2002).
C-ADE of HIV infection has been well characterized through in vitro studies in
T-cell lines such as MT-2 and SupT1/R5, showing enhancement of cell line-adapted
HIV strains or primary virus isolates by human monoclonal antibodies or sera from
HIV-infected individuals, and has been associated with increased synthesis of viral
RNA and protein and enhanced release of infectious virions (Robinson Jr. et al.
1989, 1990a, b; Robinson Jr. 2006; Willey et al. 2011). Such C-ADE is primarily
mediated by antibodies targeting the N-terminal immunodominant domain of viral
gp41. Antibody binding to gp41 initiates the complement cascade leading to deposi-
tion of complement component C3d,g on the virion. Interaction of this opsonized
virus with the cell surface complement receptor type 2 (CR2) facilitates virus
attachment and internalization. Engagement of the HIV receptor CD4 and co-
receptor CXCR4 is also required for this mode of virus uptake. Increased attach-
ment of the virus to target cell, rather than CR2-mediated signaling, has been
suggested to be responsible for enhancement of infection through C-ADE. An alter-
native route of complement component C1q binding followed by interaction with
C1q receptor has also been suggested for C-ADE (Prohaszka et al. 1997).
FcR-ADE has been demonstrated in vitro mainly in monocyte/macrophage cell
lines and primary cultures, with all three classes of FcγR, namely, FcγRI, FcγRII,
and FcγRIII, reported to support ADE (Homsy et al. 1989; Takeda et al. 1988, 1990;
Laurence et al. 1990; Connor et al. 1991; Trischmann et al. 1995). ADE by FcγRI
2 Antibody-Dependent Enhancement of Viral Infections 25
and FcγRII requires viral interaction with CD4, indicating that the FcRs facilitate
virus entry by potentiating attachment to CD4 receptors. Alternatively, FcR-
mediated endocytosis of virus–antibody complexes followed by intracellular fusion
with endosomal membrane occurring as a result of binding of virus with CD4 recep-
tors on the endosomal membranes has also been suggested. FcγRIII-mediated ADE
has been shown to be CD4-independent. FcR-ADE assumes particular importance
in the viral life cycle as it mediates infection of macrophages, which are important
for maintaining a viral reservoir during persistent infections.
Another mechanism of complement-independent, FcR-independent ADE,
mediated by antibodies specific to viral gp120, has been described (Sullivan et al.
1998; Guillon et al. 2002). Antibody binding has been shown to modulate interac-
tion of gp120 with HIV co-receptor CCR5 or induce conformational changes in
gp120, leading to its activation and subsequent promotion of membrane fusion.
Further, FcαR-mediated ADE has also been reported, with human serum IgA
showing low-level enhancement of HIV replication in vitro (Janoff et al. 1995;
Kozlowski et al. 1995).
An important feature of HIV is the rapid viral evolution during chronic infection.
While neutralizing antibodies are known to exert a selection pressure facilitating
emergence of neutralization escape mutants, enhancing antibodies have been sug-
gested to favor emergence of ADE-susceptible variants. Indeed, at later stages of
infection, patient sera have demonstrated increased proportion of enhancing anti-
bodies and potential for ADE, while virus strains isolated at such later stage have
exhibited increased susceptibility to ADE in vitro (Davis et al. 2001; Subbramanian
et al. 2002; Willey et al. 2011).
The impact of ADE on HIV vaccine safety is not clearly understood. Studies in
Rhesus monkeys have demonstrated the association of active and passive immuni-
zation with enhanced disease progression on subsequent infection (Staprans et al.
2004; Sholukh et al. 2014). In humans, ADE has been indicated in two vaccine
clinical trials. In the AIDSVAX trial, the rate of HIV infection was observed to be
higher among vaccinees with low (non-protective) antibody responses (Gilbert et al.
2005). In the RV144 trial, positive correlation of vaccine-elicited plasma IgA
responses with risk for HIV acquisition was documented (Shmelkov et al. 2014).
Further, a study investigating the effect of passive immunization of HIV-positive
pregnant women on mother-to-infant transmission suggested the possibility of
antibody-enhanced in utero transmission (Onyango-Makumbi et al. 2011). These
findings are suggestive of vaccine-induced ADE, but cannot confirm the same.
Assays capable of estimating neutralizing and enhancing potential of immune
serum, such as those involving use of complement and complement receptor carry-
ing target cells for C-ADE, have been proposed for a better understanding of the
clinical relevance of vaccine-related ADE. Such assays would also provide stronger
immune correlates during preclinical and clinical studies. Further, design of vaccine
immunogens that will elicit protective and not enhancing antibodies, by avoiding
inclusion of known enhancing epitopes, has been put forth as a mitigation strategy
for HIV-ADE (Beck et al. 2008).
these cells has been shown to be possible in the presence of appropriate protease for
HA cleavage. Increased presentation of viral antigens by the infected macrophages
leading to augmented virus-specific T-cell activation is also suggested to contribute
to viral pathogenesis. Enhancement of virus fusion by anti-HA2 subunit antibodies
is another mechanism of influenza-ADE and has been shown to be responsible for
the occurrence of vaccine-associated enhanced respiratory disease (VAERD) in pigs
immunized with inactivated H1N2 vaccine and challenged with pandemic 2009
H1N1 virus (Gauger et al. 2011; Khurana et al. 2013). Influenza-ADE is also sug-
gested by epidemiologic observations in humans. Retrospective analysis of medical
records from the 1918 influenza pandemic revealed association of prior influenza-
like illness with increased risk of medically attended illness during the pandemic
period (Shanks et al. 2016). More recently, prior receipt of seasonal influenza vac-
cine was found to increase the risk of medically attended pandemic H1N1 illness
during 2009 in Canada (Skowronski et al. 2010). Furthermore, presence of high titer
non-protective serum antibodies was associated with immune complex-mediated
severe H1N1 illness during this pandemic (Monsalvo et al. 2011; To et al. 2012).
Observation of VAERD in pigs and ferrets immunized with inactivated or HA
subunit vaccines raises concern regarding the safety of vaccines, particularly uni-
versal influenza vaccines (Gauger et al. 2011; Khurana et al. 2013; Rajao et al.
2014; Skowronski et al. 2014). However, vaccine-mediated protection and absence
of VAERD is also reported in few studies in pigs, suggesting that vaccine-induced
ADE occurs only in certain conditions, which are not yet clearly understood (Reeth
et al. 2004; Kyriakis et al. 2010; Ricklin et al. 2016). Regarding therapeutic candi-
dates, Phase 2 studies of anti-HA stalk mAbs have indicated possibility of enhanced
viral shedding in some treated human subjects. Screening for high potency protec-
tive antibodies, defining optimal dose range for effective neutralization, using cock-
tails of mAbs with different antigen specificities, has been suggested for mitigation
of ADE (Chan-Hui and Swiderek 2016).
1989; Osiowy et al. 1994). The phenomenon has also been reported in Bonnet mon-
keys developing enhanced disease following FI-RSV immunization (Ponnuraj et al.
2001, 2003). FcγR-mediated internalization of immune complexes is the suggested
mechanism, with antibodies against the viral surface glycoproteins, the attachment
glycoprotein (G) and fusion glycoprotein (F), playing an important role. Although
epithelial cells of the respiratory tract are primary targets of RSV infection, it has
been suggested that alveolar macrophages are infected through ADE, resulting in
activation of Th2 response and increased expression of TNF-α, IL-6, thus causing
enhanced disease (Gimenez et al., 1996). Further, ADE-mediated infection of lung
dendritic cells (DCs) has been demonstrated to negatively modulate DC cell func-
tion, resulting in impaired T-cell activation, and has been suggested to contribute to
RSV pathogenesis (Gomez et al. 2016). However, the clinical effects of RSV-ADE
still remain unclear with different studies reporting contradictory observations
regarding the association of maternal antibody-induced ADE and disease severity in
infants (Chanock et al. 1970; van Erp et al. 2017).
Enhanced RSV disease in FI-RSV vaccine recipients was initially attributed to
vaccine-induced ADE; however, recent studies have suggested a role of other mech-
anisms involving T cells (Huisman et al. 2009). Whether ADE would affect the
efficacy of other RSV vaccines in development remains to be seen. As far as immu-
notherapeutics are concerned, the F protein-specific humanized monoclonal anti-
body, Palivizumab, is the only clinically approved intervention. This mAb has
demonstrated ADE at sub-neutralizing concentrations in vitro, yet has been found to
have a protective effect in animal models and humans (TI-RS Group 1998; Mejıas
et al. 2004; van Mechelen et al. 2016).
Ebola virus is a member of family Filoviridae. Of the four Ebola virus species
infecting humans, Zaire ebolavirus (EBOV) is the most virulent and is the causative
agent of a severe hemorrhagic fever disease, Ebola virus disease (EVD), with a
fatality rate of ~90% (Feldmann and Geisbert 2011). The disease is zoonotic, with
initial cases occurring due to contact with infected fruit bats (reservoir) or their
contaminated material, followed by human-to-human transmission. The virus is
mainly endemic in Africa; however, due to its increased incidence and fast spread,
as observed during the 2014–2016 African outbreak, the virus is considered a pan-
demic threat.
ADE of Ebola virus infection has been demonstrated in vitro in a variety of cell
lines and is shown to be mediated by antibodies specific to the viral envelope glyco-
protein (GP) (Takada et al. 2001, 2003, 2007). Such enhancing antibodies have been
identified in the sera of EVD patients (Takada et al. 2003; Furuyama et al. 2016).
Two different mechanisms have been proposed for ADE of EBOV infection: (a)
FcγR-mediated ADE and (b) C1q-mediated ADE. In FcγR-mediated ADE, anti-
body–virus complexes bind cell surface FcγRIIA triggering phosphorylation of Src
family protein tyrosine kinases (PTKs) and activation of Src signaling pathways
2 Antibody-Dependent Enhancement of Viral Infections 29
Acknowledgments The author is grateful to Dr. A.C. Mishra, Director, IRSHA, and Dr.
V.A. Arankalle, Head, Department of Communicable Diseases, IRSHA, for their constant
support.
Conflict of Interest The authors declare that they have no competing interests.
References
Acosta EG, Bartenschlager R (2016) Paradoxical role of antibodies in dengue virus infections:
considerations for prophylactic vaccine development. Expert Rev Vaccines 15(4):467–482
Alvarez M, Rodriguez-Roche R, Bernardo L, Vazquez S, Morier L, Gonzalez D, Castro O, Kouri
G, Halstead SB, Guzman MG (2006) Dengue hemorrhagic fever caused by sequential dengue
1–3 virus infections over a long time interval: Havana epidemic, 2001–2002. Am J Trop Med
Hyg 75:1113–1117
Appanna R, Wang SM, Ponnampalavanar SA, Lum LC, Sekaran SD (2012) Cytokine factors pres-
ent in dengue patient sera induces alterations of junctional proteins in human endothelial cells.
Am J Trop Med Hyg 87:936–942
Ayala-Nunez NV, Hoornweg TE, van de Pol DP, Sjollema KA, Flipse J, van der Schaar HM, Smit
JM (2016) How antibodies alter the cell entry pathway of dengue virus particles in macro-
phages. Sci Rep 6:28768
Balsitis SJ, Williams KL, Lachica R, Flores D, Kyle JL, Mehlhop E, Johnson S, Diamond MS,
Beatty PB, Harris E (2010) Lethal antibody enhancement of dengue disease in mice is pre-
vented by Fc modification. PLoS Pathog 6(2):e1000790
Banhegyi D, Bacsi A, Toth FD, Prohaszka Z, Horvath A, Beck Z, Konya J, Fust G (2003)
Significant decrease of the enhancement/neutralization index in HIV patients during highly
active antiretroviral therapy (HAART). Immunol Lett 89(1):25–30
Bardina SV, Bunduc P, Tripathi S, Duehr J, Frere JJ, Brown JA, Nachbagauer R, Foster GA,
Krysztof D, Tortorella D et al (2017) Enhancement of Zika virus pathogenesis by preexisting
antiflavivirus immunity. Science 356(6334):175–180
Beck Z, Prohászka Z, Füst G (2008) Traitors of the immune system-enhancing antibodies in HIV
infection: their possible implication in HIV vaccine development. Vaccine 26(24):3078–3085
Beltramello M, Williams KL, Simmons CP, Macagno A, Simonelli L, Quyen NT, Sukupolvi-Petty
S, Navarro-Sanchez E, Young PR, de Silva AM et al (2010) The human immune response to
dengue virus is dominated by highly cross-reactive antibodies endowed with neutralizing and
enhancing activity. Cell Host Microbe 8(3):271–283
Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, Drake JM, Brownstein
JS, Hoen AG, Sankoh O (2013) The global distribution and burden of dengue. Nature
496(7446):504–507
32 R. Kulkarni
Boonnak K, Slike BM, Donofrio GC, Marovich MA (2013) Human FcγRII cytoplasmic domains dif-
ferentially influence antibody-mediated dengue virus infection. J Immunol 190(11):5659–5665
Braciale TJ, Hahn YS, Burton DR (2013) Adaptive immune response to viral infections. In: Knipe
DM, Howley PM (eds) Fields Virology, 6th edn. Lippincott Williams and Wilkins, Philadelphia,
pp 214–253
Capeding MR, Tran NH, Hadinegoro SR, Ismail HI, Chotpitayasunondh T, Chua MN, Luong CQ,
Rusmil K, Wirawan DN, Nallusamy R et al (2014) Clinical efficacy and safety of a novel tet-
ravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked,
placebo-controlled trial. Lancet 384:1358–1365
Cardosa MJ (1987) Dengue virus isolation by antibody-dependent enhancement of infectivity in
macrophages. Lancet 329(8526):193–194
Cardosa MJ, Porterfield JS, Gordon S (1983) Complement receptor mediates enhanced flavivirus
replication in macrophages. J Exp Med 158:258–263
Carro AC, Piccini LE, Damonte EB (2018) Blockade of dengue virus entry into myeloid cells
by endocytic inhibitors in the presence or absence of antibodies. PLoS Negl Trop Dis
12(8):e0006685
Castilla V, Piccini LE, Damonte EB (2015) Dengue virus entry and trafficking: perspectives as
antiviral target for prevention and therapy. Future Virol 10:625–645
Chan KR, Ong EZ, Ooi EE (2013) Therapeutic antibodies as a treatment option for dengue fever.
Expert Rev Anti-Infect Ther 11(11):1147–1157
Chan KR, Ong EZ, Tan HC, Zhang SL, Zhang Q, Tang KF, Kaliaperumal N, Lim AP, Hibberd
ML, Chan SH et al (2014) Leukocyte immunoglobulin-like receptor B1 is critical for antibody-
dependent dengue. Proc Natl Acad Sci U S A 111(7):2722–2727
Chan KR, Wang X, Saron WAA, Gan ES, Tan HC, Mok DZ, Zhang SL, Lee YH, Liang C, Wijaya
L et al (2016) Cross-reactive antibodies enhance live attenuated virus infection for increased
immunogenicity. Nat Microbiol 1:16164
Chan-Hui PY, Swiderek KM (2016) Immunological considerations for developing antibody thera-
peutics for Influenza A. Hum Vaccin Immunother 12(2):474–477
Chanock RM, Kapikian AZ, Mills J, Kim HW, Parrott RH (1970) Influence of immunological fac-
tors in respiratory syncytial virus disease. Arch Environ Health 21(3):347–355
Chareonsirisuthigul T, Kalayanarooj S, Ubol S (2007) Dengue virus (DENV) antibody-dependent
enhancement of infection upregulates the production of anti-inflammatory cytokines, but sup-
presses anti-DENV free radical and pro-inflammatory cytokine production, in THP-1 cells. J
Gen Virol 88:365–375
Chaturvedi UC, Agarwal R, Elbishbishi EA, Mustafa AS (2000) Cytokine cascade in dengue hem-
orrhagic fever: implications for pathogenesis. FEMS Immunol Med Microbiol 28:183–188
Chau TN, Quyen NT, Thuy TT, Tuan NM, Hoang DM, Dung NT, Lien le B, Quy NT, Hieu NT,
Hieu LT et al (2008) Dengue in Vietnamese infants—results of infection enhancement assays
correlate with age-related disease epidemiology, and cellular immune responses correlate with
disease severity. J Infect Dis 198:516–524
Chau TN, Hieu NT, Anders KL, Wolbers M, Lien le B, Hieu LT, Hien TT, Hung NT, Farrar J,
Whitehead S et al (2009) Dengue virus infections and maternal antibody decay in a prospective
birth cohort study of Vietnamese infants. J Infect Dis 200:1893–1900
Chawla T, Chan KR, Zhang SL, Tan HC, Lim APC, Hanson BJ, Ooi EE (2013) Dengue virus neu-
tralization in cells expressing fc gamma receptors. PLoS One 8(5):e65231
Chotiwan N, Roehrig JT, Schlesinger JJ, Blair CD (2014) Molecular determinants of dengue
virus 2 envelope protein important for virus entry in FcγRIIA-mediated antibody-dependent
enhancement of infection. Virology 456–457:238–246
Connor RI, Dinces NB, Howell AL, Romet-Lemonne J-L, Pasquali J-L, Fanger MW (1991) Fc
receptors for IgG (FcγRs) on human monocytes and macrophages are not infectivity receptors
for human immunodeficiency virus type 1 (HIV-1): studies using bispecific antibodies to target
HIV-1 to various myeloid cell surface molecules, including the FcyR. Proc Natl Acad Sci U S
A 88:9593–9597
2 Antibody-Dependent Enhancement of Viral Infections 33
Crill WD, Chang G-JJ (2004) Localization and characterization of flavivirus envelope glycopro-
tein cross-reactive epitopes. J Virol 78(24):13975–13986
Crill WD, Hughes HR, Trainor NB, Davis BS, Whitnet MT, Chang G-JJ (2012) Sculpting humoral
immunity through dengue vaccination to enhance protective immunity. Front Immunol 3:334
Cruz-Oliveira C, Freire JM, Conceicão TM, Higa LM, Castanho MA, Da Poian AT (2015) Receptors
and routes of dengue virus entry into the host cells. FEMS Microbiol Rev 39:155–170
Davis D, Trischmann H, Stephens DM, Lachmann PJ (2001) Antibodies raised to short synthetic
peptides with sequences derived from HIV-1 SF2 gp120 can both neutralize and enhance HIV-1
SF13: a later variant isolated from the same host. J Med Virol 64(3):207–216
Dejnirattisai W, Jumnainsong A, Onsirisakul N, Fitton P, Vasanawathana S, Limpitikul W,
Puttikhunt C, Edwards C, Duangchinda T, Supasa S et al (2010) Cross-reacting antibodies
enhance dengue virus infection in humans. Science 328:745–748
Dejnirattisai W, Wongwiwat W, Supasa S, Zhang X, Dai X, Rouvinski A, Jumnainsong A, Edwards
C, Quyen NTH, Duangchinda T et al (2015) A new class of highly potent, broadly neutralizing
antibodies isolated from viremic patients infected with dengue virus. Nat Immunol 16:170–177
Dewi BE, Takasaki T, Kurane I (2004) In vitro assessment of human endothelial cell permeability:
effects of inflammatory cytokines and dengue virus infection. J Virol Methods 121:171–180
Dhama K, Karthik K, Khandia R, Chakraborty S, Munjal A, Latheef SK, Kumar D, Ramakrishnan
MA, Malik YS, Singh R et al (2018) Advances in designing and developing vaccines, drugs,
and therapies to counter Ebola virus. Front Immunol 9:1803
Dowd KA, Jost CA, Durbin AP, Whitehead SS, Pierson TC (2011) A dynamic landscape for anti-
body binding modulates antibody-mediated neutralization of West Nile virus. PLoS Pathog
7:e1002111
Feldmann H, Geisbert TW (2011) Ebola haemorrhagic fever. Lancet 377(9768):849–862
Fibriansah G, Tan JL, Smith SA, de Alwis R, Ng TS, Kostyuchenko VA, Jadi RS, Kukkaro P, de
Silva AM, Crowe JE et al (2015) A highly potent human antibody neutralizes dengue virus
serotype 3 by binding across three surface proteins. Nat Commun 6:6341
Flipse J, Diosa-Toro MA, Hoornweg TE, van de Pol DP, Urcuqui-Inchima S, Smit JM (2016)
Antibody-dependent enhancement of dengue virus infection in primary human macrophages;
balancing higher fusion against antiviral responses. Sci Rep 6:29201
Frei JC, Wirchnianski AS, Govero J, Vergnolle O, Dowd KA, Pierson TC, Kielian M, Girvin
ME, Diamond MS, Lai JR (2018) Engineered dengue virus domain III proteins elicit cross-
neutralizing antibody responses in mice. J Virol 92(18):e01023–e01018
Furuyama W, Marzi A, Carmody AB, Maruyama J, Kuroda M, Miyamoto H, Nanbo A, Manzoor
R, Yoshida R, Igarashi M et al (2016) Fcγ-receptor IIa-mediated Src signaling pathway is
essential for the antibody-dependent enhancement of Ebola virus infection. PLoS Pathog
12(12):e1006139
Fust G, Toth FD, Kiss J, Ujhelyi E, Nagy I, Banhegyi D (1994) Neutralizing and enhancing anti-
bodies measured in complement-restored serum samples from HIV-1-infected individuals cor-
relate with immunosuppression and disease. AIDS 8(5):603–609
Gan ES, Ting DH, Chan KR (2017) The mechanistic role of antibodies to dengue virus in protec-
tion and disease pathogenesis. Expert Rev Anti-Infect Ther 15(2):111–119
Gauger PC, Vincent AL, Loving CL, Lager KM, Janke BH, Kehrli ME Jr, Roth JA (2011) Enhanced
pneumonia and disease in pigs vaccinated with an inactivated human-like (delta-cluster) H1N2
vaccine and challenged with pandemic 2009 H1N1 influenza virus. Vaccine 29(15):2712–2719
George J, Valiant WG, Mattapallil MJ, Walker M, Huang Y-JS, Vanlandingham DL, Misamore J,
Greenhouse J, Weiss DE, Verthelyi D et al (2017) Prior exposure to Zika virus significantly
enhances peak dengue-2 viremia in rhesus macaques. Sci Rep 7:10498
Ghosn J, Taiwo B, Seedat S, Autran B, Katlama C (2018) HIV. Lancet 392(10148):685–697
Gibbons RV, Kalanarooj S, Jarman RG, Nisalak A, Vaughn DW, Endy TP, Mammen MP Jr,
Srikiatkhachorn A (2007) Analysis of repeat hospital admissions for dengue to estimate the
frequency of third or fourth dengue infections resulting in admissions and dengue hemorrhagic
fever, and serotype sequences. Am J Trop Med Hyg 77:910–913
34 R. Kulkarni
Gil L, Izquierdo A, Lazo L, Valdes I, Ambala P, Ochola L, Marcos E, Suzarte E, Kariuki T, Guzmán
G et al (2014) Capsid protein: evidences about the partial protective role of neutralizing anti-
body independent immunity against dengue in monkeys. Virology 456–457:70–76
Gil L, Lazo L, Valdés I, Suzarte E, Yen P, Ramírez R, Álvarez M, Dung LT, Cobas K, Marcos E
et al (2017) The tetravalent formulation of domain III-capsid proteins recalls memory B- and
T-cell responses induced in monkeys by an experimental dengue virus infection. Clin Transl
Immunology 6(6):e148
Gilbert PB, Peterson ML, Follmann D, Hudgens MG, Francis DP, Gurwith M, Heyward WL, Jobes
DV, Popovic V, Self SG et al (2005) Correlation between immunologic responses to a recombi-
nant glycoprotein 120 vaccine and incidence of HIV-1 infection in a phase 3 HIV-1 preventive
vaccine trial. J Infect Dis 191:666–677
Gimenez HB, Keir HM, Cash P (1989) In vitro enhancement of respiratory syncytial virus infec-
tion of U937 cells by human sera. J GenVirol 70:89–96
Gimenez HB, Chisholm S, Dornan J, Cash P (1996) Neutralizing and enhancing activities of
human respiratory syncytial virus-specific antibodies. Clin Diagn Lab Immunol 3:280–286
Glezen WP, Taber LH, Frank AL, Kasel JA (1986) Risk of primary infection and reinfection with
respiratory syncytial virus. Am J Dis Child 140:543–546
Gomez RS, Ramirez BA, Cespedes PF, Cautivo KM, Riquelme SA, Prado CE, Gonzalez PA,
Kalergis AM (2016) Contribution of Fcgamma receptors to human respiratory syncytial virus
pathogenesis and the impairment of T-cell activation by dendritic cells. Immunology 147:55–72
Goncalvez AP, Engle RE, St Claire M, Purcell RH, Lai CJ (2007) Monoclonal antibody-mediated
enhancement of dengue virus infection in vitro and in vivo and strategies for prevention. Proc
Natl Acad Sci U S A 104:9422–9427
Gotoff R, Tamura M, Janus J, Thompson J, Wright P, Ennis FA (1994) Primary influenza A virus
infection induces cross-reactive antibodies that enhance uptake of virus into Fc receptor/bear-
ing cells. J Infect Dis 169:200–203
Gould EA, Buckley A (1989) Antibody-dependent enhancement of yellow fever and Japanese
encephalitis virus neurovirulence. J Gen Virol 70(Pt 6):1605–1608
Graham RR, Juffrie M, Tan R, Hayes CG, Laksono I, Ma’roef C, Erlin, Sutaryo, Porter KR,
Halstead SB (1999) A prospective seroepidemiologic study on dengue in children four to nine
years of age in Yogyakarta, Indonesia I. studies in 1995–1996. Am J Trop Med Hyg 61:412–419
Guilliams M, Bruhns P, Saeys Y, Hammad H, Lambrecht BN (2014) The function of Fcγ receptors
in dendritic cells and macrophages. Nat Rev Immunol 14:94–108
Guillon C, Schutten M, Boers PH, Gruters RA, Osterhaus AD (2002) Antibody-mediated enhance-
ment of human immunodeficiency virus type 1 infectivity is determined by the structure of
gp120 and depends on modulation of the gp120-CCR5 interaction. J Virol 76:2827–2834
Guzman MG, Kouri GP, Bravo J, Soler M, Vazquez S, Morier L (1990) Dengue hemorrhagic fever
in Cuba, 1981: a retrospective seroepidemiologic study. Am J Trop Med Hyg 42:179–184
Guzman MG, Kouri G, Valdes L, Bravo J, Alvarez M, Vazquez S, Delgado I, Halstead SB (2000)
Epidemiologic studies on dengue in Santiago de Cuba, 1997. Am J Epidemiol 152:793–799
Guzman MG, Alvarez M, Halstead SB (2013) Secondary infection as a risk factor for dengue
hemorrhagic fever/dengue shock syndrome: an historical perspective and role of antibody-
dependent enhancement of infection. Arch Virol 158(7):1445–1459
Hadinegoro SR, Arredondo-Garcia JL, Capeding MR, Deseda C, Chotpitayasunondh T, Dietze
R, Muhammad Ismail HI, Reynales H, Limkittikul K, Rivera-Medina DM et al (2015)
Efficacy and long-term safety of dengue vaccine in regions of endemic disease. N Engl J Med
373:1195–1206
Hall CB, Weinberg GA, Iwane MK, Blumkin AK, Edwards KM, Staat MA, Auinger P, Griffin
MR, Poehling KA, Erdman D, Grijalva CG, Zhu Y, Szilagyi P (2009) The burden of respiratory
syncytial virus infection in young children. N Engl J Med 360:588–598
Hallengard D, Lum FM, Kümmerer BM, Lulla A, Lulla V, García-Arriaza J, Fazakerley JK,
Roques P, Le Grand R, Merits A et al (2014) Prime-boost immunization strategies against
Chikungunya virus. J Virol 88(22):13333–13343
2 Antibody-Dependent Enhancement of Viral Infections 35
Halstead SB (1979) In vivo enhancement of dengue virus infection in rhesus monkeys by passively
transferred antibody. J Infect Dis 140:527–533
Halstead SB (2003) Neutralization and antibody-dependent enhancement of dengue viruses. Adv
Virus Res 60:421–467
Halstead SB (2008) Pathogenesis: risk factors prior to infection. In: Halstead SB (ed) Dengue.
Imperial College Press, London, pp 219–256
Halstead SB, Nimmannitya S, Yamarat C, Russell PK (1967) Hemorrhagic fever in Thailand:
recent knowledge regarding etiology. Jpn J Med Sci Biol 20:96–103
Halstead SB, Chow J, Marchette NJ (1973a) Immunologic enhancement of dengue virus replica-
tion. Nat New Biol 243:24–26
Halstead SB, Shotwell H, Casals J (1973b) Studies on the pathogenesis of dengue infection in
monkeys. II. Clinical laboratory responses to heterologous infection. J Infect Dis 128:15–22
Hawkes RA (1964) Enhancement of the infectivity of arboviruses by specific antisera produced in
domestic fowls. Aust J Exp Biol Med Sci 42:465–482
Hawkes RA, Lafferty KJ (1967) The enhancement of virus infectivity by antibody. Virology
33:250–261
Hertz T, Beatty PR, MacMillen Z, Killingbeck SS, Wang C, Harris E (2017) Antibody epitopes
identified in critical regions of dengue virus nonstructural 1 protein in mouse vaccination and
natural human infections. J Immunol 198(10):4025–4035
Homsy J, Meyer M, Tateno M, Clarkson S, Levy JA (1989) The Fc and not CD4 receptor mediates
antibody enhancement of HIV infection in human cells. Science 244(4910):1357–1360
Homsy J, Meyer M, Levy JA (1990) Serum enhancement of human immunodeficiency virus (HIV)
infection correlates with disease in HIV-infected individuals. J Virol 64:1437–1440
Huisman W, Martina BEE, Rimmelzwaan GF, Gruters RA, Osterhaus ADME (2009) Vaccine-
induced enhancement of viral infections. Vaccine 27:505–512
Injampa S, Muenngern N, Pipattanaboon C, Benjathummarak S, Boonha K, Hananantachai H,
Wongwit W, Ramasoota P, Pitaksajjakul P (2017) Generation and characterization of cross
neutralizing human monoclonal antibody against 4 serotypes of dengue virus without enhanc-
ing activity. Peer J 5:e4021
Janoff EN, Wahl SM, Thomas K, Smith PD (1995) Modulation of human immunodeficiency virus
type 1 infection of human monocytes by IgA. J Infect Dis 172:855–858
Jaume M, Yip MS, Cheung CY, Leung HL, Li PH, Kien F, Dutry I, Callendret B, Escriou N,
Altmeyer R et al (2011) Anti-severe acute respiratory syndrome coronavirus spike antibodies
trigger infection of human immune cells via a pH- and cysteine protease-independent FcγR
pathway. J Virol 85:10582–10597
Junjhon J, Edwards TJ, Utaipat U, Bowman VD, Holdaway HA, Zhang W, Keelapang P, Puttikhunt
C, Perera R, Chipman PR et al (2010) Influence of pr-M cleavage on the heterogeneity of extra-
cellular dengue virus particles. J Virol 84(16):8353–8358
Kam YW, Kien F, Roberts A, Cheung YC, Lamirande EW, Vogel L, Chu SL, Tse J, Guarner J, Zaki
SR et al (2007) Antibodies against trimeric S glycoprotein protect hamsters against SARS-CoV
challenge despite their capacity to mediate FcgammaRII-dependent entry into B cells in vitro.
Vaccine 25:729–740
Kam YW, Ong EK, Rénia L, Tong JC, Ng LF (2009) Immuno-biology of chikungunya and impli-
cations for disease intervention. Microbes Infect 11(14–15):1186–1196
Katzelnick LC, Harris E (2017) Immune correlates of protection for dengue: state of the art and
research agenda. Vaccine 35:4659–4669
Katzelnick LC, Gresh L, Halloran ME, Mercado JC, Kuan G, Gordon A, Balmaseda A, Harris
E (2017) Antibody-dependent enhancement of severe dengue disease in humans. Science
358(6365):929–932
Khurana S, Loving CL, Manischewitz J, King LR, Gauger PC, Henningson J, Vincent AL, Golding
H (2013) Vaccine-induced anti-HA2 antibodies promote virus fusion and enhance influenza
virus respiratory disease. Sci Transl Med 5(200):200ra114
36 R. Kulkarni
Kim HW, Canchola JG, Brandt CD, Pyles G, Chanock RM, Jensen K, Parrott RH (1969)
Respiratory syncytial virus disease in infants despite prior administration of antigenic inacti-
vated vaccine. Am J Epidemiol 89:422–434
Klasse PJ, Sattentau QJ (2002) Occupancy and mechanism in antibody-mediated neutralization of
animal viruses. J Gen Virol 83:2091–2108
Kliks S (1990) Antibody-enhanced infection of monocytes as the pathogenetic mechanism for
severe dengue illness. AIDS Res Hum Retrovir 6(8):993–998
Kliks SC, Nimmanitya S, Nisalak A, Burke DS (1988) Evidence that maternal dengue antibodies
are important in the development of dengue hemorrhagic fever in infants. Am J Trop Med Hyg
38:411–419
Kozlowski PA, Black KP, Shen L, Jackson S (1995) High prevalence of serum IgA HIV-1 infection-
enhancing antibodies in HIV-infected persons. Masking by IgG. J Immunol 154:6163–6173
Krilov LR, Anderson LJ, Marcoux L, Bonagura VR, Wedgwood JF (1989) Antibody-mediated
enhancement of respiratory syncytial virus infection in two monocyte/macrophage cell lines.
J Infect Dis 160:777–782
Kyriakis CS, Gramer MR, Barbe F, Van Doorsselaere J, Van Reeth K (2010) Efficacy of commer-
cial swine influenza vaccines against challenge with a recent European H1N1 field isolate. Vet
Microbiol 144(1–2):67–74
Laurence J, Saunders A, Early E, Salmon JE (1990) Human immunodeficiency virus infection of
monocytes: relationship to Fc-gamma receptors and antibody-dependent viral enhancement.
Immunology 70:338–343
Lee N, Chan PK, Ip M, Wong E, Ho J, Ho C, Cockram CS, Hui DS (2006) Anti-SARS-CoV
IgG response in relation to disease severity of severe acute respiratory syndrome. J Clin Virol
35:179–184
Li L, Lok SM, Yu I-M, Zhang Y, Kuhn RJ, Chen J, Rossmann MG (2008) The flavivirus precursor
membrane-envelope protein complex: structure and maturation. Science 319(5871):1830–1834
Li M, Zhao L, Zhang C, Wang X, Hong W, Sun J, Liu R, Yu L, Wang J, Zhang F et al (2018)
Dengue immune sera enhance Zika virus infection in human peripheral blood monocytes
through Fc gamma receptors. PLoS One 13(7):e0200478
Lidbury BA, Mahalingam S (2000) Specific ablation of antiviral gene expression in macrophages
by antibody-dependent enhancement of Ross River virus infection. J Virol 74:8376–8381
Lindenbach BD, Murray CL, Thiel H-J, Rice CM (2013) Flaviviridae. In: Knipe DM, Howley
PM (eds) Fields virology, 6th edn. Lippincott Williams and Wilkins, Philadelphia, pp 712–746
Lu J, Wang R, Xia B, Yu Y, Zhou X, Yang Z, Huang P (2018) Potent neutralization ability of a
human monoclonal antibody against serotype 1 dengue virus. Front Microbiol 9:1214
Lum FM, Couderc T, Chia BS, Ong RY, Her Z, Chow A, Leo YS, Kam YW, Rénia L, Lecuit M et al
(2018) Antibody-mediated enhancement aggravates chikungunya virus infection and disease
severity. Sci Rep 8(1):1860
Mahalingam S, Herring BL, Halstead SB (2013) Call to action for dengue vaccine failure. Emerg
Infect Dis 19:1335–1337
Marchette NJ, Halstead SB, Falkler WA Jr, Stenhouse A, Nash D (1973) Studies on the patho-
genesis of dengue infection in monkeys. III. Sequential distribution of virus in primary and
heterologous infections. J Infect Dis 128:23–30
Matsui K, Gromowski GD, Li L, Barrett ADT (2010) Characterization of a dengue type-specific
epitope on dengue 3 virus envelope protein domain III. J Gen Virol 91(Pt 9):2249–2253
McArthur MA, Sztein MB, Edelman R (2013) Dengue vaccines: recent developments, ongoing
challenges and current candidates. Expert Rev Vaccines 12(8):933–953
Mejıas A, Chavez-Bueno S, Rıos AM, Saavedra-Lozano J, Fonseca Aten M, Hatfield J, Kapur P,
Gómez AM, Jafri HS, Ramilo O (2004) Anti-respiratory syncytial virus (RSV) neutralizing
antibody decreases lung inflammation, airway obstruction, and airway hyperresponsiveness in
a murine RSV model. Antimicrob Agents Chemother 48:1811–1822
Meyer K, Ait-Goughoulte M, Keck Z-Y, Foung S, Ray R (2008) Antibody-dependent enhancement
of hepatitis C virus infection. J Virol 82(5):2140–2149
2 Antibody-Dependent Enhancement of Viral Infections 37
Prohaszka Z, Nemes J, Hidvegi T, Toth FD, Kerekes K, Erdei A, Szabó J, Ujhelyi E, Thielens N,
Dierich MP et al (1997) Two parallel routes of the complement-mediated antibody-dependent
enhancement of HIV-1 infection. AIDS 11(8):949–958
Puerta-Guardo H, Raya-Sandino A, González-Mariscal L, Rosales VH, Ayala-Dávila J, Chávez-
Mungía B, Martínez-Fong D, Medina F, Ludert JE, del Angel RM (2013) The cytokine
response of U937-derived macrophages infected through antibody-dependent enhancement of
dengue virus disrupts cell apical-junction complexes and increases vascular permeability. J
Virol 87(13):7486–7501
Rajao DS, Loving CL, Gauger PC, Kitikoon P, Vincent AL (2014) Influenza A virus hemagglutinin
protein subunit vaccine elicits vaccine-associated enhanced respiratory disease in pigs. Vaccine
32(40):5170–5176
Reeth KV, Brown I, Essen S, Pensaert M (2004) Genetic relationships, serological cross-reaction
and cross-protection between H1N2 and other influenza A virus subtypes endemic in European
pigs. Virus Res 103(1–2):115–124
Ricklin ME, Vielle NJ, Python S, Brechbühl D, Zumkehr B, Posthaus H, Zimmer G, Summerfield
A (2016) Partial protection against porcine influenza A virus by a hemagglutinin-expressing
virus replicon particle vaccine in the absence of neutralizing antibodies. Front Immunol 7:253
Robinson WE Jr (2006) Mechanism for complement-mediated, antibody dependent enhancement
of human immunodeficiency virus type 1 infection in MT2 cells is enhanced entry through
CD4, CD21, and CXCR4 chemokine receptors. Viral Immunol 19(3):434–447
Robinson WE Jr, Montefiori DC, Mitchell WM (1987) A human immunodeficiency virus type
1 (HIV-1) infection-enhancing factor in seropositive sera. Biochem Biophys Res Commun
149(2):693–699
Robinson WE Jr, Montefiori DC, Mitchell WM (1988) Antibody dependent enhancement of
human immunodeficiency virus type 1 infection. Lancet 1(8589):790–794
Robinson WE Jr, Montefiori DC, Gillespie DH, Mitchell WM (1989) Complement mediated,
antibody-dependent enhancement of HIV-1 infection in vitro is characterized by increased pro-
tein and RNA syntheses and infectious virus release. J Acquir Immune Defic Syndr 2:33–42
Robinson WE Jr, Kawamura T, Lake D, Masuho Y, Mitchell WM, Hersh EM (1990a) Antibodies
to the primary immunodominant domain of human immunodeficiency virus type 1 (HIV-1)
glycoprotein gp4l enhance HIV-1 infection in vitro. J Virol 64(11):5301–5305
Robinson WE Jr, Kawamura T, Gorny MK, Lake D, Xu JY, Matsumoto Y, Sugano T, Masuho Y,
Mitchell WM, Hersh E et al (1990b) Human monoclonal antibodies to the human immuno-
deficiency virus type 1 (HIV-1) transmembrane glycoprotein gp41 enhance HIV-1 infection
in vitro. Proc Natl Acad Sci U S A 87(8):3185–3189
Rodenhuis-Zybert IA, Moesker B, da Silva Voorham JM, van der Ende-Metselaar H, Diamond
MS, Wilschut J, Smit JM (2011) A fusion-loop antibody enhances the infectious properties of
immature flavivirus particles. J Virol 85:11800–11808
Rolph MS, Zaid A, Rulli NE, Mahalingam S (2011) Downregulation of interferon-β in antibody-
dependent enhancement of dengue viral infections of human macrophages is dependent on
interleukin-6. J Infect Dis 204(3):489–491
Rothman AL (2011) Immunity to dengue virus: a tale of original antigenic sin and tropical cyto-
kine storms. Nat Rev Immunol 11:532–543
Sangkawibha N, Rojanasuphot S, Ahandrik S, Viriyapongse S, Jatanasen S, Salitul V,
Phanthumachinda B, Halstead SB (1984) Risk factors in dengue shock syndrome: a prospective
epidemiologic study in Rayong, Thailand. I. the 1980 outbreak. Am J Epidemiol 120:653–669
Schmidt AC (2010) Response to dengue fever - the good, the bad, and the ugly? N Engl J Med
363(5):484–487
Serafin IL, Aaskov JG (2001) Identification of epitopes on the envelope (E) protein of dengue 2
and dengue 3 viruses using monoclonal antibodies. Arch Virol 146(12):2469–2479
Shanks GD, Burroughs SA, Sohn JD, Waters NC, Smith VF, Waller M, Brundage JF (2016)
Enhanced risk of illness during the 1918 influenza pandemic after previous influenza-like ill-
nesses in three military populations. Epidemiol Infect 144(10):2043–2048
2 Antibody-Dependent Enhancement of Viral Infections 39
Shao W, Li X, Goraya ML, Wang S, Chen J-L (2017) Evolution of influenza A virus by mutation
and re-assortment. Int J Mol Sci 18(8):E1650
Shi X, Deng Y, Wang H, Ji G, Tan W, Jiang T, Li X, Zhao H, Xia T, Meng Y et al (2016) A bispe-
cific antibody effectively neutralizes all four serotypes of dengue virus by simultaneous block-
ing virus attachment and fusion. MAbs 8(3):574–584
Shmelkov E, Nadas A, Cardozo T (2014) Could vaccination with AIDSVAX immunogens have
resulted in antibody dependent enhancement of HIV infection in human subjects? Hum Vaccin
Immunother 10(10):3013–3016
Sholukh AM, Byrareddy SN, Shanmuganathan V, Hemashettar G, Lakhashe SK, Rasmussen
RA, Watkins JD, Vyas HK, Thorat S, Brandstoetter T et al (2014) Passive immunization of
macaques with polyclonal anti-SHIV IgG against a heterologous tier 2 SHIV: outcome depends
on IgG dose. Retrovirology 11:8
Skowronski DM, De Serres G, Crowcroft NS, Janjua NZ, Boulianne N, Hottes TS, Rosella LC,
Dickinson JA, Gilca R, Sethi P et al (2010) Association between the 2008–09 seasonal influ-
enza vaccine and pandemic H1N1 illness during spring–summer 2009: four observational stud-
ies from Canada. PLoS Med 7(4):e1000258
Skowronski DM, Hamelin ME, De Serres G, Janjua NZ, Li G, Sabaiduc S, Bouhy X, Couture C,
Leung A, Kobasa D (2014) Randomized controlled ferret study to assess the direct impact of
2008-09 trivalent inactivated influenza vaccine on A (H1N1)pdm09 disease risk. PLoS One
9:e86555
Smith SA, de Alwis AR, Kose N, Jadi RS, de Silva AM, Crowe JE (2014) Isolation of dengue
virus-specific memory B cells with live virus antigen from human subjects following natural
infection reveals the presence of diverse novel functional groups of antibody clones. J Virol
88(21):12233–12241
Sridhar S, Luedtke A, Langevin E, Zhu M, Bonaparte M, Machabert T, Savarino S, Zambrano B,
Moureau A, Khromava A et al (2018) Effect of dengue serostatus on dengue vaccine safety and
efficacy. N Engl J Med 379(4):327–340
Staprans SI, Barry AP, Silvestri G, Safrit JT, Kozyr N, Sumpter B, Nguyen H, McClure H,
Montefiori D, Cohen JI et al (2004) Enhanced SIV replication and accelerated progression to
AIDS in macaques primed to mount a CD4 T cell response to the SIV envelope protein. Proc
Natl Acad Sci U S A 101(35):13026–13031
Subbramanian RA, Xu J, Toma E, Morisset R, Cohen EA, Menezes J, Ahmad A (2002) Comparison
of human immunodeficiency virus (HIV)-specific infection-enhancing and -inhibiting antibod-
ies in AIDS patients. J Clin Microbiol 40(6):2141–2146
Suhrbier A, La Linn M (2003) Suppression of antiviral responses by antibody-dependent enhance-
ment of macrophage infection. Trends Immunol 24:165–168
Sullivan N, Sun Y, Binley J, Lee J, Barbas CF 3rd, Parren PW, Burton DR, Sodroski J (1998)
Determinants of human immunodeficiency virus type 1 envelope glycoprotein activation by
soluble CD4 and monoclonal antibodies. J Virol 72:6332–6338
Sun H, Chen Q, Lai H (2018) Development of antibody therapeutics against flaviviruses. Int J Mol
Sci 19(1):E54
Szabo J, Prohaszka Z, Toth FD, Gyuris A, Segesdi J, Banhegyi D, Ujhelyi E, Minarovits J, Fust
G (1999) Strong correlation between the complement-mediated antibody-dependent enhance-
ment of HIV-1 infection and plasma viral load. AIDS 13:1841–1849
Takada A, Kawaoka Y (2003) Antibody-dependent enhancement of viral infection: molecular
mechanisms and in vivo implications. Rev Med Virol 13(6):387–398
Takada A, Watanabe S, Okazaki K, Kida H, Kawaoka Y (2001) Infectivity-enhancing antibodies to
Ebola virus glycoprotein. J Virol 75(5):2324–2330
Takada A, Feldmann H, Ksiazek TG, Kawaoka Y (2003) Antibody-dependent enhancement of
Ebola virus infection. J Virol 77(13):7539–7544
Takada A, Ebihara H, Feldmann H, Geisbert TW, Kawaoka Y (2007) Epitopes required
for antibody- dependent enhancement of Ebola virus infection. J Infect Dis 196(Suppl
2):S347–S356
40 R. Kulkarni
Takeda A, Tuazon CU, Ennis FA (1988) Antibody-enhanced infection by HIV-1 via Fc receptor-
mediated entry. Science 242(4878):580–583
Takeda A, Sweet RW, Ennis FA (1990) Two receptors are required for antibody-dependent
enhancement of human immunodeficiency virus type 1 infection: CD4 and FcγR. J Virol
64(11):5605–5610
Tamura M, Webster RG, Ennis FA (1991) Antibodies to HA and NA augment uptake of influenza
A viruses into cells via Fc receptor entry. Virology 182:211–219
Tamura M, Webster RG, Ennis FA (1994) Subtype cross-reactive, infection-enhancing antibody
responses to influenza A viruses. J Virol 68(6):3499–3504
Tang C-T, Li P-C, Liu I-J, Liao M-Y, Chiu CY, Chao D-Y, Wu HC (2015) An epitope-substituted
DNA vaccine improves safety and immunogenicity against dengue virus type 2. PLoS Negl
Trop Dis 9(7):e0003903
Taylor A, Foo SS, Bruzzone R, Dinh LV, King NJ, Mahalingam S (2015) Fc receptors in antibody-
dependent enhancement of viral infections. Immunol Rev 268(1):340–364
TI-RS Group (1998) Palivizumab, a humanized respiratory syncytial virus monoclonal antibody,
reduces hospitalization from respiratory syncytial virus infection in high-risk infants. Pediatrics
102(3 Pt 1):531–537
To KK, Zhang AJ, Hung IF, Xu T, Ip WC, Wong RT, Ng JC, Chan JF, Chan KH, Yuen KY (2012)
High titer and avidity of nonneutralizing antibodies against influenza vaccine antigen are asso-
ciated with severe influenza. Clin Vaccine Immunol 19(7):1012–1018
Toth FD, Szabo B, Ujhelyi E, Paloczi K, Horvath A, Fust G, Kiss J, Bánhegyi D, Hollán SR
(1991) Neutralizing and complement-dependent enhancing antibodies in different stages of
HIV infection. AIDS 5(3):263–268
Tóth FD, Mosborg-Petersen P, Kiss J, Aboagye-Mathiesen G, Zdravkovic M, Hager H, Aranyosi
J, Lampé L, Ebbesen P (1994) Antibody-dependent enhancement of HIV-1 infection in human
term syncytiotrophoblast cells cultured in vitro. Clin Exp Immunol 96(3):389–394
Trischmann H, Davis D, Lachmann PJ (1995) Lymphocytotropic strains of HIV type 1 when com-
plexed with enhancing antibodies can infect macrophages via Fc gamma RIII. AIDS Res Hum
Retrovir 11(3):343–352
Tsai T-T, Chuang Y-J, Lin Y-S, Chang C-P, Wan S-W, Lin S-H, Chen C-L, Lin C-F (2014)
Antibody-dependent enhancement infection facilitates dengue virus-regulated signaling of
IL-10 production in monocytes. PLoS Negl Trop Dis 8(11):e3320
Ubol S, Phuklia W, Kalayanarooj S, Modhiran N (2010) Mechanisms of immune evasion induced
by a complex of dengue virus and preexisting enhancing antibodies. J Infect Dis 201:923–935
van de Winkel JG, Anderson CL (1991) Biology of human immunoglobulin G Fc receptors. J
Leukoc Biol 49(5):511–524
van Erp EA, van Kasteren PB, Guichelaar T, Ahout IML, de Haan CAM, Luytjes W, Ferwerda G,
Wicht O (2017) In vitro enhancement of respiratory syncytial virus infection by maternal anti-
bodies does not explain disease severity in infants. J Virol 91(21):e00851–e00817
van Mechelen L, Luytjes W, de Haan CA, Wicht O (2016) RSV neutralization by palivizumab, but
not by monoclonal antibodies targeting other epitopes, is augmented by Fc gamma receptors.
Antivir Res 132:1–5
Vaughn DW, Green S, Kalayanarooj S, Innis BL, Nimmannitya S, Suntayakorn S, Endy TP,
Raengsakulrach B, Rothman AL, Ennis FA et al (2000) Dengue viraemia titer, antibody
response pattern, and virus serotype correlate with disease severity. J Infect Dis 181(1):2–9
Villar L, Dayan GH, Arredondo-Garcia JL, Rivera DM, Cunha R, Deseda C, Reynales H, Costa
MS, Morales-Ramírez JO, Carrasquilla G et al (2015) Efficacy of a tetravalent dengue vaccine
in children in Latin America. N Engl J Med 372:113–123
Vogelpoel LT, Baeten DL, de Jong EC, den Dunnen J (2015) Control of cytokine production
by human Fc gamma receptors: implications for pathogen defense and autoimmunity. Front
Immunol 6:79
Wahala WM, Silva AM (2011) The human antibody response to dengue virus infection. Viruses
3(12):2374–2395
2 Antibody-Dependent Enhancement of Viral Infections 41
Wang WK, Chen HL, Yang CF, Hsieh SC, Juan CC, Chang SM, Yu CC, Lin LH, Huang JH, King
CC (2006) Slower rates of clearance of viral load and virus-containing immune complexes in
patients with dengue hemorrhagic fever. Clin Infect Dis 43:1023–1030
Wang SF, Tseng SP, Yen CH, Yang JY, Tsao CH, Shen CW, Chen KH, Liu FT, Liu WT, Chen YM
et al (2014) Antibody-dependent SARS coronavirus infection is mediated by antibodies against
spike proteins. Biochem Biophys Res Commun 451:208–214
Wang Y, Si L, Luo Y, Guo X, Zhou J, Fang D, Yan H, Zeng G, Jiang L (2015) Replacement of pr
gene with Japanese encephalitis virus pr using reverse genetics reduces antibody-dependent
enhancement of dengue virus 2 infection. Appl Microbiol Biotechnol 99(22):9685–9698
Wang Q, Zhang L, Kuwahara K, Li L, Liu Z, Li T, Zhu H, Liu J, Xu Y, Xie J et al (2016)
Immunodominant SARS coronavirus epitopes in humans elicited both enhancing and neutral-
izing effects on infection in non-human primates. ACS Infect Dis 2(5):361–376
WHO (2009) Dengue: guidelines for diagnosis, treatment, prevention and control. WHO, Geneva.
www.who.int/tdr/publications/documents/dengue-diagnosis.pdf
Wilder-Smith A, Hombach J, Ferguson N, Selgelid M, O’Brien K, Vannice K, Barrett A, Ferdinand
E, Flasche S, Guzman M et al (2019) Deliberations of the strategic advisory group of experts
on immunization on the use of CYD-TDV dengue vaccine. Lancet Infect Dis 19(1):e31–e38
Willey S, Aasa-Chapman MM, O’Farrell S, Pellegrino P, Williams I, Weiss RA, Neil SJ (2011)
Extensive complement-dependent enhancement of HIV-1 by autologous non-neutralising anti-
bodies at early stages of infection. Retrovirology 8:16
Xu M, Zuest R, Velumani S, Tukijan F, Toh YX, Appanna R, Tan EY, Cerny D, MacAry P, Wang C
et al (2017) A potent neutralizing antibody with therapeutic potential against all four serotypes
of dengue virus. NPJ Vaccin 2:2
Yip MS, Leung NH, Cheung CY, Li PH, Lee HH, Daëron M, Peiris JS, Bruzzone R, Jaume M
(2014) Antibody-dependent infection of human macrophages by severe acute respiratory syn-
drome coronavirus. Virol J 11:82
Yip MS, Leung HL, Li PH, Cheung CY, Dutry I, Li D, Daëron M, Bruzzone R, Peiris JS, Jaume
M (2016) Antibody-dependent enhancement of SARS coronavirus infection and its role in the
pathogenesis of SARS. Hong Kong Med J 22(3 Suppl 4):25–31
Zellweger RM, Prestwood TR, Shresta S (2010) Enhanced infection of liver sinusoidal endothe-
lial cells in a mouse model of antibody-induced severe dengue disease. Cell Host Microbe
7:128–139
Zhang L, Zhang F, Yu W, He T, Yu J, Yi CE, Ba L, Li W, Farzan M, Chen Z et al (2006) Antibody
responses against SARS coronavirus are correlated with disease outcome of infected individu-
als. J Med Virol 78:1–8
Inflammation During Virus Infection:
Swings and Roundabouts 3
Sankar Bhattacharyya
Abstract
Inflammation constitutes a concerted series of cellular and molecular responses
that follow disturbance of systemic homeostasis, by either toxins or infectious
organisms. Leukocytes modulate inflammation through production of secretory
mediators, like cytokines and chemokines, which work in an autocrine and/or
paracrine manner. These mediators can either promote or attenuate the inflam-
matory response and depending on differential temporal and spatial expression
play a crucial role in the outcome of infection. Even though the objective is clear-
ance of the pathogen with minimum damage to host, the pathogenesis of multi-
ple human pathogenic viruses has been suggested to emanate from a dysregulation
of the inflammatory response, sometimes with fatal consequences. This review
discusses the nature and the outcome of inflammatory response, which is trig-
gered in the human host subsequent to infection by single-sense plus-strand
RNA viruses. In view of such harmful effects of a dysregulated inflammatory
response, an exogenous regulation of these reactions by either interference or
supplementation of critical regulators has been suggested. Currently multiple
such factors are being tested for their beneficial and adverse effects. A successful
use of such an approach in diseases of viral etiology can potentially protect the
affected individual without directly affecting the virus life cycle. Further, such
approaches whenever applicable would be useful in mitigating death and/or
debility that is caused by the infection of those viruses which have proven par-
ticularly difficult to control by either prophylactic vaccines and/or therapeutic
strategies using specific antiviral drugs.
S. Bhattacharyya (*)
Vaccine and Infectious Disease Research Centre, Translational Health Science and
Technology Institute, Faridabad, Haryana, India
e-mail: [email protected]
Keywords
Inflammation · RNA virus · Cytokine therapy
3.1 Introduction
The mammalian immune system has evolved arsenal and strategies to make a dis-
tinction between microbes that are either beneficial or benign or bad, an integral part
of which is differential treatment of “self” and “non-self.” Whereas recognition of
“self” as “non-self” can cause autoimmunity, the converse results in microbial colo-
nization. In fact the human gut does harbor multiple variety of microbes as natural
part of the biological ecosystem (Scarpellini et al. 2015). The recognized non-self
are counteracted by adaptive and innate effectors of the immune system, using dedi-
cated cells and biochemicals, which attempt to restrict the growth and impede colo-
nization by the pathogen. The innate response is nonspecific, while the secondary
adaptive response is specific for the pathogen or closely related species. The cellular
component includes innate immune cells like the monocytes/macrophages, neutro-
phils, and natural killer (NK) cells and adaptive immune cells like B- and
T-lymphocytes, which coordinate for an effective response. Cytokines are a dedi-
cated group of biochemicals involved in this coordination and include interferons
(IFNs), interleukins (ILs), and chemokines that are responsible for synchronizing
the initiation, regulation, and termination of an immune response. A group (~100)
of small polypeptides (<20 kDa) produced predominantly although not exclusively
by immune cells like macrophages and lymphocytes, cytokines are secreted out
exerting their function by engaging respective cell-surface receptors and depending
on biological function are labeled as either pro-inflammatory (PIC) or anti-
inflammatory (AIC) cytokines (Turner et al. 2014). On the one hand, several cyto-
kines are functionally redundant, and on the other hand, some cells can express
receptors for multiple cytokines.
releasing viral genome into the host cytosol (Kumar et al. 2018; Plemper 2011). A
culmination of the following steps results in direct translation of the genomic RNA
to produce a polyprotein, which is cleaved by virus-derived and host-origin prote-
ases to yield the multiple structural and nonstructural proteins (Fields et al. 2013).
The structural features of the genomic RNA facilitating translation can be, e.g., a
5’cap and a poly-A tail (Alphavirus, Togaviridae; Coronavirus, Coronaviridae) or a
5’cap without a poly-A tail (Flavivirus, Flaviviridae) or an internal-ribosome entry
site (IRES) serving for ribosome recruitment without a poly-A tail (hepatitis C
virus) or an IRES with a poly-A tail (Picornaviridae, Astroviridae, Caliciviridae).
After multiple rounds of translation, ribosome loading stops and the genomic RNA
is replicated by virus-encoded RNA-dependent RNA polymerase (RdRp), in endo-
plasmic reticulum (ER) membrane-associated replication complexes (RCs) during
which a double-stranded RNA intermediate is produced followed by its asymmetric
transcription to produce multiple copies of plus-sense genomic RNA. The new
genomic RNAs are packaged into virion particles that exit the cell by either secre-
tory pathway or plasma-membrane budding (for enveloped viruses) or by cell lysis
(for non-enveloped viruses) (Bird and Kirkegaard 2015; Pornillos et al. 2002).
Although viruses can replicate in multiple types of cells, the pathological outcome
manifests in only one or a few specific cell/tissue types. Independent of organismal
entry site, the likeliest primary encounter of a virus is with mononuclear-phagocytic
cells like monocytes, macrophages (Mϕ), and dendritic cells (DCs). Mϕ and DCs
are tissue-localized cells constituting the first line of cellular defense, which when
infected can undertake antiviral steps in addition to “informing” the other effectors
of the innate and adaptive immune system (Pohl et al. 2007; Ginhoux and Jung
2014). Activated DCs shift to lymph nodes, process viral antigen, and “present” or
display it for clonal expansion of cognate lymphocytes population. Mϕ, which can
be either tissue-resident or differentiated from afferent monocytes postinfection,
play a more regulatory role and are important determinants of the outcome of the
inflammatory response (Ginhoux and Jung 2014; Mercer and Greber 2013; Hou
et al. 2012; Schulz et al. 2012). Tissue-resident Mϕ , which are a distinct population
from monocyte-derived ones, include microglial cells in CNS, liver Kupffer cells,
skin Langerhans cells, etc. (Davies et al. 2013)
Monocytes/Mϕ (and many other cell types) express molecular detector proteins
called pattern recognition receptors (PRRs), specialized for interacting with signa-
ture motifs on microbe-derived molecules, termed as pathogen-associated molecu-
lar pattern (PAMP). Viral PAMPs include double-stranded (dsRNA) RNA
(replication-intermediate formed during replication) and 5′-ppp (uncapped genomic
RNA polymerized by de novo replication). Cellular PRRs specific for these include
toll-like receptors (TLRs) like TLR3 (dsRNA) and RIG-I-like receptors (RLRs) like
RIG-I, MDA5 (dsRNA, 5′-ppp end on RNA) (Jensen and Thomsen 2012). Nod-like
receptors or NLRs form another class of cytosolic PRRs that can detect virus
46 S. Bhattacharyya
infection, albeit in an indirect manner (Takeuchi and Akira 2010; Ichinohe et al.
2013). Physical engagement with PAMPs activates the respective PRRs, stimulating
alterations in conformation of these sensors that allow them to interact with adapter
molecules mediating the assembly of multi-protein complexes called inflammo-
some, in parallel to activating the expression of cytokine genes coding for type-1
interferons (IFNs) and NFκB target genes (Kawai et al. 2005; Pichlmair and Reis e
Sousa 2007; Chen and Ichinohe 2015; Seth et al. 2005). Secreted type-I IFNs attach
specific receptors, in a paracrine or autocrine manner, thereby activating the expres-
sion of many interferon-sensitive genes (ISGs) with diverse functions that confer
antiviral property to their activity (Schneider et al. 2014; Schoggins and Rice 2011).
ISGs include PRR-coding genes producing a feed-forward loop and aggravating
inflammation. In parallel, NFκB enhances expression of pro-inflammatory genes
like TNF-α, IL-1β, COX2, IL6, IL-12p40, or IL-12 besides components of NLRP3
(Tak and Firestein 2001; Bauernfeind et al. 2009). Upon assembly the NLRP3
inflammosome catalyzes caspase-1 activation, a protease which slices the precursor
form of pleiotropic pro-inflammatory cytokines like IL-1β and IL-18 generating
their active secreted forms (Garlanda et al. 2013; Biet et al. 2002). IL-1β potentiates
the antiviral response by multiple ways in addition to inducing expression of type-I
IFNs and ISGs in DCs (Ben-Sasson et al. 2011; Aarreberg et al. 2018). Chemokines
(chemotactic cytokines) flag/point to the site of infection by a concentration gradi-
ent, attracting leukocytes like neutrophils, monocytes, and lymphocytes, subse-
quently activating them to release more cytokines thereby amplifying the
inflammatory response (Sokol and Luster 2015; Ley 2014). Among these IL-12 and
IL-2 (produced predominantly by DCs) have crucial immunomodulatory functions.
IL-12 attracts CD4+ T-helper (Th) cells influencing their differentiation into IFN-γ
secreting Th1 cells in addition to augmenting the cytotoxic activity of CD8+ T cells
and NK cells (Athie-Morales et al. 2004; Henry et al. 2008). IL-2 on the other hand
increases NK-cell sensitivity to IL-12 by receptor upregulation (Wang et al. 2000).
IFN-γ which in contrast to type-I IFNs is produced exclusively by immune cells (T
and NK cells) has pleiotropic antiviral effect including the capacity to polarize
existing or newly recruited Mϕ to M1 phenotype (Hu and Ivashkiv 2009; Verreck
et al. 2004). Mϕ either resident or monocyte-derived can acquire either an M1 or an
M2 phenotype differing in ontology, phenotype, and secretome, with unidirectional
plasticity from M1 to M2 (Halstead et al. 2010; Guiducci et al. 2005; Smith et al.
2016). M1-Mϕ promotes a Th1 immune response which is necessary for resolution
of infection, while the M2-Mϕ endorses tissue repair following inflammation, sug-
gesting that a premature skew in abundance of M2-Mϕ at the expense of M1-Mϕ
would limit viral clearance leading to chronic infection and prolonged inflammatory
response (Klenerman and Hill 2005). An emerging concept in modulation of inflam-
mation involves the role of bacterial surface components like lipopolysaccharide on
concurrent viral infection (Smith et al. 2016; Wilks and Golovkina 2012). Alterations
in gut microbiome have been reported and potential influences this might have on
disease outcome have been suggested (Preveden et al. 2017; Banks et al. 2015).
Though it is difficult to ascertain the number of asymptomatic infections for any
given virus, the percentage of symptomatic infection vis-à-vis asymptomatic ones is
3 Inflammation During Virus Infection: Swings and Roundabouts 47
often a multivariate variable, being known for only a few. For example, only 1
among 4 individuals infected with DENV shows febrile symptoms. This suggests a
success for the antiviral immune mechanisms in the majority of individuals. Animal
studies using gene knockout models have given evidence of this efficacy for many
viruses (Suthar et al. 2010; Samuel and Diamond 2005; Lazear et al. 2011; Deonarain
et al. 2004; Burdeinick-Kerr et al. 2007). In case of humans, these information are
complicated by differential efficacy of these pathways, protecting or predisposing
individuals under the influence of genotype, environment, etc. (Paalani et al. 2011;
Mitchell and Aneshensel 2016; Liu and Taioli 2015) Besides, there are few studies
that indicate potential influence of medication or noninfectious ailments or societal
stress on the outcome of infection through an influence on the immune system
(Mehrbod et al. 2014; Gilbert et al. 2005; Htun et al. 2015; Jean et al. 2007).
HCV and DENV infection can cause liver damage through a chronic and acute
infection regime, respectively (Samanta and Sharma 2015; Axley et al. 2018). Liver
as an organ is characterized by a high capacity to regenerate; however, chronic
injury/scarring can lead to fibrosis, steatosis, or even hepatocellular carcinoma
resulting in liver failure (Forbes and Newsome 2016). Hepatocytes constitute two-
thirds of all liver cells and are associated with all major liver functions besides play-
ing a crucial role in innate immune signaling (Kmiec 2001; Zhou et al. 2016).
Hepatocytes are permissible to both HCV and DENV, the latter being reported to
additionally infect Kupffer cells (Chang et al. 2003; Zehender et al. 1997; Boisvert
et al. 2001; Caussin-Schwemling et al. 2001; Goutagny et al. 2003; Marianneau
et al. 1999; de Macedo et al. 2006; Huerre et al. 2001). In acute infection, the major
damage is through apoptosis following direct infection of these cells, whereas
establishment of a chronic infection usually causes a sustained inflammation lead-
ing to infiltration of polymorphonuclear cells and lymphocytes (Huerre et al. 2001;
Lim et al. 2014; Masalova et al. 2017; Deng et al. 2008; Bala et al. 2012; Sung et al.
2012). Irrespective of the virus, these infections augment PIC levels in the liver with
drastic consequences. For example, hepatocyte apoptosis caused by either direct
infection or effect of PICs like TNF-α generates apoptotic bodies which when
engulfed by Kupffer cells induce the latter to release more PIC providing a positive
loop toward inflammation (Canbay et al. 2003a; Burdette et al. 2012; Negash et al.
2013; Shimizu et al. 2005). Cytokines like TGFβ and PDGF thus released can “acti-
vate” hepatic stellate cells initiating a metabolic transformation in them to secrete
more extracellular matrix that deposits as fibrotic tissue in addition to converting
them into smooth muscle fibers (Canbay et al. 2003b; Hernandez-Gea and Friedman
2011). In addition to virus infection-induced changes, bacterial LPS can also poten-
tially “activate” hepatic stellate cells (Brun et al. 2005). HCV infection skews mac-
rophage population to M2 phenotype restraining virus clearance while promoting
hepatic stellate cell activation mediated by TGFβ (Saha et al. 2016). Additionally, in
48 S. Bhattacharyya
The central nervous system (CNS) is physiologically isolated from the rest of the
body by a specialized selectively permeable barricade called as the blood–brain bar-
rier (BBB), which allows passage to selected metabolites, respiratory gases, and an
extremely limited repertoire of circulatory tissue cells. This isolation is necessary
for protection of low regeneration capacity neuronal cells from systemic inflamma-
tion, which can also upset the structural and functional plasticity of neurons that is
dependent on cytokine signaling (Arnett et al. 2001; Gougeon et al. 2013; Mason
et al. 2001; Fischer et al. 2011; Brissoni et al. 2006). The CNS can have either neu-
ronal or non-neuronal glial cells; the latter provide vital functional support and
include microglia (macrophage-like immune cells), oligodendrocytes (which pro-
vide insulation for neurons), and astrocytes (responsible for repair of damaged neu-
ronal tissue). Microglial cells have immunomodulatory function in suppressing a
pathogenic inflammation (Seitz et al. 2018). Multiple viruses in the +ve-ssRNA
genome group, including Coronavirus, Picornavirus, Flaviviridae, and Togaviridae,
cause opportunistic infection of CNS (Bergmann et al. 2006; Koyuncu et al. 2013;
Fletcher and McKeating 2012).
In the absence of a direct admission route, these viruses undergo limited repli-
cation in peripheral tissue, before entering through either peripheral nerves or
BBB microvasculature or CNS infiltrating leukocytes (functioning as the prover-
bial “Trojan horse”) (Koyuncu et al. 2013; Jeha et al. 2003). A feature common
here is a breach of the vascular endothelial barrier at varying locations, e.g., BBB
for JEV/WNV, blood retinal barrier for ZIKV, and endothelial barriers in lungs/
peritoneum for DENV. Breach in BBB is more common for some viruses (e.g.,
WNV, JEV, ZIKAV, poliovirus) correlating with fatality. Interestingly, WNV and
JEV have been suggested to cause BBB disruption from inside the CNS (Li et al.
2015; Verma et al. 2009). Still other reports suggest infected endothelial cells to
secrete PICs that disrupt the BBB (Chen et al. 2014; Chang et al. 2017; Roach and
Alcendor 2017). The tissue damage is caused from a combination of either direct
neuronal infection which activates intrinsic apoptosis or a hyperactive inflamma-
tory response mediated by PICs or CD8+ cytotoxic T cells (CTLs) (Wang et al.
3 Inflammation During Virus Infection: Swings and Roundabouts 49
2003; Samuel et al. 2007). Infected neurons secrete chemokines that attract leuko-
cytes like monocytes and lymphocytes (Klein et al. 2005; Shrestha and Diamond
2004; Glass et al. 2005; Kelley et al. 2003; Lim et al. 2011; Bardina et al. 2015;
Durrant et al. 2015; Shrestha et al. 2008). The relation between a “good” and
“bad” immune response is, however, very tricky when it comes to the
CNS. Migration of CTLs expressing receptors for chemokines like CCL2, CCL3,
CCL4, CCL5, CXCL9–11, as well as its timing with respect to establishment of
infection, seems to play a crucial role in virus eradication and survival (Wang
et al. 2003; Shrestha and Diamond 2004; Diamond et al. 2003; Sussman et al.
1989; Getts et al. 2010; Nansen et al. 2000; Chen et al. 2004; Liu et al. 2001; Zink
et al. 2001; Winter et al. 2004). The CTLs exert their antiviral role by inducing
cell death through either a perforin-dependent or Fas-FasL-mediated mechanism
(Rossi et al. 1998; Shrestha and Diamond 2007). In addition to CTLs, other PICs
might also induce direct cell death in neurons (Dhanwani et al. 2012; Olmo et al.
2017; Baer et al. 2016; Kumar et al. 2010).
Dengue virus causes a febrile illness with can turn fatal after a subsidence of the
fever. The severity emanates from leakage of fluid from the blood vessels by a
breach of the vascular endothelium. Circulating in four serotypes, severe disease is
mostly associated with secondary infection by a serotype different from the one
causing primary infection. Neutralizing antibodies generated during primary infec-
tion incompletely neutralize the secondary infection virus and instead promote their
uptake by monocytes, by a phenomenon called antibody-dependent enhancement or
ADE (Katzelnick et al. 2017; Dejnirattisai et al. 2016). Notwithstanding a primary
or secondary infection, the pathological symptoms are considered to be the result of
an unbridled host immune response (Basu and Chaturvedi 2008; Rothman 2011).
DENV infects a variety of cells including monocytes, dendritic cells (skin
Langerhans cells), macrophages (Kupffer cells), and vascular endothelial cells,
expectedly leading to PIC secretion (Wu et al. 2000; Jessie et al. 2004; Tolfvenstam
et al. 2011). Different studies have reported a positive association of DHF/DSS
development with extraordinarily augmented levels of different PICs that include
macrophage migration inhibitory factor (MIF), IFN-α, TNF-α (Green et al. 1999;
Kurane et al. 1993; Huang et al. 2000; Chen et al. 2006). Although multiple reports
have suggested correlation between specific PIC level and plasma leakage, the
mechanism is still elusive and limited to association studies (Priyadarshini et al.
2010; Her et al. 2017; Sehrawat et al. 2018; Malavige et al. 2012). Interestingly
however, multiple similar association studies have suggested a positive association
between levels of IL10 (an AIC) and severe/critical symptoms related to dengue
infection (Malavige et al. 2013; Tsai et al. 2013; Flores-Mendoza et al. 2017). IL10,
produced by multiple immune cells, suppresses immune response through upregu-
lation of SOCS (suppressor of cytokine signaling) function and downregulation of
IFN activity, the result being decreased T-cell cytotoxicity (Halstead et al. 2010;
50 S. Bhattacharyya
Katzelnick et al. 2017; Tsai et al. 2013; Azeredo et al. 2001; Brasier et al. 2012).
The augmentation of IL10 level has been suggested to emanate from monocytes
infected by the ADE route with additional influence from high viremia (Tsai et al.
2014). IL10 is a dominant regulator of the immune system that can prolong patho-
gen clearance through a subversion of the immune response (Couper et al. 2008).
Coronavirus infections are usually benign causing self-limiting mild flu-like symp-
toms. However, recent outbreaks involving, e.g., severe acute respiratory syndrome
coronavirus (SARS-CoV), which jumped species barrier through acquisition of
minor genome mutations, have projected them as potentially severe human patho-
gens (Guan et al. 2004). Spread through aerosols, SARS-CoV primarily infect lung
cells triggering an often fatal inflammatory response clinically called acute respira-
tory distress syndrome (ARDS) that starts with severe hypoxia, pulmonary edema
progressing to systemic inflammation, and failure of multiple organs, culminating
in high rate of mortality (Peiris et al. 2003; Lew et al. 2003; Tsushima et al. 2009;
Farcas et al. 2005). Although evidence suggests that SARS-CoV can infect multiple
cell types, lung type-II pneumocytes and ciliated epithelial cells constitute primary
sites of virus replication, consequent to which these cells undergo apoptotic and/or
necrotic death attracting innate immune cells and activating them to secrete PICs
(Sims et al. 2005; Chow et al. 2004; Nicholls et al. 2003). The nature of inflamma-
tion following SARS-CoV infection is characterized by a prompt production of
PICs through immediate NFκB activation and a delayed expression of type-I IFN
genes (Shi et al. 2014; Kong et al. 2009; Wong et al. 2004). Severity of symptoms
correlates positively with IL-6 levels while exhibiting negative correlation with that
of IL-8 and TGFβ (Zhang et al. 2004). As observed with many other viral pathogen-
esis models, macrophage polarization culminating in preferential enrichment of
M2-macrophages has been suggested to be responsible for SARS-CoV pathogene-
sis (Page et al. 2012). SARS-CoV infection is also associated with hemophagocyto-
sis or engulfment of different types of blood cells by histiocytes (a class within
macrophages), which is a clinical marker of immune system hyper-activation
(Usmani et al. 2013).
Conflict of Interest The author declares that they have no competing interests.
References
Aarreberg LD et al (2018) Interleukin-1beta signaling in dendritic cells induces antiviral interferon
responses. MBio 9. ). pii: e00342-18.
Acharya D et al (2016) Interleukin-17A promotes CD8+ T cell cytotoxicity to facilitate West Nile
virus clearance. J Virol 91:e01529
Arnett HA et al (2001) TNF alpha promotes proliferation of oligodendrocyte progenitors and
remyelination. Nat Neurosci 4:1116–1122
Athie-Morales V, Smits HH, Cantrell DA, Hilkens CM (2004) Sustained IL-12 signaling is
required for Th1 development. J Immunol 172:61–69
Axley P, Ahmed Z, Ravi S, Singal AK (2018) Hepatitis C virus and hepatocellular carcinoma: a
narrative review. J Clin Transl Hepatol 6:79–84
52 S. Bhattacharyya
Huang YH et al (2000) Dengue virus infects human endothelial cells and induces IL-6 and IL-8
production. Am J Trop Med Hyg 63:71–75
Huerre MR et al (2001) Liver histopathology and biological correlates in five cases of fatal dengue
fever in Vietnamese children. Virchows Arch 438:107–115
Ichinohe T, Yamazaki T, Koshiba T, Yanagi Y (2013) Mitochondrial protein mitofusin 2 is required
for NLRP3 inflammasome activation after RNA virus infection. Proc Natl Acad Sci U S A
110:17963–17968
Jean CM, Honarmand S, Louie JK, Glaser CA (2007) Risk factors for West Nile virus neuroinva-
sive disease, California, 2005. Emerg Infect Dis 13:1918–1920
Jeha LE et al (2003) West Nile virus infection: a new acute paralytic illness. Neurology 61:55–59
Jensen S, Thomsen AR (2012) Sensing of RNA viruses: a review of innate immune receptors
involved in recognizing RNA virus invasion. J Virol 86:2900–2910
Jessie K, Fong MY, Devi S, Lam SK, Wong KT (2004) Localization of dengue virus in natu-
rally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis
189:1411–1418
Ji JD et al (2005) Inhibition of IL-10-induced STAT3 activation by 15-deoxy-Delta12,14-
prostaglandin J2. Rheumatology (Oxford) 44:983–988
Katzelnick LC et al (2017) Antibody-dependent enhancement of severe dengue disease in humans.
Science 358:929–932
Kawai T et al (2005) IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon
induction. Nat Immunol 6:981–988
Kelley TW, Prayson RA, Ruiz AI, Isada CM, Gordon SM (2003) The neuropathology of West Nile
virus meningoencephalitis. A report of two cases and review of the literature. Am J Clin Pathol
119:749–753
Klein RS et al (2005) Neuronal CXCL10 directs CD8+ T-cell recruitment and control of West Nile
virus encephalitis. J Virol 79:11457–11466
Klenerman P, Hill A (2005) T cells and viral persistence: lessons from diverse infections. Nat
Immunol 6:873–879
Kmiec Z (2001) Cooperation of liver cells in health and disease. In: Advances in anatomy, embry-
ology and cell biology, vol 161. Springer, Berlin, pp 1–151
Kong SL, Chui P, Lim B, Salto-Tellez M (2009) Elucidating the molecular physiopathology of
acute respiratory distress syndrome in severe acute respiratory syndrome patients. Virus Res
145:260–269
Koyuncu OO, Hogue IB, Enquist LW (2013) Virus infections in the nervous system. Cell Host
Microbe 13:379–393
Kumar M, Verma S, Nerurkar VR (2010) Pro-inflammatory cytokines derived from West Nile virus
(WNV)-infected SK-N-SH cells mediate neuroinflammatory markers and neuronal death. J
Neuroinflammation 7:73
Kumar CS, Dey D, Ghosh S, Banerjee M (2018) Breach: host membrane penetration and entry by
nonenveloped viruses. Trends Microbiol 26:525–537
Kurane I et al (1993) High levels of interferon alpha in the sera of children with dengue virus infec-
tion. Am J Trop Med Hyg 48:222–229
Lazear HM, Pinto AK, Vogt MR, Gale M Jr, Diamond MS (2011) Beta interferon controls West
Nile virus infection and pathogenesis in mice. J Virol 85:7186–7194
Lazear HM et al (2015) Interferon-lambda restricts West Nile virus neuroinvasion by tightening
the blood-brain barrier. Sci Transl Med 7:284ra59
Lew TW et al (2003) Acute respiratory distress syndrome in critically ill patients with severe acute
respiratory syndrome. JAMA 290:374–380
Ley K (2014) The second touch hypothesis: T cell activation, homing and polarization. F1000Res
3:37
Li F et al (2015) Viral infection of the central nervous system and neuroinflammation precede blood-
brain barrier disruption during Japanese encephalitis virus infection. J Virol 89:5602–5614
Lim JK et al (2011) Chemokine receptor Ccr2 is critical for monocyte accumulation and survival
in West Nile virus encephalitis. J Immunol 186:471–478
56 S. Bhattacharyya
Lim EJ et al (2014) Hepatitis C virus-induced hepatocyte cell death and protection by inhibition of
apoptosis. J Gen Virol 95:2204–2215
Liu B, Taioli E (2015) Seasonal variations of complete blood count and inflammatory biomarkers
in the US population – analysis of NHANES data. PLoS One 10:e0142382
Liu MT, Armstrong D, Hamilton TA, Lane TE (2001) Expression of Mig (monokine induced by
interferon-gamma) is important in T lymphocyte recruitment and host defense following viral
infection of the central nervous system. J Immunol 166:1790–1795
MacDonald AJ et al (2002) CD4 T helper type 1 and regulatory T cells induced against the same
epitopes on the core protein in hepatitis C virus-infected persons. J Infect Dis 185:720–727
Malavige GN et al (2012) Cellular and cytokine correlates of severe dengue infection. PLoS One
7:e50387
Malavige GN et al (2013) Serum IL-10 as a marker of severe dengue infection. BMC Infect Dis
13:341
Marianneau P et al (1999) Infection of primary cultures of human Kupffer cells by dengue virus:
no viral progeny synthesis, but cytokine production is evident. J Virol 73:5201–5206
Masalova OV et al (2017) Modulation of Cell Death Pathways by Hepatitis C Virus Proteins in
Huh7.5 Hepatoma Cells. Int J Mol Sci 18:E2346
Mason JL, Suzuki K, Chaplin DD, Matsushima GK (2001) Interleukin-1beta promotes repair of
the CNS. J Neurosci 21:7046–7052
Mehrbod P, Omar AR, Hair-Bejo M, Haghani A, Ideris A (2014) Mechanisms of action and effi-
cacy of statins against influenza. Biomed Res Int 2014(872370):1
Mercer J, Greber UF (2013) Virus interactions with endocytic pathways in macrophages and den-
dritic cells. Trends Microbiol 21:380–388
Mitchell UA, Aneshensel CS (2016) Social inequalities in inflammation. J Aging Health
29:769–787
Nansen A, Marker O, Bartholdy C, Thomsen AR (2000) CCR2+ and CCR5+ CD8+ T cells
increase during viral infection and migrate to sites of infection. Eur J Immunol 30:1797–1806
Negash AA et al (2013) IL-1beta production through the NLRP3 inflammasome by hepatic mac-
rophages links hepatitis C virus infection with liver inflammation and disease. PLoS Pathog
9:e1003330
Nicholls JM et al (2003) Lung pathology of fatal severe acute respiratory syndrome. Lancet
361:1773–1778
Okuda M et al (2002) Mitochondrial injury, oxidative stress, and antioxidant gene expression are
induced by hepatitis C virus core protein. Gastroenterology 122:366–375
Olenina LV et al (2005) Identification of glycosaminoglycan-binding sites within hepatitis C virus
envelope glycoprotein E2∗. J Viral Hepat 12:584–593
Olmo IG et al (2017) Zika virus promotes neuronal cell death in a non-cell autonomous manner by
triggering the release of neurotoxic factors. Front Immunol 8:1016
Paalani M, Lee JW, Haddad E, Tonstad S (2011) Determinants of inflammatory markers in a bi-
ethnic population. Ethn Dis 21:142–149
Page C et al (2012) Induction of alternatively activated macrophages enhances pathogenesis during
severe acute respiratory syndrome coronavirus infection. J Virol 86:13334–13349
Peiris JS, Yuen KY, Osterhaus AD, Stohr K (2003) The severe acute respiratory syndrome. N Engl
J Med 349:2431–2441
Perlemuter G et al (2002) Hepatitis C virus core protein inhibits microsomal triglyceride transfer
protein activity and very low density lipoprotein secretion: a model of viral-related steatosis.
FASEB J 16:185–194
Pichlmair A, Reis e Sousa C (2007) Innate recognition of viruses. Immunity 27:370–383
Plemper RK (2011) Cell entry of enveloped viruses. Curr Opin Virol 1:92–100
Pohl C, Shishkova J, Schneider-Schaulies S (2007) Viruses and dendritic cells: enemy mine. Cell
Microbiol 9:279–289
Pornillos O, Garrus JE, Sundquist WI (2002) Mechanisms of enveloped RNA virus budding.
Trends Cell Biol 12:569–579
3 Inflammation During Virus Infection: Swings and Roundabouts 57
Preveden T, Scarpellini E, Milic N, Luzza F, Abenavoli L (2017) Gut microbiota changes and
chronic hepatitis C virus infection. Expert Rev Gastroenterol Hepatol 11:813–819
Priyadarshini D et al (2010) Clinical findings and pro-inflammatory cytokines in dengue patients
in Western India: a facility-based study. PLoS One 5:e8709
Raemer PC, Kohl K, Watzl C (2009) Statins inhibit NK-cell cytotoxicity by interfering with LFA-
1-mediated conjugate formation. Eur J Immunol 39:1456–1465
Reynolds IJ, Miller RJ (1989) Ifenprodil is a novel type of N-methyl-D-aspartate receptor antago-
nist: interaction with polyamines. Mol Pharmacol 36:758–765
Rhen T, Cidlowski JA (2005) Antiinflammatory action of glucocorticoids--new mechanisms for
old drugs. N Engl J Med 353:1711–1723
Roach T, Alcendor DJ (2017) Zika virus infection of cellular components of the blood-retinal bar-
riers: implications for viral associated congenital ocular disease. J Neuroinflammation 14:43
Rossi CP, McAllister A, Tanguy M, Kagi D, Brahic M (1998) Theiler’s virus infection of perforin-
deficient mice. J Virol 72:4515–4519
Rothman AL (2011) Immunity to dengue virus: a tale of original antigenic sin and tropical cyto-
kine storms. Nat Rev Immunol 11:532–543
Saha B, Kodys K, Szabo G (2016) Hepatitis C virus-induced monocyte differentiation into polar-
ized M2 macrophages promotes stellate cell activation via TGF-beta. Cell Mol Gastroenterol
Hepatol 2:302–316.e8
Samanta J, Sharma V (2015) Dengue and its effects on liver. World J Clin Cases 3:125–131
Samuel MA, Diamond MS (2005) Alpha/beta interferon protects against lethal West Nile virus infec-
tion by restricting cellular tropism and enhancing neuronal survival. J Virol 79:13350–13361
Samuel MA, Morrey JD, Diamond MS (2007) Caspase 3-dependent cell death of neurons contrib-
utes to the pathogenesis of West Nile virus encephalitis. J Virol 81:2614–2623
Scarpellini E et al (2015) The human gut microbiota and virome: potential therapeutic implica-
tions. Dig Liver Dis 47:1007–1012
Schneider WM, Chevillotte MD, Rice CM (2014) Interferon-stimulated genes: a complex web of
host defenses. Annu Rev Immunol 32:513–545
Schoggins JW, Rice CM (2011) Interferon-stimulated genes and their antiviral effector functions.
Curr Opin Virol 1:519–525
Schulz C et al (2012) A lineage of myeloid cells independent of Myb and hematopoietic stem cells.
Science 336:86–90
Sehrawat P et al (2018) Role of cytokines as molecular marker of dengue severity. Mediterr J
Hematol Infect Dis 10:e2018023
Seitz S, Clarke P, Tyler KL (2018) Pharmacologic depletion of microglia increases viral load in
the brain and enhances mortality in murine models of flavivirus-induced encephalitis. J Virol
92:e00525
Seth RB, Sun L, Ea CK, Chen ZJ (2005) Identification and characterization of MAVS, a mitochon-
drial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell 122:669–682
Shi CS et al (2014) SARS-coronavirus open reading frame-9b suppresses innate immunity by tar-
geting mitochondria and the MAVS/TRAF3/TRAF6 signalosome. J Immunol 193:3080–3089
Shimizu S et al (2005) Liver injury induced by lipopolysaccharide is mediated by TNFR-1 but not
by TNFR-2 or Fas in mice. Hepatol Res 31:136–142
Shrestha B, Diamond MS (2004) Role of CD8+ T cells in control of West Nile virus infection. J
Virol 78:8312–8321
Shrestha B, Diamond MS (2007) Fas ligand interactions contribute to CD8+ T-cell-mediated con-
trol of West Nile virus infection in the central nervous system. J Virol 81:11749–11757
Shrestha B, Zhang B, Purtha WE, Klein RS, Diamond MS (2008) Tumor necrosis factor alpha
protects against lethal West Nile virus infection by promoting trafficking of mononuclear leu-
kocytes into the central nervous system. J Virol 82:8956–8964
Sims AC et al (2005) Severe acute respiratory syndrome coronavirus infection of human ciliated
airway epithelia: role of ciliated cells in viral spread in the conducting airways of the lungs. J
Virol 79:15511–15524
58 S. Bhattacharyya
Smith TD, Tse MJ, Read EL, Liu WF (2016) Regulation of macrophage polarization and plasticity
by complex activation signals. Integr Biol (Camb) 8:946–955
Sokol CL, Luster AD (2015) The chemokine system in innate immunity. Cold Spring Harb
Perspect Biol 7. pii: a016303.
Sugimoto K et al (2003) Suppression of HCV-specific T cells without differential hierarchy dem-
onstrated ex vivo in persistent HCV infection. Hepatology 38:1437–1448
Sung JM, Lee CK, Wu-Hsieh BA (2012) Intrahepatic infiltrating NK and CD8 T cells cause liver
cell death in different phases of dengue virus infection. PLoS One 7:e46292
Sussman MA, Shubin RA, Kyuwa S, Stohlman SA (1989) T-cell-mediated clearance of mouse
hepatitis virus strain JHM from the central nervous system. J Virol 63:3051–3056
Suthar MS et al (2010) IPS-1 is essential for the control of West Nile virus infection and immunity.
PLoS Pathog 6:e1000757
Tak PP, Firestein GS (2001) NF-kappaB: a key role in inflammatory diseases. J Clin Invest
107:7–11
Takeuchi O, Akira S (2010) Pattern recognition receptors and inflammation. Cell 140:805–820
Tan CW, Poh CL, Sam IC, Chan YF (2013) Enterovirus 71 uses cell surface heparan sulfate gly-
cosaminoglycan as an attachment receptor. J Virol 87:611–620
Thomas SJ, Grossberg GT (2009) Memantine: a review of studies into its safety and efficacy in
treating Alzheimer’s disease and other dementias. Clin Interv Aging 4:367–377
Tolfvenstam T et al (2011) Characterization of early host responses in adults with dengue disease.
BMC Infect Dis 11:209
Tsai TT et al (2013) An emerging role for the anti-inflammatory cytokine interleukin-10 in dengue
virus infection. J Biomed Sci 20:40
Tsai TT et al (2014) Antibody-dependent enhancement infection facilitates dengue virus-regulated
signaling of IL-10 production in monocytes. PLoS Negl Trop Dis 8:e3320
Tsushima K et al (2009) Acute lung injury review. Intern Med 48:621–630
Turner MD, Nedjai B, Hurst T, Pennington DJ (2014) Cytokines and chemokines: at the crossroads
of cell signalling and inflammatory disease. Biochim Biophys Acta 1843:2563–2582
Usmani GN, Woda BA, Newburger PE (2013) Advances in understanding the pathogenesis of
HLH. Br J Haematol 161:609–622
Verma S et al (2009) West Nile virus infection modulates human brain microvascular endothelial
cells tight junction proteins and cell adhesion molecules: transmigration across the in vitro
blood-brain barrier. Virology 385:425–433
Verreck FA et al (2004) Human IL-23-producing type 1 macrophages promote but IL-10-
producing type 2 macrophages subvert immunity to (myco)bacteria. Proc Natl Acad Sci U S
A 101:4560–4565
Wang KS, Frank DA, Ritz J (2000) Interleukin-2 enhances the response of natural killer cells
to interleukin-12 through up-regulation of the interleukin-12 receptor and STAT4. Blood
95:3183–3190
Wang Y, Lobigs M, Lee E, Mullbacher A (2003) CD8+ T cells mediate recovery and immunopa-
thology in West Nile virus encephalitis. J Virol 77:13323–13334
Wang T et al (2004) Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal
encephalitis. Nat Med 10:1366–1373
Wilks J, Golovkina T (2012) Influence of microbiota on viral infections. PLoS Pathog 8:e1002681
Winter PM et al (2004) Proinflammatory cytokines and chemokines in humans with Japanese
encephalitis. J Infect Dis 190:1618–1626
Wong CK et al (2004) Plasma inflammatory cytokines and chemokines in severe acute respiratory
syndrome. Clin Exp Immunol 136:95–103
Wu SJ et al (2000) Human skin Langerhans cells are targets of dengue virus infection. Nat Med
6:816–820
Zehender G et al (1997) Detection of hepatitis C virus RNA in CD19 peripheral blood mono-
nuclear cells of chronically infected patients. J Infect Dis 176:1209–1214
3 Inflammation During Virus Infection: Swings and Roundabouts 59
Zhang Y et al (2004) Analysis of serum cytokines in patients with severe acute respiratory syn-
drome. Infect Immun 72:4410–4415
Zhou Z, Xu MJ, Gao B (2016) Hepatocytes: a key cell type for innate immunity. Cell Mol Immunol
13:301–315
Zink MC et al (2001) Increased macrophage chemoattractant protein-1 in cerebrospinal fluid pre-
cedes and predicts simian immunodeficiency virus encephalitis. J Infect Dis 184:1015–1021
Interferons and Their Role in Viral
Infection 4
Suji George, Gururaj Rao Deshpande,
and Gajanan N. Sapkal
Abstract
Interferons (IFNs) are a large family of naturally occurring cytokines, discovered
in 1957. They are secreted by host immune cells in response to virus infections,
tumors, and other biological inducers as diverse countermeasures against stimu-
lation. They elicit and regulate inter- and intracellular networks of immune sys-
tem by producing interferon-induced proteins. These proteins play a vital role in
inhibiting any vulnerable step in viral replication cycle starting from viral entry/
uncoating to maturation and release of the virus. Presently many interferons are
approved as therapeutic agents, and tremendous progress has been made in
understanding their role and usage as antiviral agents. This chapter explores the
essential link of IFNs with the innate and adaptive immune system during viral
infections. Further it discusses in detail the types of IFNs, genes induced by IFNs
in the host, IFN-induced proteins and their antiviral roles, and the strategies used
by viruses to counter the effects of antiviral action by IFNs. Finally, the chapter
concludes by summarizing the therapeutic application of IFNs to fight viral
diseases.
Keywords
Interferons · Antiviral activity · Immune system
4.1 Introduction
Viruses cause infectious diseases by invading living cells to multiply and produce
progeny viruses. The host immune system plays a key role in defending against the
infection and this defense barrier consists of innate immune cells such as dendritic
cells, macrophages, and natural killer cells. These cells are capable of mounting an
immune response by recognizing a wide range of viral products. Activation of these
cells can result in rapid viral sensing and release of key antiviral cytokines, which
can inhibit viral replication and can further activate other components of innate and
adaptive immunity. Interferons (IFNs) are a group of highly specialized cytokines
capable of mediating antiviral, antigrowth, and immune modulation responses. It
was discovered by Isaacs and Lindenmann in 1957 as a molecule secreted by virus-
infected cells and was able to prevent further infection of cells that were exposed to
it (Isaacs and Lindenmann 1957). IFN activity includes the induction of the tran-
scription of IFN-stimulated gene (ISG) that actively participates in the inhibition of
crucial steps in viral replication.
IFNs are classified into three types (I, II, and III) based on the structural homology,
chromosomal location, and cell surface receptors to which they bind to induce intra-
cellular signaling pathways (de Weerd and Nguyen 2012). There are more than 10
IFN I subtypes of IFN-α, named IFN-α1, IFN-α2, etc., IFN-β, IFN-κ, IFN-ω, IFN-ε,
and limitin. Type I IFNs signal by binding to heterodimeric receptor composed of
low- (IFNAR1) and high-affinity (IFNAR2). While IFN II signals through trans-
membrane tetrameric IFN-γ receptor (IFNGR), comprising two units of IFNGR1
and IFNGR2. Type III IFNs (IFN-λ1, -λ1, -λ2, -λ3, and -λ4) signal through IFN-λ
receptor (IFNLR), a heterodimer of IFNLR1 and IL10RB (Fensterl et al. 2015).
The functions of three IFNs in some instances overlap, for example, their ability
to inhibit virus replication; at the same time many effects of signaling by type I, II,
III IFNs are distinct. These differences may be attributed to two properties. First,
IFN and its cognate receptors show differential expression as there are cell- and
tissue-specific differences. Although most of the virally infected cells can produce
IFN-α/β, IFN-γ is synthesized by certain cells of the immune system such as natural
killer T cells, B cells, and antigen-presenting cells (Samuel 2001). Type I, type II,
and IL10RB receptors are expressed in almost all cell types, except that IFNGR2 is
not found on the surface of Th1 cells and low expression of IFNAR2 is seen in the
human brain (de Weerd and Nguyen 2012). Unlike type I and II IFN receptors, the
distribution of type III IFN receptor (IFNLR1) is more restricted. IFNAR is abun-
dantly expressed only in cells of epithelial origin and hepatocytes and responds to
type III IFNs (IFN-λ) (Mordstein et al. 2010; Sheahan et al. 2014; Sommereyns
et al. 2008) Second, downstream targets of IFNs are different and this leads to
induction of different IFN-stimulated genes (Der et al. 1998), and there is only par-
tial overlap between IFNAR, IFNLR, and IFNGR. The set of genes induced by
IFNAR and IFNLR is similar and they do so by activating intracellular signaling
pathway through transcription factor complex ISG factor 3 (ISGF3) (Doyle et al.
2006). In contrast, activation of the transcription factor gamma-activated factor
(GAF) by IFNGR induces different set of genes (Decker et al. 1997; Fensterl et al.
2015) (Fig. 4.1).
4 Interferons and Their Role in Viral Infection 63
Fig. 4.1 Summary of Type I, Type II, and Type III interferon signaling: Different cell surface
receptors are involved in signaling of Type I, Type II, and Type III interferons. Binding of IFNs to
cognate receptor triggers phosphorylation of STAT through kinases (JAK-1, JAK-2, and TYK-2).
Further, phosphorylated STAT undergoes oligomerization and interacts with IRF-9 in case of Type
I and III IFNs to form a complex of ISGF3. Similarly, in the case of Type II IFNs STAT undergoes
dimerization and associates with GAF to form a complex which then triggers the transcriptional
activity
further associate to form a heterodimer. The STAT heterodimer then interacts with
IFN-regulatory factor 9 (IRF9) to form the IFN-stimulated gene factor 3 (ISGF3).
Type II IFN dimers binding to the IFNGR1/2complex activate the cascade by phos-
phorylation of pre-associated JAK1 and JAK2 tyrosine kinases and transphosphory-
lation of the receptor chains leading to the recruitment and phosphorylation of
STAT1. Phosphorylated STAT1 homodimers form IFN-γ activation factor (GAF)
and translocate to the nucleus and bind to the gamma-activated sequence (GAS)
promoter elements, respectively, resulting in expression of antiviral genes. Similarly
the ISGF3 translocates to the nucleus to induce genes regulated by IFN-stimulated
response elements (ISRE), resulting in expression of antiviral genes.
Although IFNs are not expressed in resting cells, in virus-infected cells there is a
need to accelerate the synthesis of IFNs as they are effective in limiting the virus
replication and spread. As all the viruses replicate inside the cells, detecting viral
products can be an effective strategy for inducing an immune response through
production of IFNs. Viral nucleic acid (RNA or DNA) can trigger the synthesis of
IFNs through the pattern recognition receptors (PRRs) by engaging pathogen-
associated molecular patterns (PAMPs) (Thompson et al. 2011). Nucleic acid PRRs
can be endosomal membrane or cytosolic proteins. Recognition and binding of
PAMPs activates PRRs by conformational change, often by dimerization or oligo-
merization, and initiates intracellular signaling pathways resulting in the formation
of IFNs. In uninfected normal cells, the PRRs remain in an inactive conformation
unable to evoke an immune response. Toll-like receptors (TLRs) are receptors local-
ized within endosomes and plasma membrane and are efficient sensors of PAMPs
such as single-stranded RNA (ssRNA), lipopolysaccharides, lipoproteins, and fla-
gella. TLRs (TLR3, TLR7, TLR8, and TLR9) are nucleic acid sensing receptor
(Gay et al. 2014). TLR3 is a dsRNA sensor widely expressed in innate immune cells
and involved in the recognition of RNA viruses such as influenza A virus, picorna-
viruses, and DNA viruses such as herpes simplex virus-1 (Alexopoulou et al. 2001;
Le Goffic et al. 2006; Hardarson et al. 2007; Zhang et al. 2007). TLR-7 and TLR-8
are activated by ssRNA from many viruses such as vesicular stomatitis virus and
influenza A virus (Lee et al. 2007). DNA viruses are recognized by endosomal
TLR9 and several other cytoplasmic DNA sensors such as IFN-induced 16-kDA
protein (IFI16), cyclic GMP-AMP synthase (cGAS), DEAD-Box RNA Helicase
(DDX41), and DNA-dependent activator of IRFs (DAI) (Pandey et al. 2015; Wu and
Chen 2014). TLR9 can recognize unmethylated deoxycytidylate-phosphate-
deoxyguanylate (CpG) motif in viral DNA in plasmacytoid dendritic cells
(Krug et al. 2004). RIG-I-like receptor family (RLRs) is another closely related
family consisting of retinoic acid-inducible gene (RIG-I), melanoma differentia-
tion-associated gene 5(MDA-5), and laboratory of genetics and physiology-2
(LGP2). RLRs are cytosolic RNA helicases involved in recognition of RNA viruses
(Yoneyama et al. 2015). RLRs play a crucial role in antiviral defense pathway in cell
types such as fibroblasts, epithelial cells, and dendrite cells (Thompson et al. 2011).
4 Interferons and Their Role in Viral Infection 65
RIG-I uses 5′ end of the ssRNA as a strategy to distinguish between viral and host
RNA. In addition, RIG-I can also recognize short dsRNA product produced during
viral replication (Goubau et al. 2014; Weber et al. 2013; Yoneyama et al. 2015).
MDA-5 differentiates viral and host RNA based on the length of the sequences as
long as RNA is not common in host cells (Kato et al. 2008; Züst et al. 2011).
RIG-I recognizes Vesicular Stomatitis virus (VSV), rabies virus, Newcastle dis-
ease virus, measles virus, influenza A and B, hepatitis C virus, Japanese encephalitis
virus, and Ebola virus (Fensterl et al. 2015; Thompson et al. 2011). LGP-2 can act
via two different pathways during cytoplasmic recognition. LGP2 binds to dsRNA
in tandem with MDA5 and thereby activates MDA5 (Bruns et al. 2014; Childs et al.
2013). Second, it can inhibit RIG-I activation by competing for dsRNA and by
direct interaction with RIG I (Saito et al. 2007).
Detection of PAMPs results in the activation of downstream signaling pathways
leading to the induction of IFNs. IFN-α subtypes use transcription factor IRF-7 and
IFN-λs utilize IRF-3, IRF-7, and NF-κB (Génin et al. 2009; Iversen and Paludan
2010; Onoguchi et al. 2007; Osterlund et al. 2007). IFN-β is induced by the syner-
gistic promoter binding of transcription factors IRF-3, NF-κB, and API (Ford and
Thanos 2010; Panne et al. 2007). Once activated by the PAMP, PRRs interact with
respective adapter proteins; TLR3 uses the TRIF and TLR7, TLR8, and TLR9 use
My88, while RLRs use mitochondrial and peroxisome-bound MAVS (Fensterl et al.
2015). Cytoplasmic DNA sensors use STING (stimulator of interferon genes) for
their signaling. Further the TRIF and MAVS interact with signaling proteins of the
TRAF family of ubiquitin ligases, as well as IKKα/β and TBK1 kinases which
phosphorylate and activate downstream transcription factors (Fensterl et al. 2015).
Phosphorylation of IRF3 at specific serine and threonine residues results in opening
of its conformation and subsequent dimerization and translocation to nucleus where
it binds to IFN-stimulated response elements (ISREs) in the promoter region of
IFN-β and IFN-γ (Pandey et al. 2015). NF-κB another transcription factor upon
phosphorylation is released from its inhibitor IκB to induce IFN genes (Fig 4.2).
Evidence suggests that multiple signaling pathways are activated and cooperation as
well as coordinated response by these pathways is required for the generation of
response to IFNs. Some of the major pathways involved in IFN signaling are dealt
in sufficient detail below. In addition to the signaling pathway described here, there
are many other pathways whose involvement in IFN signaling is documented; in
fact the list of newly identified IFN signaling pathway is growing and it seems prob-
able that more than one signaling pathway is involved in IFN-mediated response.
IFNs are synthesized and secreted by virus-infected cells and it can elicit biological
responses by binding to specific receptors on the surface of the target cells. Binding
66 S. George et al.
Fig. 4.2 Signaling pathways for Type I and III interferons. Receptor proteins (toll-like receptors
(TLRs), RIG-I-like receptors (RLRs), and cyclic GMP-AMP synthase (cGAS) in endosomes or
cytoplasm recognize viral RNA or DNA and trigger signaling via adapter proteins such as TRIF,
My88, MAVS, and STING, which further activate the transcription factors of the IRF family and
NF-κB resulting in the transcription of Type I and Type III interferons
the nucleus and initiates transcription by binding specific sites in the promoters
(IFN-stimulated response elements) of IFN-stimulated genes (ISGs) (Ivashkiv and
Donlin 2014; Platanias 2005). In addition, IFN-induced complex can also bind to
another site known as IFN-γ-activated site (GAS) element (Platanias 2005). IFNs
induce the expression of hundreds of genes, of these some have only ISREs or only
GAS elements in their promoter, whereas others have both elements. Thus, for opti-
mal transcriptional activation a combination of different STATS complex is required
to produce functionally distinct genes (Platanias 2005). STAT1 is activated by JAK1
and JAK2 in response to IFN-γ engagement of type II IFN receptors. Phosphorylation
of STAT1 on the tyrosine residue at position 701 (Tyr701) results in the formation
of STAT1-STAT1 homodimers which translocate to the nucleus to bring about tran-
scription by binding to GAS elements (Aaronson 2002; Boehm et al. 1997; Stark
et al. 1998). Type II IFNs are different from type I as they do not form ISGF3 com-
plexes and thereby cannot induce the formation of genes that have only ISREs in
their promoter.
In addition to the Jak STAT pathway, IFNs activates many other signaling pathways.
Such a pathway includes the Crk family of adaptor proteins which includes three
members—CrkL, CrkI, and CrkII, all of which are cellular homologues of the v-Crk
gene (Platanias and Fish 1999; Sattler and Salgia 1998). CrkL is present in the latent
form in cytoplasm and is known to constitutively associate with the guanine nucleo-
tide exchange factor (GEF) C3G. Binding of the IFN to the respective receptor
results in the association of CrkL-C3G with TYK2 and is activated by phosphoryla-
tion of the tyrosine. The active Crkl now forms a complex with STAT-5 to form
Crkl-STAT5 complex, which subsequently translocates to the nucleus and binds to
GAS elements (TTCTAGGAA) located at the promoters of ISGs. Activation of Crkl
can also result in the GEF activity of the C3G, thereby resulting in the activation of
GTPase activity of RAPI, a member of the Ras family of small GTPases which
mediate induction of the growth suppressive effects by IFNs (Platanias 2005;
Schmitt and Stork 2001). Both type I and type II IFNs activate RAPI in a Crkl-
dependent manner (Lekmine et al. 2002). RAPI has also been shown to promote
cellular proliferation; thus, the effects on cell growth depend on the stimuli and cell
type that are involved (Hattori 2003).
(SH2) and SH3 domains which interact with other signaling proteins, while the
p110 regulates the phosphorylation of the D3 position of the phosphatidylinositol
lipids of the inositol ring. Several type I IFNs (IFNs-α, IFNs-β, IFNs-ω) have been
shown to induce tyrosine phosphorylation of IRS1 and the p85 subunit of PI3K
associates with IRS1 through its SH2 domains in an IFN-dependent manner result-
ing in activation of catalytic subunit of PI3K (p110) (Uddin et al. 1995). Similar to
IRS1, the IRS2 belonging to the same family was subsequently shown to undergo
phosphorylation in type I IFN-dependent manner (Burfoot et al. 1997; Platanias
et al. 1996). Thus, type I IFN can activate the PI3K signaling pathway in an IRS-
dependent and STAT independent manner (Ivashkiv and Donlin 2014).
Similar to type I IFNs, type II IFNs activate PI3K pathway but in an IRS indepen-
dent manner. The protein that seems to play a role in type II IFN signaling is CBL
which has binding sites for the SH3 domain of p85 and is shown to be phosphory-
lated in an IFN-γ-dependent manner (Alsayed et al. 2000). As mentioned above
JAK1 and JAK2 are activated in response to IFN-γ engagement of type II IFN
receptors. The activated JAK through an intermediate can also activate the p110 of
PI3K, which in turn activates protein kinase C-δ (PKC-δ). Finally, the PKC-δ regu-
lates the phosphorylation of STAT1 on the Ser727 (Nguyen et al. 2001). Although
the modification of STAT1 on the Ser727 is not required for its translocation to
nucleus, it is essential for transcriptional activation. Many downstream effectors of
PI3K signaling pathway have been identified such as 3-phosphatidylinositol-
dependent protein kinase 1 (PDK1), serine/threonine kinases (AKT), various iso-
forms of protein kinase C (PKC), and members of the TEC family of tyrosine
kinases (Platanias 2005).
stress activated kinase 1 (MSK1), and MAPK interacting protein kinase 1 (MNK1)
then mediate gene transcription of ISG protein products. p38 MAPK pathway is
also activated by the type II IFN and type III receptor and plays important roles in
the generation of type II IFN-biological responses in several different cell types
(Fensterl et al. 2015).
Binding of IFNs to its cognate receptors activates signaling cascades which culmi-
nates into the transcriptional induction of a number of antiviral genes called
interferon-stimulated genes (ISGs). These genes encode direct antiviral molecules
with potential to regulate IFNs signaling positively or negatively. Hundreds of ISGs
have been identified by genome-wide transcriptional profiling and most of the ISGs
mainly target conserved aspects of viral infection. Specific mechanisms of the ISGs
are summarized below (Schneider et al. 2014; Schoggins and Rice 2011).
The best characterized IFN-induced ISGs are the “classical ISGs,” interferon-induced,
double-stranded RNA-activated protein kinase encoded by Eif2ak2 (PKR), MX1 (the
myxovirus (influenza virus) resistance 1), and 2′-5′ oligoadenylate synthetase/RNase L
system (OAS1, OAS2, OAS3, encoded by Oas1, Oas2, Oas3). PKR is a serine/threo-
nine protein kinase and has been shown to be an inhibitor of cellular and viral mRNA
translation by phosphorylation of the protein synthesis initiation factor eIF2. OAS
enzyme system catalyzes the formation of 2′-5′ oligoadenylates, which then activate
RNase L to degrade viral genomes. The MX1 protein is a dynamin-like large guanosine
triphosphatase (GTPase) and it acts by trapping the viral components such as the nucleo-
capsids and prevents them from reaching their destination by limiting the viral growth.
Around 20 years ago, the expression of PML was detected in 98% of acute promy-
elocytic leukemia (APL) cases due to chromosomal translocation; later it was iden-
tified as INF-induced protein. The PML is known for inhibition of replication of
RNA and DNA viruses. The induction of PML enhances the expression of distinct
PML isoforms and nuclear bodies (NBs). Several PML isoforms have a key role in
transcription regulation and posttranslational modifications such as ubiquitinyl-
ation, SUMOylation, acetylation, and phosphorylation, which leads to consolidated
antiviral activity against specific viruses (Geoffroy and Chelbi-Alix 2011).
Type-1 and type III interferons chiefly induce IFIT family proteins. The genes for
these proteins are mainly conserved in mammals, amphibians, and fish but not in
lower animals like yeasts, fruit flies, etc. Presence of tetratricopeptide (TPR) mul-
tiple repeats is a unique property of these proteins. Some members of the IFIT fam-
ily act by binding to the subunits of the eIF3 translation initiation complex and
thereby reduce the efficiency of cellular cap-dependent protein translation. Besides,
IFIT can also function as viral RNA sensor by binding to uncapped 5′-ppp RNA and
sequestering it from the actively replicating pool. Studies have also shown that IFIT
can bind other proteins such as viral E1 helicase, a protein required for replication,
and prevent virus infection.
Humans consist of four IFITM proteins (IFITM1, IFITM2, IFITM3, and
IFITM5). Besides antiviral function, IFTM has been shown to play a role in many
biological processes such as cell signaling, germ cell homing and maturation, and
bone mineralization. IFN-induced transmembrane (IFITM) protein members inhibit
endocytic fusion events of a broad spectrum of viruses. However, further work has
to be done to establish how IFITM differentially restricts different viruses (Diamond
and Farzan 2013; Zhou et al. 2013).
4 Interferons and Their Role in Viral Infection 71
4.5.8 SAMHD1
The IFN system is one of the first lines of defense mechanism mounted by the host
against the virus. Virus in due course of evolution has acquired mechanisms to
antagonize the IFN-mediated responses by inhibiting the IFN-activated signaling
cascade or else by suppressing the antiviral pathways. In the following section of
this article, a brief account of how viruses evade the IFN system is described. For a
more detailed review, refer to García-Sastre (2017).
Viruses such as flaviviruses form replication complex inside membrane vesicles
also termed as viroplasm-like structures (VLS) that protect the viral RNA from
interacting with RLRs during RNA synthesis (den Boon and Ahlquist 2010). While
in case of influenza virus, the replication occurs in nucleus far away from cytosolic
RLRs and TLRs, it has been shown that RIGI can recognize the influenza virus
RNA genome during the transit from cytoplasm to the nucleus (Jackson et al. 1982;
Weber et al. 2015). The viral RNA with unmethylated 5′ tri- or diphosphate ends is
recognized by the RLRs RIG-I and MDA 5, and this in turn activates and induces
IFNs. In order to mimic the host, some RNA viruses post-translationally modify
RNA by adding 5′ cap structures similar to eukaryotic using viral coded phospha-
tases and methyltransferases. For some viruses that lack enzymes for posttransla-
tional addition of their own RNA cap, they utilize the host cellular mRNA expressing
viral endonucleases and utilize these caps to mask viral RNA (Reguera et al. 2010).
Many DNA viruses such as herpes virus due to their large coding sequences
encode viral protein capable of interacting with key mediators of IFN signaling and
thereby suppress IFN response. For example, inhibition by herpesviruses of the
STING, a signaling molecule which is downstream of cGAS, has been reported
(Chen et al. 2016). Similar to DNA virus, some RNA viruses such as arenavirus Z
protein (Fan et al. 2010) and paramyxovirus V proteins (Andrejeva et al. 2004) bind
4 Interferons and Their Role in Viral Infection 73
to and inhibit members of RIG-I like receptors. The influenza virus PB1-F2 protein
is shown to interact with the downstream adaptor mitochondrial protein MAVS
(Varga et al. 2012). Viral proteins are also shown to interact with a number of tran-
scription factors involved in IFN induction. The binding of ORF36 protein of murine
gamma-herpesvirus to activated IRF3 in the nucleus prevents induction of IFN
mRNA synthesis (Hwang et al. 2009) The NS5 protein of yellow fever virus binds
to STAT2 and this affects its antiviral action by inhibiting binding of this transcrip-
tion factor to the IFN responsive promoter elements of the ISGs (Laurent-Rolle
et al. 2014). Similarly a number of viral encoded proteins have been shown to bind
to STAT1 and STAT2 such as the P proteins of rabies (Vidy et al. 2005), the C6
protein of vaccinia virus (Stuart et al. 2016), and the V and W proteins of paramyxo-
viruses (Rodriguez et al. 2002; Shaw et al. 2004).
Direct regulation of phosphorylation events involved in IFN induction is another
attractive strategy used by virus. These kinases are target of many viral encoded
proteins, for example, the IKKε kinase inhibition by the N protein of arenaviruses
(Pythoud et al. 2012). The NS4B protein of flaviviruses binds to and inhibits tank
binding kinase 1(TBK1) (Dalrymple et al. 2015), VP40 of Ebola virus inhibits the
JAK1 (Valmas et al. 2010), and TYK2 inhibition by LMP1 of Epstein-Barr virus
(Geiger and Martin 2006). IFN signaling is yet another pathway targeted by the
viruses to evade the host. The IFNAR1 chain of the IFN receptor is the preferred
target for degradation by phosphorylation-dependent ubiquitination, and this affects
type I signaling and antiviral defense (Liu et al. 2009).
Many viral proteases that are involved in the processing of the viral polyprotein
are also shown to cleave and degrade proteins necessary for IFN response. This is
most commonly seen in positive-strand RNA viruses. For example, the hepatitis C
virus protease cleaves MAVS (Li et al. 2005; Meylan et al. 2005), while the dengue
virus protease cleaves STING (Aguirre et al. 2012; Yu et al. 2012). Another mecha-
nism by which virus degrades host proteins is by marking the protein by addition of
K48 polyubiquitin chains makes it susceptible to proteosome degradation. This is
exemplified by NS5 of Zika virus that targets human STAT2 for proteosomal degra-
dation in infected cells (Grant et al. 2016).
Viruses have been shown to prevent gene expression by epigenetic regulation,
thereby interfering with transcription mechanism of the host. Adenovirus E1A pro-
tein has been shown to bind to and dissociate the host nuclear complex that is
required for histone monoubiquitination at H2B lysine 120 (Fonseca et al. 2012).
This modification prevents the opening of the chromatin to allow transcription, and
therefore IFN is unable to activate transcription of the ISGs. In addition to transcrip-
tional shutoff, viruses can suppress host gene expression by other mechanisms such
as the posttranscriptional inhibition of cellular RNA processing, trafficking, and
translational shutoff. The NS1 protein of influenza virus has been shown to prevent
the termination as well as polyadenylation of cellular mRNA (Nemeroff et al. 1998).
The M protein of VSV targets the nuclear pore components and thereby inhibits the
RNA export from the nucleus (von Kobbe et al. 2000). Similarly the ORF10 of the
Kaposi’s sarcoma-associated herpesvirus inhibits the translocation of mRNA of
genes coding for mitosis, gene silencing, DNA metabolic process, chromosome
74 S. George et al.
organization, cell cycle, and transcription regulation (Gong et al. 2016). These
genes although not involved in direct IFN response, may be detrimental for survival
of virus. Translational shutdown of host gene expression is another strategy used by
many viruses. This is best exemplified by the hepatitis C virus infection during
which the PKR mediated phosphorylation of the translation factor eiF2α results in
the inhibition of cellular translation, while the hepatitis mRNA translation continues
as it depends on a different mechanism of 5′ ribosomal entry site that is insensitive
to PKR mediated translation inhibition (Garaigorta and Chisari 2009). Many viruses
recruit decoy proteins to sequester the host proteins and prevent its binding to spe-
cific targets. The B18R protein of vaccinia virus, which is a poxvirus-encoded sol-
uble IFN receptor, can sequester IFN before it can bind to the IFN receptor (Symons
et al. 1995). Similarly the K3L protein of the poxvirus acts as a decoy for PKR
substrates (Beattie et al. 1991). Thus, a virus may use any of the above mechanism
or multiple mechanisms as a strategy to suppress the effect of IFN-mediated host
response.
As the virus hijacks the host immune response and causes chronic infection, for
elimination and controlling the virus replication a therapeutic intervention is needed.
The strong antiviral state conferred by the IFNs to the host cells combined with its
ability to modulate the immune system has made it an ideal candidate to be used in
antiviral therapy. IFN-α was initially used for the treatment of hepatitis C virus
(HCV) infection, but due to its short half-life and rapid elimination from the body
was unable to achieve the desired effect (Lin and Young 2014). In order to maintain
stable level in the blood, pegylated IFN was used for the treatment of chronic
HCV. IFN-α is also found to be effective against hepatitis B virus infection. Besides
the antiviral activity IFN-α is a powerful immune modulator and the studies related
to its efficiency as an adjuvant have met with limited success (Rizza et al. 2008).
IFN-α treatment is accompanied by systemic side effects such as neutropenia and
thrombocytopenia and as a remedy other IFN molecules such as albumin-IFN-αand
consensus IFN (CIFN) have been developed to replace PEG-IFN-α. CIFN with
10–100 times higher antiviral activity compared to standard IFN-α is a recombinant
protein and is found to be effective at a lower dose with limited side effects (Sjogren
et al. 2007). CIFN is a recombinant IFN with most commonly observed amino acids
from several type I IFN subtypes. Similar to type I IFNs, other classes of IFNs have
been shown to be useful in therapeutic intervention during viral infections. IFN-γ
has been shown to be protective against HIV-associated opportunistic infection with
or without highly active antiretroviral therapy (Jarvis et al. 2012). A comparison of
treatment with PEG-IFN-α and PEG-IFN-λ along with ribavirin showed higher
rates of rapid virologic response regardless of the HCV genotypes. Unlike IFN-α,
treatment of HCV with IFN-λ is much safer and less toxic as the restricted tissue
expression of the IFN-λR1 chain ensures a localized reaction (Lin and Young 2014).
4 Interferons and Their Role in Viral Infection 75
References
Aaronson DS (2002) A road map for those who don’t know JAK-STAT. Science 296(5573):1653–
1655. https://doi.org/10.1126/science.1071545
Aguirre S, Maestre AM, Pagni S, Patel JR, Savage T, Gutman D et al (2012) DENV inhibits type
I IFN production in infected cells by cleaving human STING. PLoS Pathog 8(10):e1002934.
https://doi.org/10.1371/journal.ppat.1002934
Alexopoulou L, Holt AC, Medzhitov R, Flavell RA (2001) Recognition of double-stranded RNA
and activation of NF-kappaB by toll-like receptor 3. Nature 413(6857):732–738. https://doi.
org/10.1038/35099560
Alsayed Y, Uddin S, Ahmad S, Majchrzak B, Druker BJ, Fish EN, Platanias LC (2000) IFN-
gamma activates the C3G/Rap1 signaling pathway. Journal of Immunology (Baltimore, Md.
1950) 164(4):1800–1806. http://www.ncbi.nlm.nih.gov/pubmed/10657627
Andrejeva J, Childs KS, Young DF, Carlos TS, Stock N, Goodbourn S, Randall RE (2004) The
V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its
activation of the IFN-beta promoter. Proc Natl Acad Sci U S A 101(49):17264–17269. https://
doi.org/10.1073/pnas.0407639101
Beattie E, Tartaglia J, Paoletti E (1991) Vaccinia virus-encoded eIF-2 alpha homolog abrogates
the antiviral effect of interferon. Virology 183(1):419–422. http://www.ncbi.nlm.nih.gov/
pubmed/1711259
Boehm U, Klamp T, Groot M, Howard JC (1997) Cellular responses to interferon-γ. Annu Rev
Immunol 15(1):749–795. https://doi.org/10.1146/annurev.immunol.15.1.749
Bruns AM, Leser GP, Lamb RA, Horvath CM (2014) The innate immune sensor LGP2 activates
antiviral signaling by regulating MDA5-RNA interaction and filament assembly. Mol Cell
55(5):771–781. https://doi.org/10.1016/j.molcel.2014.07.003
Burfoot MS, Rogers NC, Watling D, Smith JM, Pons S, Paonessaw G et al (1997) Janus kinase-
dependent activation of insulin receptor substrate 1 in response to interleukin-4, oncostatin M,
and the interferons. The Journal of Biological Chemistry 272(39):24183–24190. http://www.
ncbi.nlm.nih.gov/pubmed/9305869
Chen Q, Sun L, Chen ZJ (2016) Regulation and function of the cGAS-STING pathway of cytosolic
DNA sensing. Nat Immunol 17(10):1142–1149. https://doi.org/10.1038/ni.3558
Childs KS, Randall RE, Goodbourn S (2013) LGP2 plays a critical role in sensitizing mda-5 to
activation by double-stranded RNA. PLoS One 8(5):e64202. https://doi.org/10.1371/journal.
pone.0064202
Dalrymple NA, Cimica V, Mackow ER (2015) Dengue virus NS proteins inhibit RIG-I/MAVS
signaling by blocking TBK1/IRF3 phosphorylation: dengue virus serotype 1 NS4A is a
unique interferon-regulating virulence determinant. MBio 6(3):e00553–e00515. https://doi.
org/10.1128/mBio.00553-15
de Weerd NA, Nguyen T (2012) The interferons and their receptors – distribution and regulation.
Immunol Cell Biol 90(5):483–491. https://doi.org/10.1038/icb.2012.9
Decker T, Kovarik P, Meinke A (1997) GAS elements: a few nucleotides with a major impact
on cytokine-induced gene expression. J Interf Cytokine Res 17(3):121–134. https://doi.
org/10.1089/jir.1997.17.121
den Boon JA, Ahlquist P (2010) Organelle-like membrane compartmentalization of positive-strand
RNA virus replication factories. Annu Rev Microbiol 64:241–256. https://doi.org/10.1146/
annurev.micro.112408.134012
Der SD, Zhou A, Williams BR, Silverman RH (1998) Identification of genes differentially regu-
lated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proceedings of the
National Academy of Sciences of the United States of America 95(26):15623–15628. http://
www.ncbi.nlm.nih.gov/pubmed/9861020
Diamond MS, Farzan M (2013) The broad-spectrum antiviral functions of IFIT and IFITM pro-
teins. Nat Rev Immunol 13(1):46–57. https://doi.org/10.1038/nri3344
76 S. George et al.
Ivashkiv LB, Donlin LT (2014) Regulation of type I interferon responses. Nat Rev Immunol
14(1):36–49. https://doi.org/10.1038/nri3581
Iversen MB, Paludan SR (2010) Mechanisms of type III interferon expression. J Interf Cytokine
Res 30(8):573–578. https://doi.org/10.1089/jir.2010.0063
Jackson DA, Caton AJ, McCready SJ, Cook PR (1982) Influenza virus RNA is synthesized
at fixed sites in the nucleus. Nature 296(5855):366–368. http://www.ncbi.nlm.nih.gov/
pubmed/7063035
Jarvis JN, Meintjes G, Rebe K, Williams GN, Bicanic T, Williams A et al (2012) Adjunctive
interferon-γ immunotherapy for the treatment of HIV-associated cryptococcal meningi-
tis: a randomized controlled trial. AIDS (London, England) 26(9):1105–1113. https://doi.
org/10.1097/QAD.0b013e3283536a93
Kato H, Takeuchi O, Mikamo-Satoh E, Hirai R, Kawai T, Matsushita K et al (2008) Length-
dependent recognition of double-stranded ribonucleic acids by retinoic acid–inducible gene-I
and melanoma differentiation–associated gene 5. J Exp Med 205(7):1601–1610. https://doi.
org/10.1084/jem.20080091
Krug A, French AR, Barchet W, Fischer JAA, Dzionek A, Pingel JT et al (2004) TLR9-
dependent recognition of MCMV by IPC and DC generates coordinated cytokine responses
that activate antiviral NK cell function. Immunity 21(1):107–119. https://doi.org/10.1016/j.
immuni.2004.06.007
Laurent-Rolle M, Morrison J, Rajsbaum R, Macleod JML, Pisanelli G, Pham A et al (2014) The
interferon signaling antagonist function of yellow fever virus NS5 protein is activated by type
I interferon. Cell Host Microbe 16(3):314–327. https://doi.org/10.1016/j.chom.2014.07.015
Le Goffic R, Balloy V, Lagranderie M, Alexopoulou L, Escriou N, Flavell R et al (2006) Detrimental
contribution of the toll-like receptor (TLR)3 to influenza a virus–induced acute pneumonia.
PLoS Pathog 2(6):e53. https://doi.org/10.1371/journal.ppat.0020053
Le Tortorec A, Willey S, Neil SJD (2011) Antiviral inhibition of enveloped virus release by teth-
erin/BST-2: action and counteraction. Viruses 3(5):520–540. https://doi.org/10.3390/v3050520
Lee HK, Lund JM, Ramanathan B, Mizushima N, Iwasaki A (2007) Autophagy-dependent viral
recognition by plasmacytoid dendritic cells. Science 315(5817):1398–1401. https://doi.
org/10.1126/science.1136880
Lekmine F, Sassano A, Uddin S, Majchrzak B, Miura O, Druker BJ et al (2002) The CrkL adapter
protein is required for type I interferon-dependent gene transcription and activation of the small
G-protein Rap1. Biochem Biophys Res Commun 291(4):744–750. https://doi.org/10.1006/
bbrc.2002.6516
Li X-D, Sun L, Seth RB, Pineda G, Chen ZJ (2005) Hepatitis C virus protease NS3/4A cleaves
mitochondrial antiviral signaling protein off the mitochondria to evade innate immunity. Proc
Natl Acad Sci U S A 102(49):17717–17722. https://doi.org/10.1073/pnas.0508531102
Li M, Zhang D, Zhu M, Shen Y, Wei W, Ying S et al (2017) Roles of SAMHD1 in antiviral defense,
autoimmunity and cancer. Rev Med Virol 27(4). https://doi.org/10.1002/rmv.1931
Lin F, Young HA (2014) Interferons: success in anti-viral immunotherapy. Cytokine Growth Factor
Rev 25(4):369–376. https://doi.org/10.1016/j.cytogfr.2014.07.015
Liu J, HuangFu W-C, Kumar KGS, Qian J, Casey JP, Hamanaka RB et al (2009) Virus-induced
unfolded protein response attenuates antiviral defenses via phosphorylation-dependent degra-
dation of the type I interferon receptor. Cell Host Microbe 5(1):72–83. https://doi.org/10.1016/j.
chom.2008.11.008
Liu S-Y, Aliyari R, Chikere K, Li G, Marsden MD, Smith JK et al (2013) Interferon-inducible
cholesterol-25-hydroxylase broadly inhibits viral entry by production of 25-hydroxycholesterol.
Immunity 38(1):92–105. https://doi.org/10.1016/j.immuni.2012.11.005
Meylan E, Curran J, Hofmann K, Moradpour D, Binder M, Bartenschlager R, Tschopp J (2005)
Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus.
Nature 437(7062):1167–1172. https://doi.org/10.1038/nature04193
Morales DJ, Lenschow DJ (2013) The antiviral activities of ISG15. J Mol Biol 425(24):4995–
5008. https://doi.org/10.1016/j.jmb.2013.09.041
78 S. George et al.
Schmitt JM, Stork PJS (2001) Cyclic AMP-mediated inhibition of cell growth requires
the small G protein Rap1. Mol Cell Biol 21(11):3671–3683. https://doi.org/10.1128/
MCB.21.11.3671-3683.2001
Schneider WM, Chevillotte MD, Rice CM (2014) Interferon-stimulated genes: a com-
plex web of host defenses. Annu Rev Immunol 32(1):513–545. https://doi.org/10.1146/
annurev-immunol-032713-120231
Schoggins JW, Rice CM (2011) Interferon-stimulated genes and their antiviral effector functions.
Curr Opin Virol 1(6):519–525. https://doi.org/10.1016/j.coviro.2011.10.008
Shaw ML, García-Sastre A, Palese P, Basler CF (2004) Nipah virus V and W proteins have a
common STAT1-binding domain yet inhibit STAT1 activation from the cytoplasmic and
nuclear compartments, respectively. J Virol 78(11):5633–5641. https://doi.org/10.1128/
JVI.78.11.5633-5641.2004
Sheahan T, Imanaka N, Marukian S, Dorner M, Liu P, Ploss A, Rice CM (2014) Interferon lambda
alleles predict innate antiviral immune responses and hepatitis C virus permissiveness. Cell
Host Microbe 15(2):190–202. https://doi.org/10.1016/j.chom.2014.01.007
Sjogren MH, Sjogren R, Lyons MF, Ryan M, Santoro J, Smith C et al (2007) Antiviral response of
HCV genotype 1 to consensus interferon and ribavirin versus pegylated interferon and ribavi-
rin. Dig Dis Sci 52(6):1540–1547. https://doi.org/10.1007/s10620-007-9757-9
Sommereyns C, Paul S, Staeheli P, Michiels T (2008) IFN-lambda (IFN-lambda) is expressed
in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PLoS Pathog
4(3):e1000017. https://doi.org/10.1371/journal.ppat.1000017
Stark GR, Kerr IM, Williams BRG, Silverman RH, Schreiber RD (1998) How cells respond to inter-
ferons. Annu Rev Biochem 67(1):227–264. https://doi.org/10.1146/annurev.biochem.67.1.227
Stuart JH, Sumner RP, Lu Y, Snowden JS, Smith GL (2016) Vaccinia virus protein C6 inhibits type
I IFN signalling in the nucleus and binds to the transactivation domain of STAT2. PLoS Pathog
12(12):e1005955. https://doi.org/10.1371/journal.ppat.1005955
Symons JA, Alcamí A, Smith GL (1995) Vaccinia virus encodes a soluble type I interferon receptor
of novel structure and broad species specificity. Cell 81(4):551–560. http://www.ncbi.nlm.nih.
gov/pubmed/7758109
Thompson MR, Kaminski JJ, Kurt-Jones EA, Fitzgerald KA (2011) Pattern recognition recep-
tors and the innate immune response to viral infection. Viruses 3(6):920–940. https://doi.
org/10.3390/v3060920
Uddin S, Yenush L, Sun XJ, Sweet ME, White MF, Platanias LC (1995) Interferon-alpha engages
the insulin receptor substrate-1 to associate with the phosphatidylinositol 3′-kinase. J Biol
Chem 270(27):15938–15941. http://www.ncbi.nlm.nih.gov/pubmed/7608146
Valmas C, Grosch MN, Schümann M, Olejnik J, Martinez O, Best SM et al (2010) Marburg
virus evades interferon responses by a mechanism distinct from Ebola virus. PLoS Pathog
6(1):e1000721. https://doi.org/10.1371/journal.ppat.1000721
Varga ZT, Grant A, Manicassamy B, Palese P (2012) Influenza virus protein PB1-F2 inhibits the
induction of type I interferon by binding to MAVS and decreasing mitochondrial membrane
potential. J Virol 86(16):8359–8366. https://doi.org/10.1128/JVI.01122-12
Vidy A, Chelbi-Alix M, Blondel D (2005) Rabies virus P protein interacts with STAT1 and inhibits
interferon signal transduction pathways. J Virol 79(22):14411–14420. https://doi.org/10.1128/
JVI.79.22.14411-14420.2005
von Kobbe C, van Deursen JM, Rodrigues JP, Sitterlin D, Bachi A, Wu X et al (2000) Vesicular
stomatitis virus matrix protein inhibits host cell gene expression by targeting the nucleoporin
Nup98. Mol Cell 6(5):1243–1252. http://www.ncbi.nlm.nih.gov/pubmed/11106761
Weber M, Gawanbacht A, Habjan M, Rang A, Borner C, Schmidt AM et al (2013) Incoming RNA
virus nucleocapsids containing a 5′-triphosphorylated genome activate RIG-I and antiviral sig-
naling. Cell Host Microbe 13(3):336–346. https://doi.org/10.1016/j.chom.2013.01.012
Weber M, Sediri H, Felgenhauer U, Binzen I, Bänfer S, Jacob R, Brunotte L, García-Sastre A,
Schmid-Burgk JL, Schmidt T, Hornung V, Kochs G, Schwemmle M, Klenk H-D, Weber F
(2015) Influenza virus adaptation PB2-627K modulates nucleocapsid inhibition by the patho-
gen sensor RIG-I. Cell Host Microbe 17(3):309–319
80 S. George et al.
Wen Z, Darnell JE (1997) Mapping of Stat3 serine phosphorylation to a single residue (727) and
evidence that serine phosphorylation has no influence on DNA binding of Stat1 and Stat3. Nucl
Acids Res 25(11):2062–2067. http://www.ncbi.nlm.nih.gov/pubmed/9153303
Wen Z, Zhong Z, Darnell JE (1995) Maximal activation of transcription by Stat1 and Stat3 requires
both tyrosine and serine phosphorylation. Cell 82(2):241–250. http://www.ncbi.nlm.nih.gov/
pubmed/7543024
Wu J, Chen ZJ (2014) Innate immune sensing and signaling of cytosolic nucleic acids. Annu Rev
Immunol 32(1):461–488. https://doi.org/10.1146/annurev-immunol-032713-120156
Yoneyama M, Onomoto K, Jogi M, Akaboshi T, Fujita T (2015) Viral RNA detection by RIG-I-like
receptors. Curr Opin Immunol 32:48–53. https://doi.org/10.1016/j.coi.2014.12.012
Yu C-Y, Chang T-H, Liang J-J, Chiang R-L, Lee Y-L, Liao C-L, Lin Y-L (2012) Dengue virus tar-
gets the adaptor protein MITA to subvert host innate immunity. PLoS Pathog 8(6):e1002780.
https://doi.org/10.1371/journal.ppat.1002780
Zhang S-Y, Jouanguy E, Ugolini S, Smahi A, Elain G, Romero P et al (2007) TLR3 deficiency
in patients with herpes simplex encephalitis. Science 317(5844):1522–1527. https://doi.
org/10.1126/science.1139522
Zhou X, Michal JJ, Zhang L, Ding B, Lunney JK, Liu B, Jiang Z (2013) Interferon induced IFIT
family genes in host antiviral defense. Int J Biol Sci 9(2):200–208. https://doi.org/10.7150/
ijbs.5613
Züst R, Cervantes-Barragan L, Habjan M, Maier R, Neuman BW, Ziebuhr J et al (2011) Ribose
2’-O-methylation provides a molecular signature for the distinction of self and non-self mRNA
dependent on the RNA sensor Mda5. Nat Immunol 12(2):137–143. https://doi.org/10.1038/
ni.1979
Role of Cytokines in Infectious Viral
Disease 5
Pavani Sanapala and Sudhakar Pola
Abstract
Cytokines are cell signaling polypeptides. They regulate and promote immune
response, mostly related to autoimmunity. Role of cytokines in viral disease as
cytokine biology in recent studies shows multifaceted interactions that are
directly involved in the advancement of disease pathogenesis. Viruses induce the
expression of numerous cytokine protein genes that are activated from the dor-
mant state, and can also stimulate the expression or replication of extremely
identical cytokines. Cytokines are also involved in treating viral diseases, espe-
cially human interferons. The ongoing developmental swift in the area of cyto-
kines has led to the eventual growth of new antiviral therapeutic approaches
rooted in mimicking the cytokine network. Some cytokines, especially IL-12 in
combination with interferon γ, decrease the pathogenicity of viruses. Likewise,
cytokines induce or increase the viral pathogenicity, for instance, Leishmania
major, where IL-4 has a deleterious effect. This chapter details the salient fea-
tures of cytokines, classification, inter-relation of cytokines- viral expression and
signaling pathways of viruses inducing cytokines production.
Keywords
Cytokines · Chemokines · Viruses · NF-κβ activity · Signal transduction · viral
infections
5.1 Introduction
5.2 Cytokines
Cytokines are low molecular mass proteins habitually below 30 kDa in size. These
polypeptides are active on signaling molecules and cells invigorating them toward
sites of inflammation, infections, and traumas (Ferreira et al. 2018). Cytokines are
emanated by various cells at confined higher concentrations, and entailed in the cell
to cell communications either at the site they originated (autocrine effect) or at the
adjacent cells (paracrine fashion), or on distant cells (endocrine effect/systemic
effect) (Stadnyk 1994). As a result, the frontier between hormones and cytokines is
quite unclear. Indeed, standard hormones, for instance, prolactin (PRL), growth hor-
mone (GH), leptin, and erythropoietin (EPO), are all evidently cytokines confirmed
by their receptor structure and their manner of signaling. Perhaps it is just simplest
to accept that cell–cell communication and host defense went hand in hand during
evolution, and so functional and structural resemblance exists among families of
5 Role of Cytokines in Infectious Viral Disease 83
molecules that act on the immune, hematopoietic, endocrine, and nervous systems
(O’Shea et al. 2019).
Earlier many attempts for a suitable nomenclature were made, but Cohen et al.
emphasized that no single cell as a source is responsible for origination, as cyto-
kines are made by one or more cell types (Cohen et al. 1974). The first defined
cytokines were lymphokines a soluble product originated from lymphocytes in
stimuli to the polyclonal antigen. Other such previous attempts were monokines for
monocytes products. However, this nomenclature or classification did not seem apt
since few cytokines, namely, interleukin 6 and tumor necrosis factor α, were made
up of both cell types (Feldmann 1998). Cytokine may act interactively or antipa-
thetically (Zhang and An 2007).
Nearly all varied cytokines have a key role in many viral infections. Different
approaches such as viruses modulating cytokine expression, cytokines promoting
viral expression and replication, and a virus with cytokines or their receptors mim-
icking host immune response result in the cure of viral diseases.
is gathered as a result of tissues and cells analysis through in situ and ex vivo
techniques.
The resistant reaction to viral diseases is a mix of numerous actions: viral antigen
introduction to the immune framework, multiplication of particular lymphocytes,
and cytokine mediation (Cavallo et al. 1994). A sign of viral contamination is an
intense response by the affected cell. This incorporates enactment of a previous
antiviral resistant mechanism, obligation headed for programmed cell death and
generation of explicit cytokines. These actions add toward the decrease of viral
duplication and the confinement of viral spread. Cytokines and chemokines have a
vital role in the host reaction to viral contagion just like in the immunopathology
related to numerous viral ailments. Numerous viral glycoproteins in association
with specific cell receptors mediate the direct synthesis of cytokines. Furthermore,
viral RNA and viral proteins endorse viral replication and pro-inflammatory protein
expression when meddling with cell signaling factor activity such as transduction
and transcription (Mogensen and Paludan 2001).
Subsequent to viral infection, viral surface proteins in many viruses principally act
on the cellular surface protein initiating a cell response where in many cases directs
the primary signal of cytokine production. As well, some viruses in the absence of
viral proteins during initial virus infection can generate cytokines as proteins are
produced at some stage of the viral life cycle. However, the increase of viral RNA
and excess synthesis of the cellular macromolecule induce signs that activate early
host response to the infection.
process on increasing the cytoplasmic levels (Crabtree 1999). NF-κβ protein is gen-
erally originated in the cytoplasm in composite with Iκβ, an inhibitory protein.
Fig. 5.1 Activation of NF-κβ by viruses. The picture depicts different viruses, their proteins, and
pathways stimulating cellular response through cell membrane receptors
5 Role of Cytokines in Infectious Viral Disease 87
Cytokines besides their role as mediators in local and systematic host antiviral
response are able to endorse viral replication and multiply some of viruses, espe-
cially by IFNs and TNF. The well-known suitable example explained is
HIV. Activation of HIV expression from inactive state of replication is reliant partly
92 P. Sanapala and S. Pola
on the stimulation of the host cell (Rosenberg and Fauci 1990). Activation of CD4+
lymphocytes and monocytes is linked with the production of a number of cytokines
that induce the expression of HIV from chronic infected T lymphocyte and mono-
cyte cell lines. Specifically, the cytokines TNF-α and IL-6 upbeat transcriptional
and posttranscriptional expression levels (Poli et al. 1990).
Conflict of Interest The authors declare that they have no competing interests.
References
Adamson AL, Darr D, Holley-Guthrie E, Johnson RA, Mauser A, Swenson J, Kenney S (2000)
Epstein-Barr virus immediate-early proteins BZLF1 and BRLF1 activate the ATF2 transcrip-
tion factor by increasing the levels of phosphorylated p38 and c-Jun N-terminal kinases. J Virol
74(3):1224–1233
Al-Wabel A, Al-Janadi M, Raziuddin S (1993) Cytokine profile of viral and autoimmune chronic
active hepatitis. J Allergy Clin Immunol 92(6):902–908
Ameglio F, Capobianchi MR, Castilletti C, Fei PC, Fais S, Trento E, Dianzani F (1994)
Recombinant gpl 20 induces IL-10 in resting peripheral blood mononuclear cells; correlation
with the induction of other cytokines. Clin Exp Immunol 95(3):455–458
Ankel H, Capobianchi MR, Castilletti C, Dianzani F (1994) Interferon induction by HIV glycopro-
tein 120: role of the V3 loop. Virology 205(1):34–43
Arora DJ, Houde M (1992) Modulation of murine macrophage responses stimulated with influ-
enza glycoproteins. Can J Microbiol 38(3):188–192
Azimi N, Brown K, Bamford RN, Tagaya Y, Siebenlist U, Waldmann TA (1998) Human T cell
lymphotropic virus type I Tax protein trans-activates interleukin 15 gene transcription through
an NF-κB site. Proc Natl Acad Sci 95(5):2452–2457
Baba M, Imai T, Yoshida T, Yoshie O (1996) Constitutive expression of various chemokine genes
in human T-cell lines infected with human T-cell leukemia virus type 1: role of the viral trans-
activator Tax. Int J Cancer 66(1):124–129
Balkwill FR, Burke F (1989) The cytokine network. Immunol Today 10(9):299–304
Barrett KE (1996) Cytokines: sources, receptors and signaling. Baillieres Clin Gastroenterol
10(1):1–5
Baumann B, Kistler B, Kirillov A, Bergman Y, Wirth T (1998) The mutant plasmacytoma cell line
S107 allows the identification of distinct pathways leading to NF-κB activation. J Biol Chem
273(19):11448–11455
Baumforth KR, Young LS, Flavell KJ, Constandinou C, Murray PG (1999) The Epstein-Barr virus
and its association with human cancers. Mol Pathol 52(6):307
Benn J, Schneider RJ (1994) Hepatitis B virus HBx protein activates Ras-GTP complex for-
mation and establishes a Ras, Raf, MAP kinase signaling cascade. Proc Natl Acad Sci
91(22):10350–10354
Benn J, Su F, Doria M, Schneider RJ (1996) Hepatitis B virus HBx protein induces transcrip-
tion factor AP-1 by activation of extracellular signal-regulated and c-Jun N-terminal mitogen-
activated protein kinases. J Virol 70(8):4978–4985
Bixler S, Goff A (2015) The role of cytokines and chemokines in filovirus infection. Viruses
7(10):5489–5507
Blut A (2009) Influenza virus. Transfus Med Hemother 36(1):32
5 Role of Cytokines in Infectious Viral Disease 93
Hurst SM, Wilkinson TS, McLoughlin RM, Jones S, Horiuchi S, Yamamoto N, Rose-John S, Fuller
GM, Topley N, Jones SA (2001) Il-6 and its soluble receptor orchestrate a temporal switch in
the pattern of leukocyte recruitment seen during acute inflammation. Immunity 14(6):705–714
Imanishi J (2000) Expression of cytokines in bacterial and viral infections and their biochemical
aspects. J Biochem 127(4):525–530
Kallas EG, Reynolds K, Andrews J, Treanor JJ, Evans TG (1999) Production of influenza-
stimulated tumor necrosis factor-alpha by monocytes following acute influenza infection in
humans. J Interf Cytokine Res 19(7):751–755
Kanamori H, Suzuki N, Siomi H, Nosaka T, Sato A, Sabe H, Hatanaka M, Honjo T (1990)
HTLV-1 p27rex stabilizes human interleukin-2 receptor alpha chain mRNA. EMBO J 9(12):
4161. -4166, 1990
Kim S, Kehrl JH, Burton J, Tendler CL, Jeang KT, Danielpour D, Thevenin C, Kim KY, Sporn MB,
Roberts AB (1990) Transactivation of the transforming growth factor beta 1 (TGF-beta 1) gene
by human T lymphotropic virus type 1 tax: a potential mechanism for the increased production
of TGF-beta 1 in adult T cell leukemia. J Exp Med 172(1):121–129
Kuhn JH, Becker S, Ebihara H, Geisbert TW, Johnson KM, Kawaoka Y, Lipkin WI, Negredo
AI, Netesov SV, Nichol ST, Palacios G (2010) Proposal for a revised taxonomy of the fam-
ily Filoviridae: classification, names of taxa and viruses, and virus abbreviations. Arch Virol
155(12):2083–2103
Kujime K, Hashimoto S, Gon Y, Shimizu K, Horie T (2000) p38 mitogen-activated protein kinase
and c-jun-NH2-terminal kinase regulate RANTES production by influenza virus-infected
human bronchial epithelial cells. J Immunol 164(6):3222–3228
Lannuzel A, Tardieu M, Hery C, Barnier JV, Vincent JD, Guibert B, Van Tan H, Gray F (1997)
Human immunodeficiency virus type 1 and its coat protein gp 120 induce apoptosis and
activate JNK and ERK mitogen-activated protein kinases in human neurons. Ann Neurol
42(6):847–856
Lauder SN, Jones E, Smart K, Bloom A, Williams AS, Hindley JP, Ondondo B, Taylor PR, Clement
M, Fielding C, Godkin AJ (2013) Interleukin-6 limits influenza-induced inflammation and pro-
tects against fatal lung pathology. Eur J Immunol 43(10):2613–2625
Lay JD, Tsao CJ, Chen JY, Kadin ME, Su IJ (1997) Upregulation of tumor necrosis factor-alpha
gene by Epstein-Barr virus and activation of macrophages in Epstein-Barr virus-infected T
cells in the pathogenesis of hemophagocytic syndrome. J Clin Invest 100(8):1969–1979
Liang TJ, Hepatitis B (2009) The virus and disease. Hepatology 49(5 Suppl):S13–S21
Lin Y, Nomura T, Cheong J, Dorjsuren D, Iida K, Murakami S (1997) Hepatitis B virus X protein
is a transcriptional modulator that communicates with transcription factor IIB and the RNA
polymerase II subunit 5. J Biol Chem 272(11):7132–7139
Luciw PA (1996) Human immunodeficiency viruses and their replication. In: Fields BN (ed)
Virology, vol 2, 3rd edn. Lippincott-Raven, Philadelphia, pp 1881–1952
Mahe Y, Mukaida N, Kuno K, Akiyama M, Ikeda N, Matsushima K, Murakami S (1991) Hepatitis
B virus X protein transactivates human interleukin-8 gene through acting on nuclear factor κB
and CCAAT/enhancer-binding protein-like cis-elements. J Biol Chem 266(21):13759–13763
Marié I, Durbin JE, Levy DE (1998) Differential viral induction of distinct interferon-α genes by
positive feedback through interferon regulatory factor-7. EMBO J 17(22):6660–6669
Marsters SA, Ayres TM, Skubatch M, Gray CL, Rothe M, Ashkenazi A (1997) Herpesvirus entry
mediator, a member of the tumor necrosis factor receptor (TNFR) family, interacts with mem-
bers of the TNFR-associated factor family and activates the transcription factors NF-κB and
AP-1. J Biol Chem 272(22):14029–14032
Matsumori A, Yamada T, Suzuki H, Matoba Y, Sasa YS (1994) Increased circulating cytokines in
patients with myocarditis and myocardiopathy. Br Heart J 72:561–566
McColl SR, Roberge CJ, Larochelle B, Gosselin J (1997) EBV induces the production and release
of IL-8 and macrophage inflammatory protein-1 alpha in human neutrophils. J Immunol
159(12):6164–6168
McInnes IB (2017) Cytokines. In: Kelley and Firestein’s textbook of rheumatology. Elsevier,
Philadelphia, pp 396–407
96 P. Sanapala and S. Pola
McLoughlin RM, Hurst SM, Nowell MA, Harris DA, Horiuchi S, Morgan LW, Wilkinson TS,
Yamamoto N, Topley N, Jones SA (2004) Differential regulation of neutrophil-activating che-
mokines by IL-6 and its soluble receptor isoforms. J Immunol 172(9):5676–5683
Melville MW, Tan SL, Wambach M, Song J, Morimoto R, Katze MG (1999) The cellular inhibitor
of the PKR protein kinase, P58(IPK), is an influenza virus-activated co-chaperone that modu-
lates heat shock protein 70 activity. J Biol Chem 274(6):3797–3803
Modur V, Li Y, Zimmerman GA, Prescott SM, McIntyre TM (1997) Retrograde inflammatory
signaling from neutrophils to endothelial cells by soluble interleukin-6 receptor alpha. J Clin
Invest 100(11):2752–2756
Mogensen TH, Paludan SR (2001) Molecular pathways in virus-induced cytokine production.
Microbiol Mol Biol Rev 65(1):131–150
Nakagomi H, Dolcetti R, Bejarano MT, Pisa P, Kiessling R, Masucci MG (1994) The Epstein-Barr
virus latent membrane protein-1 (LMP1) induces interleukin-10 production in Burkitt lym-
phoma lines. Int J Cancer 57(2):240–244
Nathan CF (1987) Secretory products of macrophages. J Clin Invest 79(2):319–326
Natoli G, Avantaggiati ML, Chirillo P, Puri P, Ianni A, Balsano C, Levrero M (1994) Ras-and Raf-
dependent activation of c-jun transcriptional activity by the hepatitis B virus transactivatorp
X. Oncogene 9(10):2837–2843
Nicod LP (1993) Cytokines overview. Thorax 48(6):660–667
O’Shea JJ, Gadina M, Siegel RM (2019) Cytokines and cytokine receptors. In: Clinical
Immunology. Elsevier, London, pp 127–155
Oberlin E, Amara A, Bessia C, Virelizier JL, Arenzana-Seisdedos F, Schwartz O, Heard JM,
Clark-Lewis I, Legler DF, Loetscher M, Baggiolini M (1996) The CXC chemokine SDF-1
is the ligand for LESTR/fusin and prevents infection by T-cell-line-adapted HIV-1. Nature
382(6594):833–835
Oquendo J, Karray S, Galanaud P, Petit MA (1997) Effect of hepatitis B virus on tumour necrosis
factor (TNF α) gene expression in human THP-1 monocytic and Namalwa B cell lines. Res
Immunol 148(6):399–409
Orange JS, Biron CA (1996) An absolute and restricted requirement for IL-12 in natural killer cell
IFN-gamma production and antiviral defense. Studies of natural killer and T cell responses in
contrasting viral infections. J Immunol 156(3):1138–1142
Pahl HL, Baeuerle PA (1995) Expression of influenza virus hemagglutinin activates transcription
factor NF-κB. J Virol 69(3):1480–1484
Pahl HL, Baeuerle PA (1997) The ER overload response: activation of NF-κB. Trends Biochem
Sci 22(2):63–67
Paludan SR, Ellermann-Eriksen S, Mogensen SC (1998) NF-kappaB activation is responsible for
the synergistic effect of herpes simplex virus type 2 infection on interferon-gamma-induced
nitric oxide production in macrophages. J Gen Virol 79(11):2785–2793
Paludan SR, Ellermann-Eriksen S, Malmgaard L, Mogensen SC (2000) Inhibition of NO produc-
tion in macrophages by IL-13 is counteracted by HSV infection through TNF-α-induced acti-
vation of NF-κB. Eur Cytokine Netw 11(2):275–282
Paul NL, Lenardo MJ, Novak KD, Sarr T, Tang WL, Ruddle NH (1990) Lymphotoxin activation
by human T-cell leukemia virus type I-infected cell lines: role for NF-kappaB. J Virol 64(11):
5412–5419
Penna A, Del Prete G, Cavalli A, Bertoletti A, D’Elios MM, Sorrentino R, D’Amato M, Boni C, Pilli
M, Fiaccadori F, Ferrari C (1997) Predominant T-helper 1 cytokine profile of hepatitis B virus
nucleocapsid-specific T cells in acute self-limited hepatitis B. Hepatology 25(4):1022–1027
Peterson PK, Gekker G, Chao C, Hu SX, Edelman C, Balfour HH, Verhoef J (1992) Human
cytomegalovirus-stimulated peripheral blood mononuclear cells induce HIV-1 replication via a
tumor necrosis factor-alpha-mediated mechanism. J Clin Invest 89(2):574–580
Poli G, Bressler P, Kinter A, Duh E, Timmer WC, Rabson A, Justement JS, Stanley S, Fauci AS
(1990) Interleukin 6 induces human immunodeficiency virus expression in infected mono-
cytic cells alone and in synergy with tumor necrosis factor alpha by transcriptional and post-
transcriptional mechanisms. J Exp Med 172(1):151–158
5 Role of Cytokines in Infectious Viral Disease 97
Popik W, Pitha PM (1996) Binding of human immunodeficiency virus type 1 to CD4 induces
association of Lck and Raf-1 and activates Raf-1 by a Ras-independent pathway. Mol Cell Biol
16(11):6532–6541
Ramshaw IA, Ramsay AJ, Karupiah G, Rolph MS, Mahalingam S, Ruby JC (1997) Cytokines and
immunity to viral infections. Immunol Rev 159(1):119–135
Rinaldo JC, Armstrong JA, Kingsley LA, Zhou S, Ho M (1990) Relation of alpha and gamma
interferon levels to development of AIDS in homosexual men. J Exp Pathol 5(3):127–132
Roberge CJ, Poubelle PE, Beaulieu AD, Heitz D, Gosselin J (1996) The IL-1 and IL-1 recep-
tor antagonist (IL-1Ra) response of human neutrophils to EBV stimulation. Preponderance of
IL-Ra detection. J Immunol 156(12):4884–4891
Rosenberg ZF, Fauci AS (1990) Immunopathogenic mechanisms of HIV infection: cytokine
induction of HIV expression. Immunol Today 11:176–180
Rösler A, Pohl M, Braune HJ, Oertel WH, Gemsa D, Sprenger H (1998) Time course of chemo-
kines in the cerebrospinal fluid and serum during herpes simplex type 1 encephalitis. J Neurol
Sci 157(1):82–89
Ruocco MR, Chen X, Ambrosino C, Dragonetti E, Liu W, Mallardo M, De Falco G, Palmieri C,
Franzoso G, Quinto I, Venuta S (1996) Regulation of HIV-1 long terminal repeats by interaction
of C/EBP (NF-IL6) and NF-κB/Rel transcription factors. J Biol Chem 271(37):22479–22486
Schafer SL, Vlach J, Pitha PM (1996) Cooperation between herpes simplex virus type 1-encoded
ICP0 and Tat to support transcription of human immunodeficiency virus type 1 long terminal
repeat in vivo can occur in the absence of the TAR binding site. J Virol 70(10):6937–6946
Scholtissek C (1991) Synthesis and function of influenza A virus glycoproteins. Behring Inst Mitt
89:46–53
Schultz JC, Hilliard AA, Cooper LT Jr, Rihal CS (2009) Diagnosis and treatment of viral myocar-
ditis. Mayo Clin Proc 84(11):1001–1009
Setsuda J, Teruya-Feldstein J, Harris NL, Ferry JA, Sorbara L, Gupta G, Jaffe ES, Tosato G (1999)
Interleukin-18, interferon-γ, IP-10, and Mig expression in Epstein-Barr virus-induced infectious
mononucleosis and posttransplant lymphoproliferative disease. Am J Pathol 155(1):257–265
Skoner DP, Gentile DA, Patel A, Doyle WJ (1999) Evidence for cytokine mediation of disease
expression in adults experimentally infected with influenza A virus. J Infect Dis 180(1):10–14
Stadnyk AW (1994) Cytokine production by epithelial cells. FASEB J 8(13):1041–1047
Su F, Schneider RJ (1996) Hepatitis B virus HBx protein activates transcription factor NF-kappa
B by acting on multiple cytoplasmic inhibitors of rel-related proteins. J Virol 70(7):4558–4566
Sugano N, Chen W, Roberts ML, Cooper NR (1997) Epstein-Barr virus binding to CD21 activates
the initial viral promoter via NF-κB induction. J Exp Med 186(5):731–737
Tanner JE, Alfieri C, Chatila TA, Diaz-Mitoma F (1999) Induction of interleukin-6 after stimula-
tion of human B-cell CD21 by Epstein-Barr virus glycoproteins gp350 and gp220. J Virol
70(1):570–575
Thomas J, Kanangat S, Rouse BT (1998) Herpes simplex virus replication-induced expression of
chemokines and proinflammatory cytokines in the eye: implications in herpetic stromal kerati-
tis. J Interf Cytokine Res 18(9):681–690
Toso JF, Ferbas JJ, Rappocciolo G, Armstrong JA, Ho M, Rinaldo JC (1990) Stimulation of IFN-
alpha production in HLA-DR+, light density peripheral blood mononuclear cells by human
immunodeficiency virus. J Exp Pathol 5(3):123–125
Vilcek J (2003) Thecytokines: anoverview. In: Lotte M, Thompson AW (eds) The cytokine hand-
book, vol 1, 4th edn. Academic Press, Amsterdam, pp 3–18
Vingerhoets J, Michielsen P, Vanham G, Bosmans E, Paulij W, Ramon A, Pelckmans P, Kestens L,
Leroux-Roels G (1998) HBV-specific lymphoproliferative and cytokine responses in patients
with chronic hepatitis B. J Hepatol 28(1):8–16
Virelizier JL (1999) Blocking HIV co-receptors by chemokines. Dev Biol Stand 97:105–109
Vlach J, Garcia A, Jacqué JM, Rodriguez MS, Michelson S, Virelizier JL (1995) Induction of
Sp1 phosphorylation and NF-κB-independent HIV promoter domain activity in T lymphocytes
stimulated by okadaic acid. Virology 208(2):753–761
98 P. Sanapala and S. Pola
Wang HD, Yuh CH, Dang CV, Johnson DL (1995a) The hepatitis B virus X protein increases the
cellular level of TATA-binding protein, which mediates transactivation of RNA polymerase III
genes. Mol Cell Biol 15(12):6720–6728
Wang L, Mukherjee S, Jia F, Narayan O, Zhao LJ (1995b) Interaction of virion protein Vpr of
human immunodeficiency virus type 1 with cellular transcription factor Sp1 and trans-
activation of viral long terminal repeat. J Biol Chem 270(43):25564–25569
Whicher JT, Evans SW (1990) Cytokines in disease. Clin Chem 36(7):1269–1281
Whitmarsh AJ, Davis RJ (1996) Transcription factor AP-1 regulation by mitogen-activated protein
kinase signal transduction pathways. J Mol Med 74(10):589–607
Xiao G, Harhaj EW, Sun SC (2000) Domain-specific interaction with the IκB kinase (IKK) reg-
ulatory subunit IKKγ is an essential step in Tax-mediated activation of IKK. J Biol Chem
275(44):34060–34067
Yurochko AD, Huang ES (1999) Human cytomegalovirus binding to human monocytes induces
immunoregulatory gene expression. J Immunol 162(8):4806–4816
Zhang JM, An J (2007) Cytokines, inflammation and pain. Int Anesthesiol Clin 45(2):27
B Cells and Their Role in Combating Viral
Diseases 6
Devanabanda Mallaiah and Pallaval Veera Bramhachari
Abstract
Humoral immunity mediated by B cells plays an important role in combating
different types of viral diseases. Two types of B cells (B-1 and B-2) originate in
the primary lymphoid organ called bone marrow from hematopoietic stem cells
(HSCs). Both B-1and B-2 cells develop separately through sequential steps
(pro-B cells—pre-B cells—immature B cells) in bone marrow from distinct
common lymphoid progenitors before their release as immature B cells into cir-
culation. In the serous cavities, the immature B-1 cells differentiate into mature
B-1 cells through transitional B cells. The immature B-2 cells differentiate into
transitional B cells, which mature finally into marginal zone (MZ) and follicular
(FO) B cells in the secondary lymphoid organs. The B-1 cells produce poly-
specific natural antibodies (antibodies produced before infection), which provide
first line of defense. The B-1 cells mainly defend mucosal and blood-borne
pathogens in a T-cell independent manner. The MZ B cells produce immune
response against blood-borne pathogens and undergo both T-independent and
T-dependent activation. In addition, both B1 and MZ B cells behave like innate
immune cells by expressing toll-like receptors (TLR) and produce immune
response without or with their membrane-bound poly-reactive B-cell receptors
(BCR). Finally, FO B cells are the conventional B cells of adaptive immunity and
primarily responsible for T-dependent immune response by their membrane-
bound mono-reactive BCR. The antigen-activated B-2 cells differentiate into
antibodies secreting plasma cells and memory B cells. Along with natural anti-
bodies, the non-neutralizing, neutralizing, and broadly neutralizing specific
D. Mallaiah (*)
Department of Biotechnology and Bioinformatics, Yogi Vemana University,
Kadapa, Andhra Pradesh, India
P. V. Bramhachari
Department of Biotechnology, Krishna University,
Machilipatnam, Andhra Pradesh, India
Keywords
B cells · Humoral immunity · Antibodies · Viral diseases
6.1 Introduction
Serum therapy by Von Behring and Kitasato provided the first evidence that humoral
response played an important role in the host resistance against infectious agents
such as diphtheria and tetanus toxins. They explained that serum therapy gives dis-
ease resistance due to the presence of antibodies in the serum. Paul Ehrlich demon-
strated the basis for serum therapy and established the field of humoral immunity.
Today, the importance of antibodies as immunotherapeutic is well characterized and
is used to treat a wide range of infectious diseases. Antibodies are main components
in many effective vaccines and have been used to prevent many human diseases
caused by viruses (Yamada 2011; Burton 2002).
The success of immune system in combating different types of viruses depends
on both non-specific innate immune system and specific-adaptive immune system.
Humoral immunity is a branch of adaptive immunity mediated by antibodies
secreted from B cells, which plays an important role in combating viruses along
with cell-mediated immunity (Bonilla and Oettgen 2010). Humoral immune
response to antigens is categorized as primary and secondary response. In the pri-
mary response, T helper cells are of primary importance whereas in the secondary
immune response, the specific-neutralizing antibodies play a primary role against
reinfection and T cells are of secondary importance. The primary immune response
can take 1–2 weeks and IgM is the predominant type of antibody elicited. The sec-
ondary immune response is more rapid than primary immune response and pre-
dominant type of antibody formed is IgG (Lefevre et al. 2009).
The T helper cells, typically follicular T helper cells (TFH cells), were activated
by the same antigen and cooperate with antigen-activated mature B cells to differ-
entiate into plasma cells and memory B cells. The activated B cells remain in the
margins of T-cell zone and differentiate into short-lived plasma cells. The activated
B cells which initiate germinal centers differentiate into long-lived plasma cells and
memory B cells. The high-affinity and antigen-specific antibodies are secreted and
maintained by both short-lived and long-lived plasma cells. The long-lived plasma
cells migrate to survival niches in the bone marrow and persist for several years
(Gourley et al. 2004). The protective natural antibodies are produced before the
foreign antigen exposure or pathogens by B-1 cells and marginal zone B cells in a
T-cell independent manner. About 80% of all circulating natural antibodies are IgM
type and remaining are IgG and IgA (Palma et al. 2018). B cells provide several
lines of protection against viruses. In the first line, natural antibodies provide
6 B Cells and Their Role in Combating Viral Diseases 101
defense against initial viral infections. The long-lived plasma cells generate high
amounts of neutralizing IgG and provide second line of defense, and reactive long-
lived memory B cells provide third line of defense against viruses (Dörner and
Radbruch 2007).
Natural antibodies are encoded by germ line variable genes and not shaped by
post-recombination processes such as somatic hypermutation and class switching.
Natural antibodies recruit antigens into the spleen to prevent infection of vital
organs and also to induce early neutralizing antibodies without T-cell help. Natural
antibodies activate complement to enhance B- and T-cell-specific immune
responses. Therefore, natural antibodies are considered an important link between
innate and adaptive immunity (Matter and Ochsenbein 2008). The natural antibod-
ies are poly-reactive that they can bind to different structurally unrelated antigens
and also auto-reactive that they can bind to self-antigens. Along with natural anti-
bodies, poly-reactive antigen-binding B cells provide protection against multiple
pathogens. In addition to poly-reactive antibodies, the specific neutralizing and
cross-reactive antibodies also provide effective antiviral immune response (Warter
et al. 2012).
The highly specific antibodies protect from viral infection by both neutralizing
and Fc-mediated effector mechanisms. Neutralizing antibodies inhibit the viruses
by binding through their antigen-binding sites or Fab region and also through their
constant or Fc region. The non-neutralizing antibodies bind to many epitopes
including neutralizing antibody targets and induce the destruction of microbe or
infected host cells by innate immune system (Hua and Ackerman 2017). Recently,
novel broadly neutralizing antibodies were characterized, which are able to neutral-
ize diverse isolates of viruses (Sok and Burton 2018). This chapter describes about
different types of B cells, antibodies, and their role in combating different types of
viruses (HIV, influenza, and hepatitis).
6.2 B Cells
The B lymphocytes are subset of lymphocytes, which express diverse specific sur-
face immunoglobulins and constitute 5–15% of total circulating lymphoid cells.
The antigen-independent development of B cells starts from the fetal liver and bone
marrow of an adult, where the hematopoietic stem cells differentiate into pro-B
cells, which in turn differentiate pre-B cells to immature B cells. Along with the
development of phenotypically distinct precursor cells, rearrangements of VDJ
genes occur parallelly, which contribute to formation of diverse specific immature
B cells with membrane-bound IgM. Immature B cells differentiate into mature B
cells with membrane-bound IgM and IgD as its BCR in the secondary lymphoid
organs. Two types of B lymphocytes (B-1 and B-2) have been identified. Mature B
lymphocytes are activated by different types of antigens. There are three types of
antigens activating the B lymphocytes. (1) T-independent antigen type 1, for exam-
ple, LPS (lipopolysaccharide), which is polyclonal activator of both mature and
immature B cells. LPS activates the B cells through TLR. (2) T-independent antigen
102 D. Mallaiah and P. V. Bramhachari
Fig. 6.1 Schematic diagram of B-cell development and different humoral effector immune cells
non-covalently attached with BCR and their cytoplasmic domains certainly con-
tains conserved motifs for tyrosine phosphorylation and Src family kinase dock-
ing respectively (LeBien and Tedder 2008). In addition to production of antibodies,
B cells also actively participate in cellular immune response mediated by T cells.
The B cells directly modulate effector, memory, and regulatory T-cell functions via
antigen-specific but antibody-independent mechanisms. B cells modulate T cells by
antigen presentation, by providing co-stimulation, and by secreting cytokines. B
cells are functionally subdivided into two types: (1) Be-1 cells and (2) Be-2 cells. In
the presence of TH1-type cytokines, the Be-1 cells do not secrete significant
amounts of IL-4, IL-13, or IL-2 but secrete IFN-γ and IL-12 including IL-10, TNF-
α, and IL-6. In the presence of TH2-type cytokines, Be-2 cells do not secrete signifi-
cant amounts of IFN-γ and IL-12 but secrete IL-4, IL-13, or IL-2 including IL-10,
TNF-α, and IL-6. The regulatory B cells also called B-10 cells produce IL-10 cyto-
kines that suppress the CD4+ T-cell responses. In addition to suppression, cytokines
producing B cells enhance the T-cell-mediated immune responses (Lund and
Randall 2010).
6.3 Antibodies
The B lymphocytes differentiate finally into plasma cells, which are the source for
antibodies. The two competing natural selection and clonal selection theories for
antibody formation were proposed by Jerne and Burnet, respectively, but finally,
clonal selection theory was supported by experimental evidence (Burnet 1976).
Antibodies are chemically glycoprotein molecules. They are also called immu-
noglobulins and are present on the surface of B cells as BCR or free molecules in
blood, plasma, and extracellular fluids. The fluids formerly were called humors and
are part of humoral immune response. They have two principal functions in
humoral defense. The first function is that it recognizes and binds to antigenic
104 D. Mallaiah and P. V. Bramhachari
Fig. 6.3 Antiviral mechanism of antibodies. (a) Neutralization, (b) antibody-bound virus or
infected cell interacts with Fc receptor on innate effector cell, (c) complement component-bound
antibody initiates MAC formation on infected cell or interacts with complement-component recep-
tor on innate effector cell, (d) infected cell bound by antibody interacts with dendritic cell to
release type 1 interferon which in turn stimulates NK cells and antiviral proteins in infected cells,
(e) antigen presentation of viral peptides to T cells. MAC membrane attack complex, APC antigen-
presenting cell, MHC major histocompatibility complex, TCR T-cell receptor
the replication, genetic material expression, and release of pathogen. (5) Inhibition
at later steps—antibodies inhibit the liberation of virus or budding (Forthal 2014).
Viruses are intracellular pathogens and need host cells machinery for their survival.
After synthesizing many viral copies, viruses burst out and reinfect healthy new
host cells. These viruses directly kill the host cells and are called acutely cytopathic
viruses (polioviruses, rabies, and small pox viruses). Some viruses do not kill the
host cells and they are called non-cytopathic viruses (hepatitis B and C, herpes
virus, herpes simplex virus 1 and 2, cytomegalovirus, and Epstein-Barr viruses).
The role of immune system in antiviral defense is both protective and pathogenic,
where it eliminates the viruses by effective immune response and in addition also
causes more damage to host cells than viruses themselves. With regard to effective
immunity, the viruses have developed an array of strategies to escape elimination by
immune system through co-evolution (Dörner and Radbruch 2007).
Viruses are chemically nucleoprotein particles with a size of ~20–200 nm. Due
to their small size, viruses move freely in the lymphatic system and interact with B
cells in secondary lymphoid organs. The highly repetitive structures of viral parti-
cles allow the cross-linking of BCRs, which is an initial step in the activation of B
cells. Further, the repetitive viral structures also bind with natural antibodies, fix
complement, and also carry TLR ligands (DNA or RNA) for TLR 7/8 or 9 to acti-
vate B cells directly (Zabel et al. 2013).
Influenza or flu viruses cause respiratory diseases and continuously threaten human
health. Genome high mutational rates of influenza viruses cause emergences of new
strains by genetic drifts. Influenza affects 5–30% of global population and causes
hospitalization and death of many people. They are enveloped viruses and contain
single-stranded, segmented, and negative sense 7–8 RNA strands. Influenza viruses
are categorized into four types: A, B, C, and D. Influenza A virus is again classified
based on the antigenic properties of two surface viral glycol proteins such as hem-
agglutinin (HA) and neuraminidase (NA) with 18 (H1-H18) and 11 (N1-N11) anti-
genic subtypes, respectively. Influenza A (H1N1 and H3N2) virus causes seasonal
influenza epidemics in humans along with other members of influenza viruses such
as influenza B virus (Sautto et al. 2018).
Two important hypotheses such as original antigenic sin (OAS) and immune
imprinting or antigenic seniority explain how humoral immunity gets affected by
influenza viruses. Both are dependent on humoral immune memory response rather
than on de novo humoral immune response to drifted or altered influenza viruses
(Guthmiller and Wilson 2018). The original antigenic sin concept refers that the first
exposure of influenza variant in early life dictates lifelong immunity to all variants
of influenza viruses in subsequent encounters. The immune memory produced by
the first influenza variant influences the immune response to subsequently expose
influenza distinct variants, but how this sequential exposure shapes the immune
response remains obscure. The antigenic seniority concept better explains the hier-
archical nature of immune response to previously exposed variants of influenza
108 D. Mallaiah and P. V. Bramhachari
virus. According to the antigenic seniority concept, the first exposed influenza vari-
ant in childhood takes the senior antigenic position in immune repertoire and subse-
quent exposed strains take the junior positions. The immune response to the first
exposure is larger than the responses to subsequent exposures. Understanding of
how previous exposure shapes the antibody responses to vaccination and infection
is critical for the development of universal influenza vaccine (Henry et al. 2018).
Neutralizing antibodies produced against HA head region of influenza are strain-
specific and bind to highly variable regions. Antibodies neutralize more strains of
influenza and are called broadly neutralizing or cross-reactive antibodies. The
broadly neutralizing antibodies produced against influenza virus mainly target the
conserved regions of HA stem domain. The existence of non-neutralizing antibod-
ies also clears the influenza virus infection by exploiting non-neutralizing effector
mechanisms (Sicca et al. 2018).
The natural antibodies against influenza virus are produced by B-1 cells and
therefore reported to suggest that innate and acquired humoral immunity comes
from separate effector arms of immune system (Baumgarth et al. 1999).
The HVR1 antibodies are type-specific but some evidence shows that anti-
HVR1 antibodies are cross-reactive. Some neutralizing antibodies against E1 and
E2 envelope proteins show cross-neutralizing potential (Drummer 2014). However,
there is strong evidence on the production of broadly neutralizing antibodies
against HCV infections (Kinchen et al. 2018). The HCV virus also has immuno-
logic regions for virus escape or non-neutralizing antibodies in the envelope glyco-
proteins (Fuerst et al. 2018). Similar to HIV virus, the humoral immune response
also evolves with time in HCV. The evolution of Ab specificity (non-neutralizing—
autologous neutralizing—heterologous neutralizing—broadly neutralizing) during
the course of infection produces increased breadth in the neutralizing activity
(Murira et al. 2016). There are also reports on natural antibodies which recognize
the linear epitopes on hepatitis C envelope glycoprotein E2 to confer additional
neutralization (Tarr et al. 2012).
B cells are the main players of humoral immunity against viruses. B cells also act as
antigen-presenting cells, contributing to cell-mediated immunity. Both B-1 and B-2
cells secrete natural and highly specific antibodies for the prevention and control of
viral infections, respectively. B-2 cells capture the viral antigens through BCR and
differentiate into memory B cells and plasma cells, which secrete different types of
specific adaptive antibodies. Recently, many studies reported the generation of
broadly neutralizing antibodies, which can neutralize different strains of viruses.
Understanding the production of antibodies and their antiviral mechanisms will pro-
vide new ways to develop novel, efficient prophylactic and therapeutic vaccines.
Acknowledgements The authors are grateful to YVU Kadapa and Krishna University,
Machilipatnam, for the support extended.
Conflict of Interest The authors declare that they have no competing interests.
References
Baum LL (2010) Role of humoral immunity in host defense against HIV. Curr HIV/AIDS Rep
7(1):11–18
Baumgarth N, Herman OC, Jager GC, Brown L, Herzenberg LA, Herzenberg LA (1999) Innate
and acquired humoral immunities to influenza virus are mediated by distinct arms of the
immune system. Proc Natl Acad Sci U S A 96(5):2250–2255
Bonilla FA, Oettgen HC (2010) Adaptive immunity. J Allergy Clin Immunol 125(2 Suppl 2):
S33–S40
Burnet FM (1976) A modification of Jerne’s theory of antibody production using the concept of
clonal selection. CA Cancer J Clin 26(2):119–121
Burton DR (2002) Antibodies, viruses and vaccines. Nat Rev Immunol 2(9):706–713
110 D. Mallaiah and P. V. Bramhachari
Chan CE, Chan AH, Hanson BJ, Ooi EE (2009) The use of antibodies in the treatment of infectious
diseases. Singap Med J 50(7):663–672; quiz 673
Dörner T, Radbruch A (2007) Antibodies and B cell memory in viral immunity. Immunity
27(3):384–392
Drummer HE (2014) Challenges to the development of vaccines to hepatitis C virus that elicit
neutralizing antibodies. Front Microbiol 5:329
Forthal DN (2014) Functions of antibodies. Microbiol Spectr 2(4):1–17
Fuerst TR, Pierce BG, Keck ZY, Foung SKH (2018) Designing a B cell-based vaccine against a
highly variable hepatitis C virus. Front Microbiol 8:2692
Gourley TS, Wherry EJ, Masopust D, Ahmed R (2004) Generation and maintenance of immuno-
logical memory. Semin Immunol 16(5):323–333
Guthmiller JJ, Wilson PC (2018) Harnessing immune history to combat influenza viruses. Curr
Opin Immunol 53:187–195
Henry C, Palm AE, Krammer F, Wilson PC (2018) From original antigenic sin to the universal
influenza virus vaccine. Trends Immunol 39(1):70–79
Hey A (2015) History and practice: antibodies in infectious diseases. Microbiol Spectr
3(2):AID-0026-2014
Hoffman W, Lakkis FG, Chalasani G (2016) B cells, antibodies, and more. Clin J Am Soc Nephrol
11(1):137–154
Hua CK, Ackerman ME (2017) Increasing the clinical potential and applications of anti-HIV anti-
bodies. Front Immunol 8:1655
Irshad M, Khushboo I, Singh S, Singh S (2008) Hepatitis C virus (HCV): a review of immunologi-
cal aspects. Int Rev Immunol 27(6):497–517
Kinchen VJ, Zahid MN, Flyak AI, Soliman MG, Learn GH, Wang S, Davidson E, Doranz BJ,
Ray SC, Cox AL, Crowe JE Jr, Bjorkman PJ, Shaw GM, Bailey JR (2018) Broadly neutral-
izing antibody mediated clearance of human Hepatitis C virus infection. Cell Host Microbe
24(5):717–730.e5
Lapa D, Garbuglia AR, Capobianchi MR, Del Porto P, Hepatitis C (2019) Virus genetic variability,
human immune response, and genome polymorphisms: which is the interplay? Cells 8(4):E305
LeBien TW, Tedder TF (2008) B lymphocytes: how they develop and function. Blood
112(5):1570–1580
Lefevre EA, Carr BV, Prentice H, Charleston B (2009) A quantitative assessment of primary and
secondary immune responses in cattle using a B cell ELISPOT assay. Vet Res 40(1):3
Lopalco L (2010) Natural anti-CCR5 antibodies in HIV-infection and exposure. J Transl Med
9(Suppl 1):S4
Lund FE, Randall TD (2010) Effector and regulatory B cells: modulators of CD4+ T cell immu-
nity. Nat Rev Immunol 10(4):236–247
Maddaly R, Pai G, Balaji S, Sivaramakrishnan P, Srinivasan L, Sunder SS, Paul SF (2010)
Receptors and signaling mechanisms for B-lymphocyte activation, proliferation and differ-
entiation--insights from both in vivo and in vitro approaches. FEBS Lett 584(24):4883–4894
Matter MS, Ochsenbein AF (2008) Natural antibodies target virus-antibody complexes to orga-
nized lymphoid tissue. Autoimmun Rev 7(6):480–486
Mayr LM, Su B, Moog C (2017) Non-neutralizing antibodies directed against HIV and their func-
tions. Front Immunol 8:1590
Montecino-Rodriguez E, Dorshkind K (2012) B-1 B cell development in the fetus and adult.
Immunity 36(1):13–21
Mouquet H (2014) Antibody B cell responses in HIV-1 infection. Trends Immunol 35(11):549–561
Murira A, Lapierre P, Lamarre A (2016) Evolution of the humoral response during HCV infection:
theories on the origin of broadly neutralizing antibodies and implications for vaccine design.
Adv Immunol 129:55–107
Palma J, Tokarz-Deptuła B, Deptuła J, Deptuła W (2018) Natural antibodies – facts known and
unknown. Cent Eur J Immunol 43(4):466–475
Phogat S, Wyatt RT, Karlsson Hedestam GB (2007) Inhibition of HIV-1 entry by antibodies: poten-
tial viral and cellular targets. J Intern Med 262(1):26–43
6 B Cells and Their Role in Combating Viral Diseases 111
Sautto GA, Kirchenbaum GA, Ross TM (2018) Towards a universal influenza vaccine: different
approaches for one goal. Virol J 15(1):17
Sicca F, Neppelenbroek S, Huckriede A (2018) Effector mechanisms of influenza-specific antibod-
ies: neutralization and beyond. Expert Rev Vaccines 17(9):785–795
Sok D, Burton DR (2018) Recent progress in broadly neutralizing antibodies to HIV. Nat Immunol
19(11):1179–1188
Suzuki K, Maruya M, Kawamoto S, Fagarasan S (2010) Roles of B-1 and B-2 cells in innate and
acquired IgA-mediated immunity. Immunol Rev 237(1):180–190
Tarr AW, Urbanowicz RA, Jayaraj D, Brown RJ, McKeating JA, Irving WL, Ball JK (2012)
Naturally occurring antibodies that recognize linear epitopes in the amino terminus of the hepa-
titis C virus E2 protein confer noninterfering, additive neutralization. J Virol 86(5):2739–2749
Ward S (2001) Natural anti-HIV antibodies. Trends Immunol 22(10):544
Warter L, Appanna R, Fink K (2012) Human poly- and cross-reactive anti-viral antibodies and
their impact on protection and pathology. Immunol Res 53(1–3):148–161
Webster DP, Klenerman P, Dusheiko GM (2015) Hepatitis C. Lancet 385(9973):1124–1135
Yamada T (2011) Therapeutic monoclonal antibodies. Keio J Med 60(2):37–46
Yuseff MI, Pierobon P, Reversat A, Lennon-Duménil AM (2013) How B cells capture, process and
present antigens: a crucial role for cell polarity. Nat Rev Immunol 13(7):475–486
Zabel F, Kündig TM, Bachmann MF (2013) Virus-induced humoral immunity: on how B cell
responses are initiated. Curr Opin Virol 3(3):357–362
Zolla-Pazner S (2016) Non-neutralizing antibody functions for protection and control HIV in
humans and SIV and SHIV in non-human primates. AIDS 30(16):2551–2553
Part II
Role of Adaptive Immunity in Combating Viral
Diseases
Significant Role of Cellular Activation
in Viral Infections 7
Bojjibabu Chidipi, Samuel Ignatious Bolleddu,
Ganugula Mohana Sheela, and Alavala Matta Reddy
Abstract
Immune response during viral infection is the result of pathogen recognition and
antibody production by the memory cells residing in the host target tissue. These
cells by origin are differentiated from the cytotoxic CD8+ T cells and play a
major role in developing adaptive immunity. Early activation of these T cells by
ribosomal binding proteins during various cell death pathways can induce a criti-
cal autoimmune response suitable to develop enhanced strategic immune thera-
pies for many chronic viral infections. In addition, there are various genes which
get expressed in good amount in the process of T-cell activation during infections
like Epstein-Barr virus. Focusing on these changes can help in determining the
pathogenic process and orientation of the viral infection. Since we can only con-
trol but not cure most of the viral infections using present medications, following
these novel strategies can help in focusing on the host cell death pathways,
thereby developing the drugs that can give complete immunity against virus
invasion by facilitated viral cell elimination from the humoral environment.
Keywords
Cytotoxic CD8+ T cells · Epstein-Barr virus · Humoral environment · Cell death
pathways · Memory cells
B. Chidipi · S. I. Bolleddu
Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
G. M. Sheela
Department of Biotechnology, Krishna University,
Machilipatnam, Andhra Pradesh, India
A. Matta Reddy (*)
School of Life and Health Sciences, Adikavi Nannaya University,
Rajamahendravaram, Andhra Pradesh, India
7.1 Introduction
The main role of the immune system is to identify the presence of any foreign pro-
tein complexes or antigens and initiate an immune response to eliminate them with-
out harming the metabolism of the host by implementing the pathogen-specific
defense response. Immune response during viral infection is the result of pathogen
recognition and antibody production by the memory cells residing in the host target
tissue. B lymphocytes produce the antibodies during the humoral immune response
while T cells initiate cell-mediated immunity (Finotti et al. 2018).
During viral infections, the cellular activation of various lymphocytes leads to
degranulation of various inflammatory mediators like histamines, TNFα, and vari-
ous amino acids within the first few seconds. The host inborn resistant reaction is a
profoundly strong boundary of barrier containing numerous intrinsic safe cells, for
example, dendritic cells (DCs), macrophages, and common executioner (NK) cells.
These cells are fit for perceiving a broad scope of viral items through a complex and
covering atomic system of pattern recognition receptors (PRRs), including toll-like
receptors (TLRs) just as cytosolic sensors, for example, the helicases, retinoic acid-
inducible gene 1 (RIG-I) and MDA5. Actuation of intrinsic cells through these
PRRs advances fast popular detecting and the arrival of critical antiviral cytokines
with the possibility of quickly controlling viral replication and driving further natu-
ral and versatile invulnerable reactions. Specifically, the sort I interferon (IFN-I) is
a pleiotropic cytokine family comprising 13 IFNα isoforms and one IFNβ isoform
that move through a common pervasively communicated receptor (IFNαβR).
Eicosanoids like leukotrienes and prostaglandins are released as a secondary
mediator after few minutes. In a prolonged immune response, cytokines and chemo-
kines along with growth factors are released (Bull and Plummer 2014; Frossi et al.
2018; Yu et al. 2016; Brown et al. 2018; Gieseck et al. 2018).
The viral reproduction process in the host cells after the viral invasion starts with
the synthesis of new viral proteins and nucleic acids intracellularly. The new viral
particles or virions are then self-assembled by the aggregation of the macromole-
cules. These virions are seen in the blood circulation invading the healthy cells after
the rupture of the infected host cells. The envelope or capsids of the virus particles
offer protection and platform for inducing an immune response. Motioning through
IFNαβR prompts both autocrine and paracrine actuation, bringing about phosphory-
lation of flag transducer and activator of interpretation 1 and 2 (STAT1 and STAT2).
This actuates several IFN-I-animated qualities (ISGs) that advance an antiviral state
and initiate various safe cells (Brown et al. 2018). Despite this complex inborn reac-
tion, numerous infections, including those that accomplish interminable disease,
can stifle and additionally avoid these early natural host reactions to support their
replication and transmission. In this segment, we centered on IFN-I to show the
dynamic changes that the natural resistant framework experiences and the critical
and expansive results that such changes can have amid perpetual viral diseases
(Ning et al. 2018).
This new wave of viral invasion directly stimulates glycoprotein inducing cells
for the production of the antiviral proteins called interferons. Vital cells of the
7 Significant Role of Cellular Activation in Viral Infections 117
immune system like leucocytes, fibroblasts, and natural killer cells play a vital role
in the production of interferons and lysosomes that lyse a wide variety of virus par-
ticles, thereby recognizing the antigen and developing a suitable antibody.
Within days after HIV discovery in plasma from tainted patients, and inside hours
after contamination with LCMV Cl13 in mice, raised dimensions of fundamental
IFN-I are distinguished. Moreover, upgraded ISG articulation is visible in tissues as
well as T cells after LCMV Cl13, HCV, and SIV diseases (Fig. 7.1). Interestingly,
qualities with intrinsic insusceptible capacity are imperceptible right on time after
in vivo HBV contamination in chimpanzees (Haas and Trumpp 2018).
Early activation of these T cells by ribosomal binding proteins can induce a critical
immune response suitable to develop enhanced strategic immune therapies for
many chronic infections. Several studies confirm T lymphocytes functional modifi-
cations as the key pathological change responsible for adaptive immunity. A few
non-hematopoietic and hematopoietic cells, including macrophages, ordinary DCs
(cDCs), and plasmacytoid DCs (pDCs), can add to the first vigorous IFNI reaction
MMP-9
Pro-inflammatory Anti-inflammatory
1. Antibodies play a vital role in the treatment of viral infections, but a new class of
human monoclonal antibodies called super-antibodies are drawing attention in
immunotherapy. These antibodies are rarely seen in human infections. They are
majorly generated during screening using the B-cell approach. This strategy is
mainly implemented to increase the half-life and immune functions in the pro-
phylaxis and treatment of a wide range of viral infections. This strategy can be a
promising approach in the future (Walker and Burton 2018).
7 Significant Role of Cellular Activation in Viral Infections 119
2. The current antiviral therapies for HBV are effective but need to be administered
for a long period without allowing the virus to rebound or replicate. There are
some immunotherapies which focus on the response of the host to the virus and
maintaining the viral load under control. One of the best strategies is to develop
the antiviral drug based on the toxic outcome and therapeutic efficiency to reduce
the viral levels by immunotherapy studies in the varied populations (Bertoletti
and Le Bert 2018).
3. The new approach in the targeting of the immunodeficiency diseases like HIV,
HBV, and HCV is to target the T regulatory cells which play a major role in
protecting important organs like the liver and kidneys from injury due to immu-
nosuppression. This approach will help protect a vast majority of the white blood
cells that are destroyed by viral infections whose primary goal is to eliminate the
large populations of the macrophages (Karkhah et al. 2018).
4. Another very successful strategy in the treatment of viral infections soon after
transplantation of the stem cells of the blood is to utilize the T lymphocytes taken
from the infected donors who are specific to virus. This can treat patients suffer-
ing from primary immunodeficiency disorders like HIV. This type of therapy is
also known as adoptive immunotherapy (Keller et al. 2018).
5. In one approach DNA was collected from different patients with immunodefi-
ciency disorders and transcriptional analysis was done on the genome to study
the genetic sequences coding for viral replication by nuclear protein binding.
The outcome is studied through mass spectroscopy and the allele or non-allele
dependent messenger RNA transcription is done in possible ways (Lummus
et al. 2018).
6. Both the lymphocytes aggregate and get compromised in the lung injuries like
asthma induced by dust particles. To study the extent of the exposure and the
injury, various fluids from the bronchiolar tissues and the alveoli are collected
and antibodies are confirmed by ELISA or western blotting techniques to study
autoimmunity associated with it. Even the lung tissues can be stained for the
identification of particular proteins (Poole et al. 2018).
There are various genes which get expressed in good amount during T-cell activa-
tion during infections like Epstein-Barr virus. The natural regulatory T cells are
usually involved in the activation of the receptors on the cell surface like the CD25.
The T lymphocytes develop in the thymus and are differentiated into regulatory and
non-regulatory cells when they migrate to the peripheral region of the thymus gland.
These regulatory cells are further divided into natural regulatory and the induc-
ible regulatory T cells and include various other subtypes. All these cells are classi-
fied based on their role in the process of eliciting the immune response according to
the invading mechanism of the pathogen.
120 B. Chidipi et al.
The pathophysiology of viral infection is very complex unless we can identify the
type of virus species more specifically under careful correlation with the clinical
symptoms of the infection. This can help in determining the pathogenic process and
orientation of the disease. All the cells in the immune system rely on the energy
pathways to survive and produce the response related to their metabolism. Amino
acids like arginine, tryptophan, and alanine play a major role in the creation of the
immune response during pathological conditions and inhibition of the unwanted
immune response during viral attack. Also, the HIV proteins Vpr and Vif initiate
IRF3 debasement (31), and direct HIV gp120 communication with pDCs restrains
TLR9 (however not TLR7) reactions, including PDC actuation and IFNα discharge.
Additionally the HIV capsid connects with chosen have cofactors to avoid PRR
detecting in macrophages. The previously mentioned hidden dimensions of natural
qualities ahead of schedule after in vivo HBV contamination might be mostly clari-
fied by HBV polymerase impedance with IRF3 phosphorylation or potentially by
hindrance of MAVS motioning by the viral HBx protein. Also, by the diminished
TLR2 articulation in hepatocytes, monocytes, and Kupffer cells (liver-inhabitant
macrophages) from ceaseless HBV patients, HBV dissolvable antigen can decrease
articulation of TLR2 in hepatic cell lines. Thus, HBV dissolvable antigen likewise
stifles TLR9 (however not TLR7) motioning in pDCs, and TLR9 articulation is
diminished in blood pDCs from incessantly contaminated patients. Notwithstanding
the direct IFN-I inhibitory exercises of viral proteins, infections that build up unend-
ing diseases additionally advance host immunomodulatory reactions that may cause
deviant working of natural cells (Coe et al. 2014).
7 Significant Role of Cellular Activation in Viral Infections 121
Since we can only control but not cure most of the viral infections, these strategies
can help in focusing on the host cell death pathways, thereby developing drugs that
can give complete immunity against the virus by facilitated viral cell elimination.
Some of the virus will escape the host immune system and become persistent in the
human body causing infection in the future immunocompromised pathological
conditions.
IFN-I antiviral impacts are interceded by acceptance of a few ISGs that have the
capacity to impede viral replication through different systems [e.g., protein interpre-
tation restraint, viral RNA debasement (investigated by Brown et al. 2018). IFN-I
likewise coordinates both inborn and versatile resistant cells, for example, DCs,
macrophages, NK cells, and T cells following viral diseases, giving enactment and
survival signals. It reliably examines persevering LCMV contamination, in which
the infection is cleared from the blood and most tissues by 2–3 months postinfec-
tion, showing that mice lacking IFNαβR build up long-lasting viremia. Besides,
Sandler et al. exhibited that treatment of rhesus macaques with IFN αβR killing
counteracting agent amid serious SIV contamination brought about improved CD4+
T-cell consumption, diminished antiviral quality articulation, decreased extents of
cytotoxic NK cells, and expanded SIV repositories, all associated with quicker
movement to simian AIDS. What is more, treatment with recombinant IFN-I can
regularly support the host barrier and upgrade viral control (Cooney et al. 2018).
A plethora of examples of such infections are human immunodeficiency virus,
Epstein-Barr virus, hepatitis B and C virus, and so on. These viruses can be preva-
lent irrespective of the advancement in the treatment and vaccination. For instance,
amid incessant HCV disease, PDC IFN-I creation limit is disabled, in any event to
a limited extent, by monocyte-determined tumor rot factor α (TNFα) and interleukin
122 B. Chidipi et al.
(IL)-10. It ought to be noticed that not all parts of natural reactions are weakened
amid unending viral diseases; in fact, select pathways are improved. For example,
though LCMV Cl13-tainted mice react ineffectively to TLR9 ligand (CpG) chal-
lenge, they produce unusually high IFN-I levels upon TLR4 ligand (LPS) incite-
ment, prompting quick passing. By these perceptions, unending HCV contamination
likewise advances hyperresponsive macrophages, including Kupffer cells, adding to
the constant liver irritation that is normal for endless HCV disease. At long last,
macrophages treated with HIV and antigen-displaying cells detached from HIV-
contaminated patients show hyperresponsiveness to random TLR ligands and com-
mensal microbes, individually. Together, these investigations feature that although
natural resistant cells are equipped for IFN-I reaction in the beginning of most con-
stant viral contaminations, blocking of intrinsic pathways by viral proteins associ-
ated with host immunomodulatory atoms prompts a significant (yet inadequate)
constriction of IFN-I at later phases of the disease (Alvarado-Mora and Pinho 2013).
There are various novel treatment strategies focusing on the host cell death path-
ways to cure these persistent diseases. One involves clearing the latent infected cells
first which have the potential to cause infection; in the future, this can be done by
screening and eluting these cells. But these latent cells are very hard to differentiate
from the healthy cells in the absence of the infection. This strategy is very success-
ful in targeting the HIV virus with antiretroviral therapy. The very high rate of muta-
tions in the virus genome can also be a problem to cure the infection when they
acquire resistance to the antiretroviral drugs administered after the diagnosis and
treatment of HIV (Liang et al. 2015).
Wang and colleagues demonstrated that treatment with recombinant IFNα5 and
IFNβ between days 2 and 5 after LCMV Cl13 contamination (maybe identical to
deferring the IFN-I lessening mentioned above) brought about early popular regula-
tion joined by improved infection explicit CD8+ T-cell reactions, despite the fact
that a similar treatment in the unending period of disease had no impact.
Also, prophylactic treatment with pegylated IFNα2a (pIFNα) preceding SIV
mucosal test prompted expanded protection from contamination interceded to some
degree by an expansion in CD56+ NK cells; in any case, persistent pIFNα treatment
brought about an IFN-I desensitization state with decreased ISG levels and expanded
cell-related viral burdens. Outstandingly, pIFNα additionally improves articulation
of a few ISGs known to be direct enemy of HIV movement. In any case, treatment
with pIFNα in HIV-tainted patients has yielded mixed outcomes; only few investi-
gations have appeared.
In some infections like hepatitis, there are few transcriptionally very active frag-
ments which can code for the viral DNA or RNA and produce new viral fragments.
They invade the host cells and remain attached to the host genome. These fragments
may remain in the body for long periods of time and relapse during favorable condi-
tions producing the antigens. These infective active fragments may get inserted in
the host genome and remain inactive for a period of time which is known as the
latent phase during which screening for the infection is very tiresome (Battivelli
et al. 2018).
7 Significant Role of Cellular Activation in Viral Infections 123
Acknowledgments The authors are grateful for the support extended from Adikavi Nannaya
University, Osmania University, Krishna University, and the University of South Florida, USA.
Conflict of Interest The authors declare that they have no competing interests.
References
Alvarado-Mora MV, Pinho JR (2013) Epidemiological update of hepatitis B, C and delta in Latin
America. Antivir Ther 18(3 Pt B):429–433
Battivelli E, Dahabieh MS, Abdel-Mohsen M, Svensson JP, Da Silva IT, Cohn LB, Verdin E (2018)
Chromatin functional states correlate with HIV latency reversal in infected primary CD4+ T
cells. bioRxiv:242958
Bertoletti A, Le Bert N (2018) Immunotherapy for chronic hepatitis B virus infection. Gut Liver
12:497
Brown GD, Willment JA, Whitehead L (2018) C-type lectins in immunity and homeostasis. Nat
Rev Immunol 18(6):374–389
Bull MJ, Plummer NT (2014) Part 1: the human gut microbiome in health and disease. Integr Med
13(6):17–22
Coe DJ, Kishore M, Marelli-Berg F (2014) Metabolic regulation of regulatory T cell development
and function. Front Immunol 5:590
Cooney J, Allison C, Preston S, Pellegrini M (2018) Therapeutic manipulation of host cell death
pathways to facilitate clearance of persistent viral infections. J Leukoc Biol 103(2):287–293
Finotti G, Migliorati D, Costantini M (2018) Multisensory integration, body representation and
hyperactivity of the immune system. Conscious Cogn 63:61–73
Frossi B, Mion F, Sibilano R, Danelli L, Pucillo CEM (2018) Is it time for a new classification
of mast cells? What do we know about mast cell heterogeneity? Immunol Rev 282(1):35–46
Gao Y, Deason KL, Jain A, Dozmorov I, Wakeland EK, Pasare C (2018) Transcriptional profiling
of pathogen-specific CD4 T cells reveals key events that regulate Th17 cell priming and dif-
ferentiation. J Immunol 200(1):47
Gasteiger G, D’Osualdo A, Schubert DA, Weber A, Bruscia EM, Hartl D (2017) Cellular innate
immunity: an old game with new players. J Innate Immun 9:111–125
Gieseck RL, Wilson MS, Wynn TA (2018) Type 2 immunity in tissue repair and fibrosis. Nat Rev
Immunol 18(1):62–76
Haas S, Trumpp A (2018) An intrinsic interferon program protects stem cells from viral infection.
Dev Cell 44(3):279–280
Karkhah A, Javanian M, Ebrahimpour S (2018) The role of regulatory T cells in immunopathogen-
esis and immunotherapy of viral infections. Infect Genet Evol 59:32–37
Keller MD, Hanley PJ, Hoover J, Roesch L, McCormack S, Lang H, Abraham A, Williams K,
Davila B, Adams R, Leiding JW, Bollard C (2018) Third-party T cell immunotherapy for viral
infections in primary immunodeficiency disorders. J Allergy Clin Immunol 141(2):AB198
Li W, Han J, Wu H (2016) Regulatory T-cells promote hepatitis B virus infection and hepatocel-
lular carcinoma progression. Chronic Dis Transl Med 2(2):67–80
Liang S, Shen Z, Yan J, Liang F, Tang Z, Liu W, Xing H (2015) Low virologic failure and drug
resistance among HIV-infected patients receiving hospital-based ART while care and outreach
through community in Guangxi, China. Front Public Health 3:244
Libby P, Hansson GK (2018) Adaptive immunity in acute coronary syndromes: chicken or egg?
Eur Heart J 39(13):1098–1099
Lummus ZL, Kesavalu B, Kaufman K, Yao J, Weirauch MT, Kottyan LC, Muñoz X (2018)
Regulatory variants of ATF3, CDH17 and FAM71A are risk factors for diisocyanate induced
occupational asthma (DA). J Allergy Clin Immunol 141(2):AB198
124 B. Chidipi et al.
Abstract
Viral infectious diseases are mainly associated with immunological responses trig-
gered by immunocytes present in the circulation like macrophages. A better under-
standing of these immunity mechanisms will help us to control the progression of
the disease to chronic conditions. Microvesicles are an extracellular group of
plasma membrane secretions, which play a significant role in the spread of infec-
tion or evasion of viral entities to surrounding healthy cells acting as a viral vehicle.
Their involvement in the viral cycle can be utilized productively in hampering the
period by developing the antibodies that shield the evasion. These extracellular
vesicles will facilitate the communication between the cells in the body humoral
environment leading to host and viral interactions in the immune system. Thereby
attempting to study the role of macrophages in the immunity process will help us
to eliminate the viral pathogens by either delaying or damaging their spread to
healthy cells. Focusing on these mechanisms of the microvesicles and macro-
phages will help lead to the early cure of many life-threatening infectious diseases
like HIV, Hepatitis, and Dengue by developing a single specific antibiotic.
Keywords
Macrophages · Viral cycle · HIV · Hepatitis · Dengue · Humoral environment ·
Extracellular vesicles
8.1 Introduction
Viral infectious diseases are mainly associated with immunological responses trig-
gered by immunocytes like Macrophages. A better understanding of these immunity
mechanisms and the knowledge related to the prevalence of the infection will help
us to control the progression of the disease to chronic conditions by actual estima-
tion of the number of infected individuals by screening methods (Birdwell et al.
2018). The primary route of entry of the virus into the human body is through the
epithelial lining, every time epithelial cells will be the first cells to get infected or
encounter the virus particles. When infected, one of the surface of the epithelial
cells will get polarized since both the inner and outer surfaces of the epithelial cells
are morphologically distinct (Hoebe et al. 2018). With or without finishing the life
cycle inside the epithelium these viruses may exit the cells to invade one or other
surfaces (Baraldo et al. 2018).
The process by which a viral infection leads to disease is known as viral patho-
genesis, the consequences and the period of the viral infection depend on various
host and viral-related interplaying factors. They can develop as an acute infection
but may last for months to years depending on the favorable viral factors. If the
disease becomes chronic it may easily get transmitted to others and also recovery
from such conditions is sporadic (Jaimes and Whittaker 2018). The cell damage
created by the infection depends on the type of virus because not all viruses can
damage cells even though they replicate and widely spread throughout the body.
Some viruses belonging to the class known as retrovirus can cause persistent infec-
tion but cannot cause cell death as they are released from the cells without any lysis
by a unique phenomenon called budding (Shaheen 2018).
Whereas picornavirus can cause lysis of cell and cause cell damage and rhinovi-
rus cause mucus secretion and poliovirus paralysis the tissues causing tremendous
cell damage. The progressive clinical symptoms are noticed with these infections
until the viral population minimizes to noninfectious levels (Carrasco et al. 2018).
An infection must utilize cell procedures to repeat. The viral replication cycle can
create dramatic biochemical and fundamental changes in the host cell, which may
cause cell harm. These changes, called cytopathic (causing cell harm) impacts, can
change cell function. Some infected cells, for example, those contaminated by the
regular cold infection known as rhinovirus, kick the bucket through lysis (bursting)
or apoptosis (customized cell demise or “cell suicide”), discharging all offspring
virions without a moment’s delay. The side effects of viral maladies result from the
insusceptible reaction to the infection, which endeavors to control and dispense
with the virus from the body, and from cell harm brought about by the virus
(McDougall et al. 2019).
Numerous viral infections, for example, HIV (human immunodeficiency infec-
tion), leave the contaminated cells of the safe framework by a procedure known as
8 Macrophages/Microvesicles and Their Task in Viral Diseases 127
sprouting, where virions go to the cell separately. Amid the growing process, the
cell does not experience lysis and is not promptly slaughtered. In any case, the harm
to the cells that the infection taints may make it outlandish for the cells to work
ordinarily, even though the cells stay alive for a timeframe. Most beneficial viral
diseases pursue comparative strides in the infection replication cycle: connection,
entrance, un-coating, replication, gathering, and discharge. In Fig. 8.1, the host cell
is annihilated toward the finish of the replication cycle—recollect this does not gen-
erally occur: here and there the host cell lives on and keeps on reproducing the
infection (Khalili and Malcolm 2019).
Connection An infection joins to a particular receptor site on the host cell film
through connection proteins in the capsid or using glycoproteins inserted in the viral
envelope. The explicitness of this communication decides the host—and the cells
inside the host—that can be tainted by a specific infection. This can be outlined by
thinking about a few keys and a few locks, where each key will fit just a single
explicit lock (Zhao et al. 2019).
Entry Phase The nucleic acid of bacteriophages enters the host cell exposed, leaving the
capsid outside the cell. Plant and creature infections can enter through endocytosis, in
which the cell film encompasses and overwhelms the whole virus. Some wrapped infec-
tions come to the cell when the viral envelope melds legitimately with the cell layer. Once
Fig. 8.1 Microvesicle formation mechanism. (Top panel) In resting cells, both phosphatidylserine
and phosphatidylethanolamine are negatively charged and segregated in the inner side of the
plasma membrane, on the other hand sphingomyelin and phosphatidylcholine, both are positively
charged head group, are in the outer side of the plasma membrane (Seigneuret et al. 1984). (Lower
panel) The “flippase” and “floppase are an ATP-dependent transmembrane enzyme. Flippase
reacts with phosphatidylserine and help to inward folding, meanwhile floppase intermediate the
both anionic and cationic phospholipids to outward fold (Daleke 2003; Laberge et al. 2018)
128 B. Chidipi et al.
inside the cell, the viral capsid is debased, and the viral nucleic acid is discharged, which
at that point ends up accessible for replication and translation (Appaiah et al. 2019).
Replication Phase The replication instrument relies upon the viral genome. The
viral infections generally use cell proteins and compounds to make extra DNA that
is translated to ambassador RNA (mRNA), which is then used to coordinate protein
union. RNA infections, for the most part, utilize the RNA center as a layout for the
union of viral genomic RNA and mRNA. The viral mRNA guides the host cell to
orchestrate viral catalysts and capsid proteins, and gather new virions (Ilan 2019).
There are individual cases to this example. On the off chance that a host cell does
not give the compounds essential to viral replication, viral qualities supply the data
to coordinate the union of the missing proteins. Retroviruses, for example, HIV,
have an RNA genome that must be turned around translated into DNA, which at that
point is consolidated into the host cell genome (Solomon and Geretti 2019).
The way that HIV creates its very own portion compounds not found in the host
has enabled scientists to develop drugs that hinder these chemicals. These medica-
tions, including the turnaround transcriptase inhibitor AZT, inhibit HIV replication
by diminishing the movement of the protein without influencing the host's diges-
tion. This methodology has prompted the advancement of an assortment of medica-
tions used to treat HIV and has been successful at decreasing the number of
irresistible virions (duplicates of viral RNA) in the blood to non-discernible dimen-
sions in numerous HIV-contaminated people (Roder et al. 2019).
Budding Phase The last phase of viral replication is the arrival of the new virions
delivered in the host living being, the place they can taint neighboring cells and rehash
the replication cycle. As you have adapted, some infections are discharged when the
host cell bites the dust, and different infections can leave contaminated cells by sprout-
ing through the layer without legitimately slaughtering the phone (May 2019).
8.3 Microvesicles
Microvesicles have many common characteristics with that of the enveloped viruses.
During viral infections, many infected cells secrete a vast number of microvesicles
that may or may not be similar to the biogenic and biophysical characteristics of
viral counterparts. For instance, HIV virions will shed from the plasma membrane
to but not from the cytoplasm in a similar budding mechanism like microvesicles
spread from cell to cell. Moreover, it is tough to separate HIV particles from exo-
some vesicles during centrifugation as they both have identical biological properties
(Bello-Morales et al. 2018a).
Microvesicles as discussed above will play a vital role in assisting the virally
infected cells in promoting the intercellular microvesicle-mediated communication.
These vesicles, when fused to the surface of the other cells, will eventually transfer the
cellular components exerting a biological function to the recipient cells. During infec-
tion with herpes simplex virus, microvesicles promote the viral pathogenesis and
infectivity by using the exosome components and endosome makers like tetraspanin.
130 B. Chidipi et al.
Fig. 8.2 Size and number of microvesicles. (a) Cryo-electron micrograph of extracellular vesicles
secreted by MLP-29 cells (Conde-Vancells et al. 2008). The percentage of vesicles that belonged
to each category and their size distribution within each category are shown in cultured HMC-1
cells (b, c) (Zabeo et al. 2017)
These particles are similar to the endosome vesicles in size making them use the exo-
some cellular pathways for their invasion and evasion (Sharma et al. 2018).
The primary functions of the microvesicles in the viral infection are to modulate
the cellular processes like angiogenesis, cell proliferation, cell invasion, gene regu-
lation, and immune regulation. These can also generate the essential ligands for
receptor cell surfaces to initiate the signaling pathways and release their contents
into the plasma membrane by endocytosis. Their involvement in the viral cycle can
be utilized in hampering the period by developing antibodies that shield the evasion.
Virally infected cells will sometimes secrete microvesicles to remove unwanted cel-
lular proteins from the cytoplasm thereby regulating the signaling complexes and
pathological process (Meckes and Raab-Traub 2011).
8 Macrophages/Microvesicles and Their Task in Viral Diseases 131
Extracellular vesicles facilitate the connection between the cells in the body humoral
environment leading to host and viral interactions in the immune system. These
extracellular vesicles are usually the exosome or microvesicles containing RNA’s,
proteins, and lipids (Birdwell et al. 2018). These involve in the extracellular com-
munication by a unique mechanism in which the cells selectively discharge mem-
brane and elute the internal components. This mechanism is identified using the
exosome secreted by antigen-presenting cells major histocompatibility complex 2
using the immunogold labels (Paolicelli et al. 2018).
Even in the tumor environment, extracellular vesicles (EVs) play a vital role in
the mediation of communication between the tumor cells. EVs contribute to regulat-
ing the bystander effects in which the stressed tumor cells will cause DNA damage
to the neighboring healthy cells (Birdwell et al. 2018). Evidence shows that exo-
somes help in the cross-communication between cells from which they were derived
from, for example, between macrophages, dendrocytes, both T and B lymphocytes,
and natural killer cells in the cancer environment. These exosomes will influence
the functions of the cells by modifying their genotype due to mutations in the cancer
cells. These are playing a vital role in the generation of the vaccines for various
diseases involved with macrovesicle mediation (Raposo et al. 1996).
The significant contribution of these vesicles in their release and function by
biogenesis will be the novel tool for the detection and treatment of the viral dis-
eases. It has been revealed that they contribute to homeostasis in the central nervous
system trauma conditions (Baraldo et al. 2018). During the therapeutic purpose, the
same exosomes play a vital role in targeting and modifying specific tissue cells and
combating the significant barriers in immunological diseases. They also help in the
development of various vaccines against those infectious diseases that mimic the
microvesicles for exploiting the host immune system (Samuel et al. 2017).
Macrophages play a vital role in the generation of the immune response in the
human body (Baraldo et al. 2018). Thereby attempting to study the role of macro-
phages in the immunity process will help us to eliminate the viral pathogens by
either delaying or damaging their spread to healthy cells. Macrophages also contrib-
ute a vital role in increasing the genome load for viral infections like infectious
bronchitis virus as they accumulate in the respiratory tract (Shen and Ren 2018).
During viral infections, a large number of infected cells will undergo a pro-
grammed cell death process called apoptosis during which the macrophages elimi-
nate the cells preventing further necrosis (Shaheen 2018). They also play a supportive
role in the activation of the tissue repair process by activating the inflammatory
cascade by recruiting inflammatory mediators thereby initiating the wound healing
process (Pegtel et al. 2014).
132 B. Chidipi et al.
In infections like hepatitis, the microvesicles budding from the infected cells will
participate in the viral cycle along with the virus. This was the first time discovered
under the electron microscope when observing the infected liver cells for the herpes
simplex virus virions. In hepatitis infection, microvesicles harbor the virions as part
of the viral cycle, and when these are ingested by the macrophages they are infect-
ing the naïve cells as part of the productive infection. In hepatitis, virus particles are
depending on the microvesicles to promote the disease effectively in the host, which
can be used therapeutically by shielding the microvesicles with antibodies (Huang-
Doran et al. 2017).
In dengue infection, the virus uses the arthropod vesicles to promote the disease
from the mosquito to humans. These extracellular vesicles contain the viral RNA
and proteins that are infectious. In the treatment of the dengue infection effectively
scientists are using the glycoprotein present in the tetraspanin domain of the
microvesicle as a marker to identify the pathophysiology and blocking the spread of
disease by targeting the microvesicle-mediated transmission by DNA silencing and
gene isolation studies. These changes are confirmed by immunoprecipitation stud-
ies (Vora et al. 2018).
In HIV infection microvesicles play a vital role in the remodeling of the pulmo-
nary vasculature. The actual reason for the cardiac regeneration in the humans is
anonymous for recent times and later discovered to be due to the extracellular vesi-
cles released from the human monocyte-derived macrophages. The main constitu-
ent of these vesicles is miRNA-130a that plays a significant role in the decrease in
the expression of the phosphatase and tensin molecules that are responsible for the
perivascular inflammation in the HIV patients (Sharma et al. 2018).
With respect to entry and departure of virus, four noteworthy stages have been
proposed to portray these procedures: capsid get together and DNA bundling in the
core; essential envelopment and de-envelopment at the atomic envelope; tegumen-
tation and optional envelopment in the cytoplasm; and, at long last, exocytosis of
viral particles at the plasma layer and additionally cell-to-cell transmission at cell
intersections (Lannes et al. 2019). A unique method of viral communication in
human tissues is cell-to-cell spread, that is, the immediate entry of descendant’s
infection from a contaminated cell to a nearby one (Bracq et al. 2018) (Fig. 8.3).
It is broadly acknowledged that this system of dissemination speaks to a resistant
avoidance technique since it shields the infection from insusceptible observation.
Be that as it may, as referenced above, HSV-1 may utilize a few methods of spread
to go from tainted to uninfected cells (You et al. 2018). Numerous viewpoints con-
cerning the procedure of viral spread, for example, the instruments of viral depar-
ture from epithelial cells and spread to neurons and the other way around, are not
seen yet (Bayliss and Piguet 2018).
Elucidating the devices of viral scattering and the ensuing passage into neighbor-
ing cells remains a vital advance to comprehend the viral cycle in the host. In this
unique situation, discharged vesicles have risen as another object of consideration
given their capacity to take part in the intercellular correspondence process amid
8 Macrophages/Microvesicles and Their Task in Viral Diseases 133
Fig. 8.3 Intercellular structures and processes involved in cell-to-cell transmission of HIV-1. (a–
g) Schemes represent the different pathways for HIV-1 cell-to-cell transfer between donor cells (in
green) and target cells (in pink) (Bracq et al. 2018)
viral diseases. Extracellular vehicles (EVs) are a very different gathering of emitted
layer vesicles, which have been separated from most cell types and natural liquids.
Three noteworthy subgroups of EVs have been recognized: apoptotic bodies;
microvesicles (MVs), which get from the shedding of the plasma film; and exo-
somes, which are intraluminal vesicles discharged to the endless extracellular supply
of multivesicular bodies (MVBs) with the plasma layer (Fig. 8.4) (Roth et al. 2019).
Exosome usually is 30–100 nm in the distance across, while MVs have a differ-
ent size running from 100 nm to 1 m in breadth. EVs have been appeared to be
associated with various physiological and neurotic procedures, for example, aggra-
vation and the insusceptible reaction, cell bond, coagulation, squander the board,
tumor movement, and viral spread. Oligodendrocytes (OLs) are the myelin-shaping
cells of the focal sensory system (CNS). The myelin sheath is an electrically pro-
tecting layer that encompasses axons in both the focal and fringe sensory systems,
permitting salutatory conduction of activity potential (Domingues et al. 2018).
All neural cell types discharge EVs, which have a focal job in procedures, for
example, myelination or guideline of synaptic movement and may be associated with
the pathogenesis of a few neurodegenerative maladies or, despite what might be
expected, in neuroprotection. To be sure, OLs discharge exosomes conveying myelin
proteins, for example, proteolipid protein (PLP), the real myelin protein in the CNS;
134 B. Chidipi et al.
Fig. 8.4 Multiple stages of the retroviral life cycle, however, no current antiretroviral drug blocks
the expression of HIV proteins and RNA from integrated viral DNA, or their subsequent sorting
into exosomes and secretion from infected cells (Patters and Kumar 2018)
named L-particles, are like the virions in appearance and do not have the viral
nucleocapsid and the genome and along these lines are not irresistible (Bello-
Morales et al. 2018a). In any case, L-particles have been appeared to encourage
HSV-1 disease by conveying viral proteins and the cellular factors required for
infection replication and invulnerable avoidance (Bello-Morales and López-
Guerrero 2018).
Hence, it has been confirmed that the comparative capacities watched for vhs and
TIF among virions and L-particles recommend that viral connection, combination,
and the arrival of covering proteins are the equivalent for both. Likewise, L-particles
share comparative gathering and departure pathways with virions, recommending
that the covering and glycoproteins are adequate to provoke auxiliary envelopment.
It has been exhibited that useful viral proteins can be exchanged to uninfected
onlooker cells by means of L-particles, a procedure that may show a methodology
for viral invulnerable break. Different particles, the previral DNA replication-
encompassed particles (PREPs), are morphologically like L-particles; however,
they vary in their relative protein arrangements (Sun et al. 1999). Nonetheless, to
date, there is no proof of virions being bundled inside EVs. Here, we propose a
unique job for MVs in spread of infection. Recent discoveries show out of the blue
that virions might be exchanged from tainted to uninfected cells through MVs. By
methods of transmission electron microscopy (TEM), we identified miniaturized
scale vesicles containing HSV-1 virions. What is more, we found that the no lenient
Chinese hamster ovary (CHO) cell line was powerless to HSV disease directly after
immunization with infection containing MVs recently detached from supernatant of
infected HOG cells. These outcomes recommend that MVs emitted by HOG cells
tainted with HSV-1 may be associated with viral spread and may add to evading
resistant reconnaissance (Bello-Morales et al. 2018b).
Acknowledgments The authors are grateful for the support extended from Adikavi Nannaya
University, Krishna University, and University of South Florida, USA.
Conflict of Interest The authors declare that they have no competing interests.
References
Appaiah CB, Manur J, Sriram B (2019) Reducing conjugative plasmids in bacteria. Google Patents
Baraldo S, Contoli M, Bonato M et al (2018) Deficient immune response to viral infections in
children predicts later asthma persistence. Am J Respir Crit Care Med 197(5):673–675
Bayliss RJ, Piguet V (2018) Masters of manipulation: viral modulation of the immunological syn-
apse. Cell Microbiol 20(10):e12944
Bello-Morales R, López-Guerrero JA (2018) Extracellular vesicles in herpes viral spread and
immune evasion. Front Microbiol 9:2572
Bello-Morales R, Praena B, de la Nuez C et al (2018a) Role of microvesicles in the spread of her-
pes simplex virus 1 in oligodendrocytic cells. J Virol 92(10):e00088
136 B. Chidipi et al.
Abstract
Viral diseases are a major cause of morbidity and mortality and result in a signifi-
cant public health burden. T lymphocytes first identified in the chordate lineage
and constitute a highly sophisticated branch of adaptive immune system. Apart
from B cells, it is the only cell type that exhibits antigenic specificities; achieved
by gene rearrangement. T cells are unique with respect to diversity of their sub-
sets, which have distinct effector specificities, proliferative abilities, memory
generation, and life span. T cells are impactful in viral infections by virtue of
their capability to combat intracellular pathogens. The effector functions of T
cells are mediated through cytokines/chemokines and by direct cytotoxicity of
infected cells. T cell response can be beneficial or detrimental to host; prognosis
depending on qualitative and quantitative differences in the response. Persistent
viral infections are associated with functionally suboptimal, exhausted T cell
responses, which are unable to clear virus. Specific subsets such as regulatory T
cells (Tregs) dampen antiviral responses; thereby favouring viral persistence.
However, Tregs protect the host from immunopathology by limiting perpetual
inflammation. Certain other subsets such as Th17 cells may contribute to autoim-
mune component of viral infections. The importance of T cells is highlighted by
the fact that modern vaccination and therapeutic approaches focus on modulating
T cell frequencies and effector functions. This chapter emphasises the under-
standing how T cells influence outcomes of viral infections, modern vaccination
and therapeutic strategies with thrust on T cell biology.
A. Jagadeesh
Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul
National University, Seoul, South Korea
A. M. V. N. Prathyusha · G. M. Sheela · P. V. Bramhachari (*)
Department of Biotechnology, Krishna University, Machilipatnam, Andhra Pradesh, India
Keywords
Adaptive immune system · T cell responses · Viral infections · Therapeutic
strategies
9.1 Introduction
Viruses are infectious agents consists of nucleic acids coated in a simple protein
casing, infects, replicates in host cells and causes acute, chronic infections.
Mammals established a refined immune system to cope with several viral and bacte-
rial infections (García-Sastre and Biron 2006). Remarkably, adaptive arm of
immune responses is significant in action against virus particles and infected cells.
T Lymphocytes are the key players in adaptive, cell-mediated immune responses
and also in elimination of foreign pathogens by activating various immune responses.
There exist many viruses for which both CD8+ and CD4+ T cells, reported to play
key role in viral control viz. measles virus (Nelson et al. 2017), cytomegalovirus
(CMV) (Wehrens et al. 2016), hepatitis C virus (HCV) (Sheiko et al. 2016) and HIV
(Jones and Walker 2016). Typically, TH1 cells are assumed to be efficient in antiviral
T cell response. Nonetheless, many viruses can inhibit TH1 response by downregu-
lating interferons release from infected cells, which greatly influence outcome of
virus infection (Laidlaw et al. 2016). The humoral immune response involves anti-
bodies specific for virus to block host–virus interactions, neutralize virus and recog-
nise viral antigens on infected cells and activates antibody-dependent cytotoxic
cells (ADCC) or complement-mediated lysis to kill infected cells. However, if these
antibodies are ineffective, viruses are able to infect host cells where adaptive arm
acts to control viral pathogenesis (Rosendahl Huber et al. 2014).
Virus infects host cells if humoral immunity fails, viruses use protein synthesis
and replication machinery of host cells for their replication and synthesis of their
own proteins. Some newly synthesized proteins may degrade into peptide frag-
ments. If these peptides have sufficient binding affinity, to class I MHC molecules
they appear on cell surface of an infected cell as class I MHC–peptide complex.
This complex activates CD8+ T cells and induces infected cell apoptosis by releas-
ing cytotoxic granules and production of TNF-α and IFN-γ. Activation of CD8+ T
cells also occurs in draining lymph nodes, where antigen-presenting cells (APCs)
encounter naïve T cells. Priming of naïve T cells will not only occur through classi-
cal pathway via infection of cell, but also through cross-presentation of viral pep-
tides on MHC class I molecules, taken up from extracellular sources. Priming of T
cells triggers a massive expansion of antigen-specific T cells. Their progeny usually
accumulate in large numbers of armed effector T cells and these normally contribute
to the eradication of viral pathogens.
T cells in chronic viral infections typically exhibit strong impairments in the
production of cytokines (IFN-g, TNF and IL-2) and express high levels of inhibitory
receptors viz. PD-1 (programmed cell death-1) and Lymphocyte-activation gene 3
(Lag-3). These phenotypic changes along with failure of immune system to clear
9 T Cells in Viral Infections: The Myriad Flavours of Antiviral Immunity 141
There are more than 8000 cases of respiratory diseases among which the Severe
acute Respiratory Syndrome (SARS) is caused by novel coronavirus (SARS-CoV),
and contributed to 10% of mortality in 2002–2003. Pro-inflammatory responses
enhance disease progression. The mechanism of immune evasion is mainly charac-
terised by poor antigen presentation by antigen presentating cells (Legge and
Braciale 2003). The antigen presentation is key for activation of T cell and produces
chemokines and cytokines that regulate disease progression (Seder et al. 2008). The
immune evasion of novel coronavirus targets APC and suppresses T cell activation.
Dendritic cell immunisation activates T cells. As a result, the production of IFN-
gamma, IL-2 and TNF-alfa are released (Zhao et al. 2010). Therefore, the viral titre
gets reduced as immunisation with Dendritic cells (DC) by T cell activation suc-
cessfully suppressed viral pathogenesis. It can be characterised as a potent immuno-
gen to activate immune response.
West Nile Virus is a positive sense single-stranded RNA belongs to Flaviviridae and
transmitted by mosquito vectors and originated in the USA in 1999, it causes mos-
quito borne encephalitis. It is asymptomatic in majority of individuals and symptoms
usually are arthralgia, myalgia and cephalea. The minor part of pathogenesis can be
neurologic deficits and neuroinvasive in elderly people. Viral pathogenesis mainly
142 A. Jagadeesh et al.
The viral hemorrhage fever (VHF) majorly regulates T cell priming, effector T
cell response and T cell activation (Dahlke et al. 2017). Early during VHF, the
antigen-presenting cells produce high levels of IFN-gamma and TNF-alfa and
IL-6. As a result, activation of T cells increases viremia and overproduction of
IFN-gamma and TNF-alfa this results in over activation of T cells (Perdomo-Celis
and Salvato 2019) This condition is termed as cytokine storm. During VHF the
proportions of regulatory T lymphocytes decreases and therefore, this enhances
disease pathogenicity.
The chronic and viral infections effect a plethora of T cell populations drastically.
The CD4+ T cells and CD8+ T cells are major players in regulating viral pathogen-
esis. The other differentiated T cell called regulatory T cell, which regulates immune
responses and inflammatory responses. During chronic and viral infections, T reg
plays important role in regulating immune responses and various cytokines (Keynan
et al. 2008). As a result, it regulates effector T cells population. In chronic viral
infections, depletion of Treg cells, CD8+ T cell proliferation (Boettler et al. 2005),
IFN-gamma production and increase cytolytic activity are predominant (Haeryfar
et al. 2005). In HIV infection, due to decrease in Treg population, hyper activation
of CD4+ and CD8+ T cells is predominant (Oswald-Richter et al. 2004) As a result,
it favours viral replication.
Human Papilloma Virus (HPV), a small DNA virus majorly infects birds, reptiles
and mammals, there are 300 viral genotypes that have been discovered till now
(Vande and Klingelhutz 2013). The HPV infects mucosal and/or cutaneous skin and
causes benign or malignant tumours. HPV associates with cervical cancer, oral
squamous carcinoma and Head and Neck cancers (Forman et al. 2012). The HPV
oncoproteins, E6 and E7, majorly regulates the host immune responses and plays
vital role in tumorigenesis (den Boon et al. 2015). The HPV16 viral particle involves
in the host immune dysregulation by epigenetic mechanisms. The viral pathogene-
sis regulates the synthesis of chemokines that required for T cell activation, i.e.
CXCL14 (Cicchini et al. 2016). The CXCL14 synthesis regulated epigenetically by
HPV viral protein, i.e. E7. Likewise, the HPV viral protein interacts with the host
DNMT1 and stimulates the methylase activation (Burgers et al. 2007) and as a
result, CXCL14 is repressed. Therefore, evasion of immune responses against virus,
by inhibiting the T cell activation. The HPV16 E7 suppresses the production of pro-
inflammatory responses like IL-8, IL18, CCL2, CCL20 (Cho et al. 2001; Guess and
McCance 2005; Huang and McCance 2002; Kleine-Lowinski et al. 2003)
144 A. Jagadeesh et al.
There are more than 350 million people infected with the Hepatitis B virus. The
virus mainly infects liver and causes chronic and acute pathology. The infection
carried commonly by mother to child during birth. The one in all among the viruses,
which is a non-retroviruses that uses the host RNA polymerase for its transcription
and as a result, the closed circular DNA gets transcribed and involved in the host
disease pathogenesis. The adaptive immunopathogenesis plays important role in the
disease progression. The liver residing APC plays an important role in the activation
of naïve CD4 T and CD8 T cells via cross-priming and facilitates the persistence of
the virus (Lan et al. 2016). The downregulation of the TLR-7 and TLR-9 in plasma-
cytoid dendritic cell and as a result, the IFN signalling is inactivated and cytokines
are inhibited (Seeger and Mason 2000). The NK cells are also major cells providing
immune to the HBV by expressing death ligand and inactivating the CD4 T cells
Bertoletti and Ferrari 2016). The CD8 T cells exhaustion is the major episode in the
disease pathology by expressing death receptors such as TIM-3, PD-1 and 2B4,
poor proliferative signal, IL-2 and IFN-gamma (Raziorrouh et al. 2010). The CTL
apoptosis of CTL by upregulation of various apoptotic genes likewise, Bim and
TRAIL-R2. Due to downregulation of T-bet results in exhaustion of the T cells.
Zika virus is mainly infected by mosquito borne flavivirus and it has unexpected
links with microcephaly and Guillain–Barre syndrome. It is hypothesised as the
infection resultant to the testis damage. The Zika viral infection results in the expres-
sion of TIM-4, which is one of the phagocytic markers and facilitates viral replica-
tion (Osuna et al. 2016; Zhang et al. 2018). The population of lymphocyte subsets
are reduced in the period of infection. The virus getting successfully evading the
host immune responses. The current study in combatting the viral immune evasion
9 T Cells in Viral Infections: The Myriad Flavours of Antiviral Immunity 145
mainly on designing the RNA vaccines and T cell epitope tetramer for activation of
T cells, recombinant vector-based vectors (Zang et al. 2018).
Conflict of Interest The authors declare that they have no competing interests.
References
Aguilar-Valenzuela R, Netland J, Seo Y-J, Bevan MJ, Grakoui A, Suthar MS (2018) Dynamics
of tissue- specific CD8+ T cell responses during West Nile virus infection. J Virol
92:e00014–e00018
Ashrafi GH, Haghshenas MR, Marchetti B, O’Brien PM, Campo MS (2005) E5 protein of
human papillomavirus type 16 selectively downregulates surface HLA class I. Int J Cancer
113:276–283
Ashrafi GH, Brown DR, Fife KH, Campo MS (2006) Down-regulation of MHC class I is a prop-
erty common to papillomavirus E5 proteins. Virus Res 120(1–2):208–211
Bertoletti A, Ferrari C (2016) Adaptive immunity in HBV infection. J Hepatol 64:S71–S83
Boettler T, Spangenberg HC, Neumann-Haefelin C et al (2005) T cells with a CD4+CD25+ regu-
latory phenotype suppress in vitro proliferation of virus-specific CD8+ T cells during chronic
hepatitis C virus infection. J Virol 79:7860–7867
Burgers WA, Blanchon L, Pradhan S, de Launoit Y, Kouzarides T, Fuks F (2007) Viral oncopro-
teins target the DNA methyltransferases. Oncogene 26:1650–1655
Cho YS, Kang JW, Cho M, Cho CW, Lee S, Choe YK, Kim Y, Choi I, Park SN, Kim S, Dinarello
CA, Yoon DY (2001) Down modulation of IL-18 expression by human papillomavirus type 16
E6 oncogene via binding to IL-18. FEBS Lett 501:139–145
Cicchini L, Westrich JA, Xu T, Vermeer DW, Berger JN, Clambey ET, Lee D, Song JI, Lambert
PF, Greer RO, Lee JH, Pyeon D (2016) Suppression of antitumor immune responses by human
papillomavirus through epigenetic downregulation of CXCL14. MBio 7:e00270-16
Dahlke C, Lunemann S, Kasonta R, Kreuels B, Schmiedel S, Ly ML, Fehling SK, Strecker T,
Becker S, Altfeld M et al (2017) Comprehensive characterization of cellular immune responses
following ebola virus infection. J Infect Dis 215(2):287–292
Den Boon JA, Pyeon D, Wang SS, Horswill M, Schiffman M, Sherman M, Zuna RE, Wang
Z, Hewitt SM, Pearson R, Schott M, Chung L, He Q, Lambert P, Walker J, Newton MA,
Wentzensen N, Ahlquist P (2015) Molecular transitions from papillomavirus infection to cervi-
cal precancer and cancer: role of stromal estrogen receptor signaling. Proc Natl Acad Sci U S
A 112:E3255–E3264
Duangchinda T, Dejnirattisai W, Vasanawathana S, Limpitikul W, Tangthawornchaikul N, Malasit
P et al (2011) Immunodominant T-cell responses to dengue virus NS3 are associated with
DHF. Proc Natl Acad Sci U S A 107(39):16922–16927
Forman D, de Martel C, Lacey CJ, Soerjomataram I, Lortet-Tieulent J, Bruni L, Vignat J, Ferlay J,
Bray F, Plummer M, Franceschi S (2012) Global burden of human papillomavirus and related
diseases. Vaccine 30(Suppl 5):F12–F23
García-Sastre A, Biron CA (2006) Type 1 interferons and the virus-host relationship: a lesson in
detente. Science 312(5775):879–882
Guess JC, McCance DJ (2005) Decreased migration of Langerhans precursor-like cells in response
to human keratinocytes expressing human papilloma virus type 16 E6/E7 is related to reduced
macrophage inflammatory protein-3α production. J Virol 79:14852–14862
Haeryfar SM, DiPaolo RJ, Tscharke DC, Bennink JR, Yewdell JW (2005) Regulatory T cells
suppress CD8+ T cell responses induced by direct priming and cross-priming and moderate
immunodominance disparities. J Immunol 174:3344–3351
9 T Cells in Viral Infections: The Myriad Flavours of Antiviral Immunity 147
Huang SM, McCance DJ (2002) Down regulation of the interleukin-8 promoter by human papil-
lomavirus type 16 E6 and E7 through effects on CREB binding protein/p300 and P/CAF. J
Virol 76:8710–8721
James EA, Gates TJ, LaFond RE, Yamamoto S, Ni C, Mai D, Gersuk VH, O’Brien K, Nguyen QA,
Zeitner B, Lanteri MC, Norris PJ, Chaussabel D, Malhotra U, Kwok WW (2016) Neuroinvasive
West Nile infection elicits elevated and atypically polarized T cell responses that promote a
pathogenic outcome. PLoS Pathog 12:e1005375
Jones RB, Walker BD (2016) HIV-specific CD8+ T cells and HIV eradication. J Clin Invest
126(2):455–463
Keynan Y, Card CM, McLaren PJ, Dawood MR, Kasper K, Fowke KR (2008) The role of regula-
tory T cells in chronic and acute viral infections. Clin Infect Dis 46(7):1046–1052
Kleine-Lowinski K, Rheinwald JG, Fichorova RN, Anderson DJ, Basile J, Munger K, Daly CM,
Rosl F, Rollins BJ (2003) Selective suppression of monocyte chemoattractant protein-1 expres-
sion by human papillomavirus E6 and E7 oncoproteins in human cervical epithelial and epider-
mal cells. Int J Cancer 107:407–415
Kumar P, Sulochana P, Nirmala G, Chandrashekar R, Haridattatreya M, Satchidanandam V (2004)
Impaired T helper 1 function of non-structural protein 3-specific T cells in Japanese patients
with encephalitis with neurological sequelae. J Infect Dis 189:880–891
Laidlaw BJ, Craft JE, Kaech SM (2016) The multifaceted role of CD4+ T cells in CD8+ T cell
memory. Nat Rev Immunol 16(2):102
Lan S, Wu L, Wang X, Wu J, Lin X, Wu W et al (2016) Impact of HBeAg on the maturation and
function of dendritic cells. Int J Infect Dis 46:42e8
Lanteri MC, Diamond MS, Law JP, Chew GM, Wu S, Inglis HC, Wong D, Busch MP, Norris PJ,
Ndhlovu LC (2014) Increased frequency of Tim-3 expressing T cells is associated with symp-
tomatic West Nile virus infection. PLoS One 9:e92134
Legge KL, Braciale TJ (2003) Accelerated migration of respiratory dendritic cells to the regional
lymph nodes is limited to the early phase of pulmonary infection. Immunity 18:265–277
Li S, Labrecque S, Gauzzi MC, Cuddihy AR, Wong AH, Pellegrini S, Matlashewski GJ, Koromilas
AE (1999) The human papilloma virus (HPV)-18 E6 oncoprotein physically associates with
Tyk2 and impairs Jak-STAT activation by interferon-α. Oncogene 18:5727–5737
Mittal D, Kassianos AJ, Tran LS, Bergot AS, Gosmann C, Hofmann J, Blumenthal A, Leggatt
GR, Frazer IH (2013) Indoleamine 2,3-dioxygenase activity contributes to local immune
suppression in the skin expressing human papillomavirus oncoprotein e7. J Invest Dermatol
133:2686–2694
Mongkolsapaya J, Dejnirattisai W, Xu XN, Vasanawathana S, Tangthawornchaikul N et al (2003)
Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever. Nat Med
9:921–927
Nelson AN, Putnam N, Hauer D, Baxter VK, Adams RJ, Griffin DE (2017) Evolution of T cell
responses during measles virus infection and RNA clearance. Sci Rep 7(1):11474
Osuna CE et al (2016) Zika viral dynamics and shedding in rhesus and cynomolgus macaques. Nat
Med 22(12):1448–1455
Oswald-Richter K, Grill SM, Shariat N et al (2004) HIV infection of naturally occurring and
genetically reprogrammed human regulatory T-cells. PLoS Biol 2:E198
Pauken KE, Wherry EJ (2015) Overcoming T cell exhaustion in infection and cancer. Trends
Immunol 36(4):265–276
Perdomo-Celis F, Salvato MS (2019) T-cell response to viral hemorrhagic fevers. Vaccine 7:11
Raziorrouh B, Schraut W, Gerlach T, Nowack D, Gruner NH, Ulsenheimer A et al (2010) The
immunoregulatory role of CD244 in chronic hepatitis B infection and its inhibitory potential
on virus-specific CD8+ T-cell function. Hepatology 52:1934–1947
Rosendahl Huber S, van Beek J, de Jonge J, Luytjes W, van Baarle D (2014) T cell responses to
viral infections–opportunities for peptide vaccination. Front Immunol 5:171
Seeger C, Mason WS (2000) Hepatitis B virus biology. Microbiol Mol Biol Rev 64:51e68
Seder RA, Darrah PA, Roederer M (2008) T-cell quality in memory and protection: implications
for vaccine design. Nat Rev Immunol 8:247–258
148 A. Jagadeesh et al.
Sheiko MA, Golden-Mason L, Giugliano S, Hurtado CW, Mack CL, Narkewicz MR, Rosen HR
(2016) CD4+ and CD8+ T cell activation in children with hepatitis C. J Pediatr 170:142–148
Shrestha B, Diamond MS (2007) Fas ligand interactions contribute to CD8+ T-cell-mediated con-
trol of West Nile virus infection in the central nervous system. J Virol 81:11749–11757
Turtle L, Bali T, Buxton G, Chib S, Chan S, Soni M, Hussain M, Isenman H, Fadnis P, Venkataswamy
MM, Satishkumar V (2016) Human T cell responses to Japanese encephalitis virus in health
and disease. J Exp Med 213(7):1331–1352
Utzschneider DT, Charmoy M, Chennupati V, Pousse L, Ferreira DP, Calderon-Copete S, Danilo
M, Alfei F, Hofmann M, Wieland D, Pradervand S (2016) T cell factor 1-expressing mem-
orylike CD8+ T cells sustain the immune response to chronic viral infections. Immunity
45(2):415–427
Vande Pol SB, Klingelhutz AJ (2013) Papillomavirus E6 oncoproteins. Virology 445:115–137
Wehrens EJ, Wong K, Gupta A, Benedict C, Zuniga E (2016) IL-27 suppresses CD4 and CD8 T cell
cytotoxicity and viral control during cytomegalovirus infection. J Immunol 196(1 Suppl):217.4
Weiskopf D, Angelo MA, de Azeredo EL, Sidney J, Greenbaum JA, Fernando AN et al (2013)
Comprehensive analysis of dengue virus-specific responses supports an HLA-linked protective
role for CD8+ T cells. Proc Natl Acad Sci U S A 110(22):E2046–E2053
Zhang Y, Zhang H, Ma W, Liu K, Zhao M, Zhao Y, Lu X, Zhang F, Li X, Gao GF, Liu WJ (2018)
Evaluation of Zika virus-specific T-cell responses in immunoprivileged organs of infected
Ifnar1−/−mice. J Vis Exp (140):e58110. https://doi.org/10.3791/58110
Zhao J, Zhao J, Perlman S (2010) T cell responses are required for protection from clinical disease
and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J Virol
84(18):9318–9325
Potentiality of Toll-Like Receptors (TLRS)
in Viral Infections 10
A. M. V. N. Prathyusha, Prudhvi Lal Bhukya,
and Pallaval Veera Bramhachari
Abstract
Living organisms exist in nature usually perceptible to infectious agents like
viruses, bacteria, and many other pathogens. Immune system of host organism
typically elicits both nonspecific innate and “specific” adaptive immune
responses against foreign pathogens. Cells of innate immunity expresses a diver-
sity of pattern recognition receptors (PRRs), which were developed during evo-
lutionary process, recognizes conserved structures of distinct pathogens known
as pathogen-associated molecular patterns (PAMPs). There are several classes of
PRRs like Toll-like receptors (TLRS), RLRs, NLRs, and many DNA and RNA
sensors. Among these PRRs, Toll-like receptors plays an important role in elicit-
ing innate immunity, development of B and T cell responses and as well as
pathogen-specific adaptive immune response. Immune responses in any viral
infections are elicited by the recognition viral PAMPs like nucleic acids such as
DNA and RNA, viral capsid proteins by the host PRR. TLRs sense these viral
PAMPs and induce the antiviral response by inducing the immune active chemo-
kines and cytokines. This chapter focuses on the responses of different TLRs
with viral PAMPS, immune responses mediated by TLRS in viral infections,
agonists of TLRs for treatment of viral infections.
Keywords
Viral Infections · PAMP · PRR · TLRs · Innate immune responses
10.1 Introduction
TLR1 and TLR2 regulate innate immune response by recognizing PAMPs-like gly-
coproteins and lipoproteins to activate immune cells (Ma et al. 2015). TLR2-
dependent secretion of pro-inflammatory cytokines and cell surface molecules
which activate promotion of T cells function in non-parenchymal liver cells like
Kupffer cells (Lin et al. 2010). Preiss et al. (2008) reported that stimulation of TLR2
leads to activation of NF-κB and other cytokines like tumor necrosis factor alpha
(TNF-α) and interleukin 8 (IL-8) in MyD88-dependent manner to suppress invading
pathogens in both hepatoma cell lines and human primary hepatocytes. In an in vitro
study carried out by Thompson et al. (2009) reported that when Hepatoma cell lines
stimulated with IlL-1 receptor and TLR2, replication and capsids formation of HBV
is inhibited. Secretions of pro-inflammatory cytokines viz. IL6, IL12p40, and
TNF-α were observed in macrophages in a TLR2-dependent fashion when triggered
with full-length HBV-HBc antigen (Cooper et al. 2005). Zhang et al. (2012) reported
that TLR2 activates antiviral signalling pathways like NFKB, PI3/Akt, and MAPK
producing various pro-inflammatory cytokines in hepatoma cell lines. In a similar
study, it is observed that in HepG2.2.15 cells and primary woodchuck hepatocytes
TLR2-mediated innate immune response that leads to decreased replication and
gene expression of HBV and woodchuck hepatitis virus. Even though TLR2 inhib-
its replication and nucleocapsid formation of HBV while virus has evolved a clever
strategy to counteract antiviral function of TLR2 by using its antigens like HBeAg,
HBsAg, and by whole HBV virions to decrease expression level of TLR2, which
ultimately affects amount of inflammatory cytokines. In many other DNA and RNA
viruses, like EBV, HSV, human cytomegalovirus, HCV, RSV, and LCMV antiviral
action mediated by TLR2 was observed. The antiviral response of TLR2 was
observed in many cell types like glial cells of nervous system in LCMV virus infec-
tion, monocytes in EBV infection, microglial cells in HSV infection and leukocytes
in HSV infection (Zhou et al. 2009; Gaudreault et al. 2007; Aravalli et al. 2008;
Murawski et al. 2009). Transmembrane domain of HIV-1 envelope interacts with
152 A. M. V. N. Prathyusha et al.
(Yamamoto et al. 2004). In comparison with other TLRs, TLR4 has a unique prop-
erty as it combines with two different signalling adaptors molecules like MyD88
and TRIF activates transcription of several pro-inflammatory cytokines like IL6,
IL12, TNFα, and type 1 interferon (Yamamoto et al. 2004; Evans et al. 2003). In
case of MyD88-dependent pathway, TLR4 which is present on cells’ surface dimer-
izes with myeloid differentiation 2 proteins (MD-2), which further results in recruit-
ment of two adaptor proteins known as TIRAP and MyD88. This series of events
results in activation of pro-inflammatory transcription factors NF-κB, AP-1, IRF5,
and production of pro-inflammatory cytokines (Rosadini et al. 2015). TLR4 is
known to internalize into endosomes and recruits TRAM and TRIF adaptor proteins
and activates transcription factor like IRF3 which induces production of type I inter-
feron (Kawai and Akira 2011).
TLR5 recognizes flagellin protein as a ligand from invading motile bacterial patho-
gens and mediates immune responses by the production of cytokines. When TLR5
receptor is activated by bacterial flagellin, TLR5 activates NF-κB and further stimu-
lates TNFα production to elicit efficient immunity against invading pathogens
(Srivastava et al. 2013). Increasing numbers of recent studies have demonstrated the
efficacy of flagellin in adjuvant activity and also about bridging innate and adaptive
immune responses via TLR5. Isogawa in 2005 reported that intravenous injection of
TLR5-specific ligand flagellin from Salmonella muenchen in HBV transgenic mice
exhibited complete inhibition of HBV replication in liver non-cytopathically within
24 h in an enhanced α/β interferon dependent way (Isogawa et al. 2005). This study
states that ligand specific for TLR5 can be used as therapeutic targets for inhibiting
the replication of HBV. Zhao et al. developed a new vaccine with the adjuvant activ-
ity of fusion protein Flagellin (FliC) from Salmonella abortus equi and gD protein
from EHV-1 virus. This novel vaccine significantly induced specific antibody
responses and increased IFN-γ and IL-4 levels in host (Zhao et al. 2019).
Following this LRR domain, there are transmembrane region and cytoplasmic TIR
receptor domain. Interaction of TLR7 with its ligand-like pathogens ssRNAs results
in activation of MyD88-IRF7-dependent innate immune signalling pathways to
elicit cytokine production (Sajadi et al. 2013), promote formation of autophago-
somes which directly combat pathogen invasion (Song et al. 2018). Viruses evolved
several strategies to combat TLR7 pathway-dependent autophagy and enhance their
replication in host cells. EV71 and CA16 virus evade immune pathways by lower-
ing production of INF-1, downstream signaling molecules in TLR7 pathway this
results in reduced autophagosomes production and in turn autophagy (Song et al.
2018). This ultimately leads to successful replication of viruses in host cells.
Despite other TLRs in viral infections, TLR8 recognizes the PAMPs expressed by
infectious agents and stimulates innate immune responses for production of cyto-
kines to combat invading pathogens (Sarvestani et al. 2012). TLR8 is present on
endosomes which specifically recognizes GU and G-rich nucleotides in ssRNAs of
pathogens. TLR8 recognizes many viral genomes like HCV and HIV. TLR8 medi-
ates its antiviral action through MyD88-dependent manner (Heil et al. 2004; Zhang
et al. 2016).
NF-κB through its numerous gene products. Therefore, HSV impairs both TLR-
facilitated NF-κB pathway but also inhibits/activates NF-κB signaling through its
own gene proteins in TLR independent manner to ensure viral replication and
immune escape. Studies on HSV discovered that TLR2 and NF-κB-dependent path-
ways are harnessed for viral replication.
Association of viral outbreaks with public health concern globally, there is a dire
need for the development of safe and effective innate immune strategies to combat
viral infections. Innate immunity delivers the first line of defense against viruses,
especially TLRs, constitute critical components of innate immune pathways.
Therefore, copious viruses evolved strategies to deploy TLR signaling to escape
defensive responses of host. Agonist therapy might control viral infections during
very early stage as they block virus attachment to cell surface via TLRs or by pre-
venting TLR-mediated immune activation. Agonists are molecules that mimic the
viral pattern recognition molecules and bind to TLRs to elicit both innate and adap-
tive immune response. Furthermore, agonist viral therapy may be useful for prophy-
lactic or therapeutic treatment for viruses. Several agonists were successfully
employed in preclinical and clinical settings against different viruses. Each TLR
responds to diverse agonists, viz. bacterial and fungal components as well as viral
RNA, except for TLR10, whose agonist is unknown (Lee et al. 2006). TLR3, 4, 7,
8, and 9 agonists were used in nonhuman primate models for DENV and hepatitis
B virus (Chihab et al. 2019) and in murine models for HSV types-1 and -2. These
agonists were discovered to reduce viral symptoms and replication. Conversely,
TLR7 and TLR8 agonists were also reported to be used in treating viral infections
along with inflammatory disorders. However, the first identified TLR guanosine
analog Isatoribine has comparable chemical structural features in common with
ribonucleoside ribavirin, component of current standard for HCV care therapy.
TLR7 agonist Isatoribine may signify an important and unique approach for chronic
HCV therapy (Xiang et al. 2007). Interestingly, both TLR7 (Imidazoquinoline) and
TLR8 (R848 (resiquimod) agonists were used to treat HSV-2 induced psoriasis and
genital lesions (Mark et al. 2007; Gotovtseva et al. 2008) and related imiquimod is
used in treatment of genital warts caused by human papillomavirus.
Nonetheless, carboxy-terminal domain and amino-terminal amphipathic helix
are indispensable for antiviral activity against HCV (Helbig et al. 2011) while SAM
domain (Upadhyay et al. 2014) is obligatory for activity against TBEV and carboxy-
terminal domain for DENV, respectively (Helbig et al. 2013). Notably, Viperin is
another agonist that comprises carboxy-terminal domain, central domain with con-
served CXXCXXC motif typical for radical SAM family of enzymes (SAM domain)
and amino-terminal domain that mimics TLR ligands that acts as antiviral agents
(Vanwalscappel et al. 2018). TLR7/8 agonist R848 is deemed as the most potent
inhibitor and might be for prophylactic or therapeutic treatment of ZIKA. RNA-seq
analysis recognized several genes that were strongly induced by R848 in
156 A. M. V. N. Prathyusha et al.
TLRs represent as primary sensors of innate immune response against viral patho-
gens. Priming of TLR results in the activation of cellular signaling pathways
involved in both innate and adaptive immune responses. TLRs in mount protective
immune system against infection and their cross talk among other PRRs in associa-
tion with pathogen recognition. Triggering of TLRs 7, 8, and 9 prompts secretion of
pro-inflammatory cytokines (IFN-α, IL-12, and TNF-α) that ensues in TH1-like
immune responses, important in antiviral and tumor immunity. TLR activation by
topical administration of TLR7/8/9 agonists represents a powerful treatment modal-
ity for epithelial viral and neoplastic lesions. Currently, imiquimod represents the
only TLR agonist licensed for the treatment of humans (e.g., CA, actinic keratosis,
superficial basal cell carcinoma). Several other agonists of TLR7, 8, and 9 were
shown to be effective therapeutic agents against infections, cancer, and allergic dis-
orders, but are not yet approved for treatment in humans. Agonists of TLRs 4, 7, 8,
and 9 were also used as adjuvants for prophylactic and therapeutic vaccination.
Acknowledgments The authors are grateful to NIV Pune and Krishna University, Machilipatnam
for the support extended.
Conflict of Interest The authors declare that they have no competing interests.
10 Potentiality of Toll-Like Receptors (TLRS) in Viral Infections 157
References
Aillot L, Bonnin M, Ait-Goughoulte M, Bendriss-Vermare N, Maadadi S, Dimier L et al (2018)
Interaction between toll-like receptor 9-CpG oligodeoxynucleotides and hepatitis B virus viri-
ons leads to entry inhibition in hepatocytes and reduction of alpha interferon production by
plasmacytoid dendritic cells. Antimicrob Agents Chemother 62(4):e01741-17
Al-Anazi MR, Matou-Nasri S, Abdo AA, Sanai FM, Alkahtani S, Alarifi S et al (2017) Association
of toll-like receptor 3 single-nucleotide polymorphisms and hepatitis C virus infection. J
Immunol Res 2017:1590653
Amin OE, Colbeck E, Daffis S, Pattabiraman D, Sitali C, Rosenberg W et al (2019) In vitro modu-
lation by TLR8 agonist GS-9688 of multiple regulatory cell types in patients with chronic
hepatitis B. J Hepatol 70:E445
Aravalli RN, Hu S, Lokensgard JR (2008) Inhibition of toll-like receptor signaling in primary
murine microglia. J Neuroimmune Pharmacol 3(1):5–11
Bourquin C, Schmidt L, Lanz AL, Storch B, Wurzenberger C, Anz D et al (2009) Immunostimulatory
RNA oligonucleotides induce an effective antitumoral NK cell response through the TLR7. J
Immunol 183(10):6078–6086
Budimir N, De Haan A, Meijerhof T, Waijer S, Boon L, Gostick E et al (2013) Critical role of
TLR7 signaling in the priming of cross-protective cytotoxic T lymphocyte responses by a
whole inactivated influenza virus vaccine. PLoS One 8(5):e63163
Chang WJ, Toledo-Pereyra LH (2012) Toll-like receptor signaling in liver ischemia and reperfu-
sion. J Investig Surg 25(4):271–277
Chihab H, Zaidane I, Elhabazi A, Jadid FZ, El Fihri R, Elmessaoudi-Idrissi M, Chair M et al
(2019) Toll-like receptor 9 polymorphisms and hepatitis B virus clearance in Moroccan chronic
carriers. Gene 687:212–218
Cooper CL, Davis HL, Angel JB, Morris ML, Elfer SM, Seguin I et al (2005) CPG 7909 adjuvant
improves hepatitis B virus vaccine seroprotection in antiretroviral-treated HIV-infected adults.
AIDS 19(14):1473–1479
Dowling JK, Dellacasagrande J (2016) Toll-like receptors: ligands, cell-based models, and read-
outs for receptor action. Methods Mol Biol 1390:3–27
Evans JT, Cluff CW, Johnson DA, Lacy MJ, Persing DH, Baldridge JR (2003) Enhancement of
antigen-specific immunity via the TLR4 ligands MPL™ adjuvant and Ribi. 529. Expert Rev
Vaccines 2(2):219–229
Fischer J, Koukoulioti E, Schott E, Fülöp B, Heyne R, Berg T et al (2018) Polymorphisms in the
toll-like receptor 3 (TLR3) gene are associated with the natural course of hepatitis B virus
infection in Caucasian population. Sci Rep 8(1):12737
Gantier MP, Tong S, Behlke MA, Xu D, Phipps S, Foster PS et al (2008) TLR7 is involved in
sequence-specific sensing of single-stranded RNAs in human macrophages. J Immunol
180(4):2117–2124
Gaudreault E, Fiola S, Olivier M, Gosselin J (2007) Epstein-Barr virus induces MCP-1 secretion
by human monocytes via TLR2. J Virol 81(15):8016–8024
Gay NJ, Gangloff M (2007) Structure and function of toll receptors and their ligands. Annu Rev
Biochem 76:141–165
Gotovtseva EP, Kapadia AS, Smolensky MH, Lairson DR (2008) Optimal frequency of imiquimod
(aldara) 5% cream for the treatment of external genital warts in immunocompetent adults: a
meta-analysis. Sex Transm Dis 35(4):346–351
Hackstein H, Hagel N, Knoche A, Kranz S, Lohmeyer J, Von Wulffen W et al (2012) Skin TLR7
triggering promotes accumulation of respiratory dendritic cells and natural killer cells. PLoS
One 7(8):e43320
Hayashi T, Yao S, Crain B, Chan M, Tawatao RI, Gray C et al (2012) Treatment of autoimmune
inflammation by a TLR7 ligand regulating the innate immune system. PLoS One 7(9):e45860
Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S et al (2004) Species-specific
recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303(5663):1526–1529
158 A. M. V. N. Prathyusha et al.
Helbig KJ, Eyre NS, Yip E, Narayana S, Li K, Fiches G, McCartney EM, Jangra RK, Lemon SM,
Beard MR (2011) The antiviral protein viperin inhibits hepatitis C virus replication via interac-
tion with nonstructural protein 5A. Hepatology 54(5):1506–1517
Helbig KJ, Carr JM, Calvert JK, Wati S, Clarke JN, Eyre NS, Narayana SK, Fiches GN, McCartney
EM, Beard MR (2013) Viperin is induced following dengue virus type-2 (DENV-2) infec-
tion and has anti-viral actions requiring the C-terminal end of viperin. PLoS Negl Trop Dis
7(4):e2178
Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H et al (2000) A toll-like receptor recog-
nizes bacterial DNA. Nature 408(6813):740
Isogawa M, Robek MD, Furuichi Y, Chisari FV (2005) Toll-like receptor signaling inhibits hepati-
tis B virus replication in vivo. J Virol 79(11):7269–7272
Jahanban-Esfahlan R, Seidi K, Majidinia M, Karimian A, Yousefi B, Nabavi SM et al (2019) Toll-
like receptors as novel therapeutic targets for herpes simplex virus infection. Rev Med Virol
29(4):e2048
Kader M, Smith AP, Guiducci C, Wonderlich ER, Normolle D, Watkins SC et al (2013) Blocking
TLR7-and TLR9-mediated IFN-α production by plasmacytoid dendritic cells does not dimin-
ish immune activation in early SIV infection. PLoS Pathog 9(7):e1003530
Karimi-Googheri M, Arababadi MK (2014) TLR3 plays significant roles against hepatitis B virus.
Mol Biol Rep 41(5):3279–3286
Kawai T, Akira S (2011) Toll-like receptors and their crosstalk with other innate receptors in infec-
tion and immunity. Immunity 34(5):637–650
Krieg AM (2007) The toll of too much TLR7. Immunity 27(5):695–697
Latz E, Schoenemeyer A, Visintin A, Fitzgerald KA, Monks BG, Knetter CF et al (2004) TLR9
signals after translocating from the ER to CpG DNA in the lysosome. Nat Immunol 5(2):190
Le Goffic R, Pothlichet J, Vitour D, Fujita T, Meurs E, Chignard M et al (2007) Cutting edge: influ-
enza a virus activates TLR3-dependent inflammatory and RIG-I-dependent antiviral responses
in human lung epithelial cells. J Immunol 178(6):3368–3372
Lee HK, Dunzendorfer S, Soldau K, Tobias PS (2006) Double-stranded RNA-mediated TLR3
activation is enhanced by CD14. Immunity 24(2):153–163
Lin SC, Lo YC, Wu H (2010) Helical assembly in the MyD88–IRAK4–IRAK2 complex in TLR/
IL-1R signalling. Nature 465(7300):885
Ma Z, Zhang E, Yang D, Lu M (2015) Contribution of toll-like receptors to the control of hepa-
titis B virus infection by initiating antiviral innate responses and promoting specific adaptive
immune responses. Cell Mol Immunol 12(3):273
Mark KE, Corey L, Meng TC, Magaret AS, Huang ML, Selke S, Slade HB, Tyring SK, Warren
T, Sacks SL, Leone P, Bergland VA, Wald A (2007) Topical resiquimod 0.01% gel decreases
herpes simplex virus type 2 genital shedding: a randomized, controlled trial. J Infect Dis
195(9):1324–1331
Medvedev R (2017) Impact of oxidative stress by hepatitis C virus-mediated Nrf2 inhibition
on autophagy and viral particle release (Doctoral dissertation, Johann Wolfgang Goethe-
Universität Frankfurt am Main)
Murawski MR, Bowen GN, Cerny AM, Anderson LJ, Haynes LM, Tripp RA et al (2009)
Respiratory syncytial virus activates innate immunity through toll-like receptor 2. J Virol
83(3):1492–1500
Nicodemus CF, Berek JS (2010) TLR3 agonists as immunotherapeutic agents. Immunotherapy
2(2):137–140
Pasare C, Medzhitov R (2005) Control of B-cell responses by toll-like receptors. Nature
438(7066):364
Preiss S, Thompson A, Chen X, Rodgers S, Markovska V, Desmond P, Visvanathan K, Li K,
Locarnini S, Revill P (2008) Characterization of the innate immune signalling pathways in
hepatocyte cell lines. J Viral Hepat 15(12):888–900
Rosadini CV, Zanoni I, Odendall C, Green ER, Paczosa MK, Philip NH et al (2015) A single bac-
terial immune evasion strategy dismantles both MyD88 and TRIF signaling pathways down-
stream of TLR4. Cell Host Microbe 18(6):682–693
10 Potentiality of Toll-Like Receptors (TLRS) in Viral Infections 159
Rudd BD, Burstein E, Duckett CS, Li X, Lukacs NW (2005) Differential role for TLR3 in respira-
tory syncytial virus-induced chemokine expression. J Virol 79(6):3350–3357
Sajadi SMA, Mirzaei V, Hassanshahi G, Khorramdelazad H, Daredor HY, Hosseini SMH et al
(2013) Decreased expressions of toll-like receptor 9 and its Signaling molecules in chronic
hepatitis B virus–infected patients. Arch Pathol Lab Med 137(11):1674–1679
Sarvestani ST, Williams BR, Gantier MP (2012) Human toll-like receptor 8 can be cool too: impli-
cations for foreign RNA sensing. J Interf Cytokine Res 32(8):350–361
Sato A, Linehan MM, Iwasaki A (2006) Dual recognition of herpes simplex viruses by TLR2 and
TLR9 in dendritic cells. Proc Natl Acad Sci 103(46):17343–17348
Song J, Hu Y, Li J, Zheng H, Wang J, Guo L et al (2018) Suppression of the toll-like receptor
7-dependent type I interferon production pathway by autophagy resulting from enterovirus
71 and coxsackievirus A16 infections facilitates their replication. Arch Virol 163(1):135–144
Srivastava T, Sharma M, Yew KH, Sharma R, Duncan RS, Saleem MA et al (2013) LPS and
PAN-induced podocyte injury in an in vitro model of minimal change disease: changes in TLR
profile. J Cell Commun Signal 7(1):49–60
Takeda K, Akira S (2004) TLR signaling pathways. Semin Immunol 16(1):3–9
Thompson AJ, Colledge D, Rodgers S, Wilson R, Revill P, Desmond P et al (2009) Stimulation
of the interleukin-1 receptor and toll-like receptor 2 inhibits hepatitis B virus replication in
hepatoma cell lines in vitro. Antivir Ther 14(6):797–808
Thompson MR, Kaminski JJ, Kurt-Jones EA, Fitzgerald KA (2011) Pattern recognition receptors
and the innate immune response to viral infection. Viruses 3(6):920–940
Triantafilou K, Orthopoulos G, Vakakis E, Ahmed MAE, Golenbock DT, Lepper PM et al (2005)
Human cardiac inflammatory responses triggered by Coxsackie B viruses are mainly toll-like
receptor (TLR) 8-dependent. Cell Microbiol 7(8):1117–1126
Upadhyay AS, Vonderstein K, Pichlmair A, Stehling O, Bennett KL, Dobler G, Guo JT, Superti-
Furga G, Lill R, Overby AK, Weber F (2014) Viperin is an iron-sulfur protein that inhibits
genome synthesis of tick-borne encephalitis virus via radical SAM domain activity. Cell
Microbiol 16(6):834–848
Vanwalscappel B, Tada T, Landau NR (2018) Toll-like receptor agonist R848 blocks Zika virus
replication by inducing the antiviral protein viperin. Virology 522:199–208
Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA (2004) Toll-like receptor 3
mediates West Nile virus entry into the brain causing lethal encephalitis. Nat Med 10(12):1366
Xiang AX, Webber SE, Kerr BM, Rueden EJ, Lennox JR, Haley GJ et al (2007) Discovery of
ANA975: an oral prodrug of the TLR-7 agonist isatoribine. Nucleosides Nucleotides Nucleic
Acids 26(6–7):635–640
Yamamoto M, Takeda K, Akira S (2004) TIR domain-containing adaptors define the specificity of
TLR signaling. Mol Immunol 40(12):861–868
Zhang X, Kraft A, Broering R, Schlaak JF, Dittmer U, Lu M (2012) Preclinical development of
TLR ligands as drugs for the treatment of chronic viral infections. Expert Opin Drug Discovery
7(7):597–611
Zhang Y, El-Far M, Dupuy FP, Abdel-Hakeem MS, He Z, Procopio FA et al (2016) HCV RNA
activates APCs via TLR7/TLR8 while virus selectively stimulates macrophages without induc-
ing antiviral responses. Sci Rep 6:29447
Zhao Y, Chang J, Zhang B, Tong P, Wang C, Ran D et al (2019) TLR-5 agonist Salmonella abortus
equi flagellin FliC enhances FliC-gD-based DNA vaccination against equine herpesvirus 1
infection. Arch Virol 164(5):1371–1382
Zhou Y, Ye L, Wan Q, Zhou L, Wang X, Li J et al (2009) Activation of toll-like receptors inhibits
herpes simplex virus-1 infection of human neuronal cells. J Neurosci Res 87(13):2916–2925
Activation of Complement System
During Viral Infections: Prospects 11
and Future Challenges
Abstract
The complement system is homeostatic system evolved to remain constant check
on pathogen but as per the recent knowledge it is involved in many biological
processes including complementing adaptive immunity. It gets activated in most
of the viral infections and leads to neutralization of virus via opsonization of
C3b, aggregation, phagocytosis, membrane attack complex (MAC) mediated
lysis of virus or virus-infected cells. Primary work of complement in viral dis-
eases clears the virus by MAC or by opsonization nonetheless it offers favorable
milieu locally during localized infection. It increases vascular permeability, gen-
erates edema, recruits phagocytes by chemotaxis, mediates release of cytokines
depending on the cell type. This acute inflammation generated due to local acti-
vation of complement in initial stage of infection is crucial. C3a and C5a ana-
phylatoxins modulate adaptive immunity generation via modulating priming of
T cells and enhancing Th1 immunity. In the absence of certain complement com-
ponents and its receptors, some viruses become more pathogenic than in general,
denote complement functions at more than one step in different viruses of their
pathogenesis.
Keywords
Complement components · Complement activation · Viral infection · Pathogenesis
P. L. Bhukya (*)
Hepatitis Division, National Institute of Virology, Pune, Maharashtra, India
P. V. Bramhachari
Department of Biotechnology, Krishna University,
Machilipatnam, Andhra Pradesh, India
11.1 Introduction
Complement system is a group of soluble and cell surface proteins that starts the
series of activation cascade of proteins against the inflammatory signal. These pro-
teins are present in all vertebrates in inactive form zymogen while activated form is
serine proteases. However, a host cell does not activate complement due to presence
of complement regulatory proteins present on their surface. There are mainly three
major pathways by which complement system gets activated. These are classical
pathway, alternate pathway, lectin pathway; converge at single step of C3 conver-
tase formation, and cleavage of C3 component.
Classical pathway (CP) is antibody dependent since it needs antibody for its
activation, upon binding of IgG or IgM to the antigen C1 complex can bind to anti-
body. C1q multimeric protein possesses globular heads that bind to Fc portion of
antibody while C1r and C1s subsequently get activated and further cleaves C4 and
C2. The activation product C4bC2a forms classical pathway C3 convertase leaving
away C4a and C2b. The alternate pathway (AP) activated by tick over mechanism
of C3, that is, spontaneous hydrolysis of C3 into C3H2O further binds to foreign
surface. Factor B binds to bound C3H2O followed by factor D that cleaves factor B
into Ba and Bb, this generates alternate pathway C3 convertase C3bBb. Properdin
11 Activation of Complement System During Viral Infections: Prospects and Future… 163
Fig. 11.1 The complement system activation during viral infection, classical alternate, and lectin
pathway
C3b when cleaved into iC3b and C3dg by factor I (Atkinson et al. 2018), C3dg
binding to antigen interacts with cr2 receptor on B cells which is B cell co-receptor
and it has very strong costimulatory effect on B cells (Roozendaal and Carroll 2007)
CR2-mediated signaling helps B cells to survive in germinal center (81) it helps DC
for long retention of antigen thereby it contributes to enhanced B cell memory
(Fischer et al. 1998) C3a and C5a has a regulatory effects on B cells such as C3a
inhibits polyclonal response and C5a promotes migration of B cells to the site of
complement activation (Burg et al. 1995; Fischer and Hugli 1997; Ottonello et al.
1999).
In the absence of C3a-C3aR signaling, DCs fail to induce potent CD4 T cell
response against antigen (Sacks 2010) anaphylatoxins provide a co-stimulatory and
survival signal to T cells (Strainic et al. 2008). Anaphylatoxins’ interaction with
their respective receptors has a very important role which decides the consequence
of APC and T cell interaction, induction of Th1, Th2, Th17, and T reg cells (Strainic
et al. 2013). Especially it has been shown that complement enhances CD4 and CD8
T cell response against virus infection through anaphylatoxin receptors (Kolev et al.
2014). In addition, complement also enhances CD8 T cell immunity in lymphocytic
choriomeningitis virus infection. (Fang et al. 2007). These mechanisms composed
of direct engagement between complement proteins with CRs on T cells, while
indirect regulation via APC engagement, and alteration of cytokine profiles through
CR–TLR cross talk.
11 Activation of Complement System During Viral Infections: Prospects and Future… 165
Acknowledgments The authors are grateful to NIV, Pune and Krishna University, Machilipatnam
for the support extended.
Conflict of Interest The authors declare that they have no competing interests.
References
Agrawal P, Nawadkar R, Ojha H, Kumar J, Sahu A (2017) Complement evasion strategies of
viruses: an overview. Front Microbiol 8:1117. https://doi.org/10.3389/fmicb.2017.01117
Atkinson JP, Du Clos TW, Mold C, Kulkarni H, Hourcade D, Wu X (2018) 21 - the human comple-
ment system: basic concepts and clinical relevance. In: Clinical Immunology, 5th edn, pp 299–
317.e1. https://doi.org/10.1016/B978-0-7020-6896-6.00021-1
Burg M, Martin U, Rheinheimer C, Köhl J, Bautsch W, Böttger EC, Klos A (1995) IFN-gamma
up-regulates the human C5a receptor (CD88) in myeloblastic U937 cells and related cell lines.
J Immunol 155(9):4419–4426
Dudkina NV, Spicer BA, Reboul CF, Conroy PJ, Lukoyanova N, Elmlund H, Law RH, Ekkel SM,
Kondos SC, Goode RJ, Ramm G, Whisstock JC, Saibil HR, Dunstone MA (2016) Structure of
the poly-C9 component of the complement membrane attack complex. Nat Commun 7:10588.
https://doi.org/10.1038/ncomms10588
Dunkelberger JR, Song WC (2010) Complement and its role in innate and adaptive immune
responses. Cell Res 20(1):34–50. https://doi.org/10.1038/cr.2009.139
166 P. L. Bhukya and P. V. Bramhachari
Abstract
MicroRNAs (miRNAs) are small noncoding RNA molecules that act as central
regulators of gene expression. miRNAs have diverse functions in physiology. As
regulatory molecules, miRNAs are involved in the manifestation of various viral
diseases in humans. Viruses can take advantage of host miRNAs to facilitate their
survival and replication while, for host cells, miRNAs can serve as direct antivi-
ral entities. miRNAs modulate viral pathogenesis either by directly binding to
viral genomes or by modulating cellular antiviral responses. Circulating miR-
NAs are increasingly being recognized as an emerging class of disease biomark-
ers. These miRNAs exhibit a consistent expression profile among healthy
individuals. Another level of complexity is added by the miRNAs encoded by the
viral genomes using the host RNAi machinery. These vir-miRNAs help in the
viral infection establishment. In this chapter, we discuss in brief about what are
miRNAs, their functional role, different viral miRNAs, and cellular miRNAs
involved in the pathogenesis of classical viral infections.
Keywords
MicroRNAs · Virus · Replication · Cellular miRNA · Viral miRNA
MicroRNAs (miRNAs) are 21–23 nucleotide small noncoding RNA molecules that
post-transcriptionally regulate gene expression by binding to complementary
sequences on target mRNA transcripts (mRNAs) resulting in translational repression
D. Tiraki (*)
Department of Communicable Diseases, Interactive Research School for Health Affairs
(IRSHA), Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India
or target degradation (Girard et al. 2008; Bartel 2009). miRNAs have attracted
immense attention during the last decade as tiny regulators with profound impact on
eukaryotic gene expression (Scaria et al. 2007). During the last decade, miRNA
research has gone from discovering the existence of miRNAs in mammals to explor-
ing their therapeutic applications in numerous diseases.
miRNAs are well conserved in eukaryotic organisms and are vital and evolution-
arily ancient component of genetic regulation (Tanzer and Stadler 2004). miRNAs
can specifically bind to 3′UTRs (Grimson et al. 2007), 5′UTR (Lytle et al. 2007), or
the coding region of mRNA transcripts (Friedman et al. 2008). miRNAs have
diverse functions in physiology; ranging from cell differentiation, proliferation,
apoptosis to the endocrine system, hematopoiesis, limb morphogenesis, fat metabo-
lism, etc. (Girard et al. 2008). They display different expression profiles from tissue
to tissue, reflecting diversity in cellular phenotypes. A direct role of miRNAs in
immune response also has been demonstrated (Gantier et al. 2007). miRNAs are
generally tissue specific and cell associated, however, they have also been found in
the extracellular milieu (circulating miRNAs). Circulating miRNAs in serum and
plasma are increasingly being recognized as an emerging class of potentially useful
disease biomarkers readily detectable in the blood (Wang et al. 2009; Zhang et al.
2010a, b).
A single miRNA may control the expression of more than one target mRNAs. The
specificity required for the binding interactions is attributed to the 5′ region of
miRNA. The target interactions are restricted to a region confined to ~6–8 nt
sequence at the 5′ end of the miRNA, known as the “seed sequence” (Wee et al.
2012). This seed sequence is highly conserved among different species and change
in the sequence alters the target binding (Ameres et al. 2007). The loops formed in
the miRNA:mRNA duplexes may also have a profound effect on the efficiency of
miRNA-mediated gene regulation (Ye et al. 2008). The properties associated with
the target:miRNA interactions such as sequence, structure-associated free energy,
and evolutionary conservation are used in the current target prediction algorithms
(Doench and Sharp 2004; Martin et al. 2007).
miRNAs direct the RISC to downregulate gene expression by one of the two post-
transcriptional mechanisms; mRNA cleavage or translational repression. The degree
of miRNA: mRNA complementarity determines which mechanism is followed.
Higher degree of complementarity directs toward the Ago-catalyzed degradation of
target miRNA sequences through the mRNA cleavage mechanism. Conversely, a
central mismatch bypasses degradation and facilitates the translational repression
process.
miRNAs are often tissue specific and cell-associated, however, they are also found
in the extracellular milieu. Hence, based on their location in the body, they can be
classified as (a) cell-associated miRNAs and (b) circulating miRNAs (cell-free
miRNAs). Based on their origin, miRNAs are also classified as (a) host-encoded
miRNAs and (b) virus-encoded miRNAs.
170 D. Tiraki
All miRNAs originate in particular cells/tissues and play a vital role in the regula-
tion of cellular processes there. For example, miR-122, a liver-specific miRNA is
abundantly present in the liver and constitutes ~70% of total miRNA pool. Cell-
associated miRNAs play a role in the pathogenesis of diseases, which is attributed
to their dysregulation in the tissue of their origin. Thus, these miRNAs can act as
biomarkers or indicators of diseases. For example, miR-122 has been reported as an
indicator of liver disease progression (Girard et al. 2008), miR-208 acts as a predic-
tive marker for myocardial injury (Ji et al. 2009).
Cell-free miRNAs that are found circulating in the extracellular milieu are called as
circulating miRNAs. In 2008, Lawrie et al. first reported the presence of miRNAs in
serum (Lawrie et al. 2008). Cell-free miRNAs are not only detected in plasma and
serum, but also in body fluids such as urine, tears, breast milk, amniotic fluid, pleu-
ral fluid, cerebrospinal fluid, and saliva (Chen et al. 2008; Weber et al. 2010). Due
to their remarkable stability and ease of access, circulating miRNAs serve as an
important tool as a biomarker of various diseases (Mitchell et al. 2008). These cell-
free miRNAs act as mediators for cell–cell communication and immune regulation
(Valadi et al. 2007). Altered levels of circulating miRNAs have been reported in
different pathological conditions (Laterza et al. 2009; Mitchell et al. 2008; Mo et al.
2012; Nogales-Gadea et al. 2014; Zhang et al. 2010a, b). The origin of miRNAs is
diverse and microRNAs are released in circulation by blood cells (Hunter et al.
2008); organs of the body (Laterza et al. 2009) and tumors (Mo et al. 2012).
Circulating miRNAs are widely used as biomarkers in a variety of pathological
conditions and few of them have been summarized in Table 12.1. As biomarkers,
these miRNAs help in various ways like tracking the progression of the disease, as
a prognostic tool, and as a diagnostic tool.
This class of miRNAs includes all the miRNAs that originate in the mammalian
system through the canonical miRNA biogenesis pathways. These include both the
cell-associated and circulating miRNAs discussed above.
are 5 IFN-inducible miRNAs, miR-196,-296, -351, -431, and -448 target HCV
genome and inhibit replication (Pedersen et al. 2007), miR-23 inhibits PRRSV
replication by targeting the PRRSV RNAs (Zhang et al. 2014a, b), let 7c inhibits
H1N1 by targeting the 3′UTR of viral M1 gene (Ma et al. 2012).
2. miRNAs modulate host factors to provide a less conducive environment for virus
replication. Examples are miR-146 that is involved in the pathogenesis of
Dengue virus (Wu et al. 2013), Chikungunya (Selvamani et al. 2014), and VSV
(Hou et al. 2009), miR-23 that inhibits PRRSV replication by upregulating type
1 IFNs (Zhang et al. 2014a, b), miR-30e∗, which suppresses DENV replication
through NF-κB-dependent IFN production (Zhu et al. 2014a), miR-21 attenuates
human cytomegalovirus (HCMV) replication by targeting Cdc25a, a cell cycle
regulator (Fu et al. 2014), miR-23a facilitates HSV-1 replication by suppressing
IFN regulatory factor 1 (IRF1) (Ru et al. 2014), miR-15b modulates HBV repli-
cation by targeting HNF1α (Dai et al. 2014). Qian et al. have reported the role of
miR-122 in Borna virus-induced disease. miR-122 exerted a direct antiviral
function by translation and replication inhibition and also by indirectly acting
through IFN to increase the host immunity (Qian et al. 2010).
12.6.1.5 Hepatitis A
Using computational analysis and experimental validation, Shi et al. reported the
production of HAV-encoded miRNA on the antigenome, hav-miR-N1-3p, in
KMB17 and HEK 293 T cells (Shi et al. 2014a). Parallelly in the same year, the
same group reported identification of two more HAV-encoded miRNAs, hav-miR-
1-5p and hav-miR-2-5p using deep sequencing and in silico approaches in KMB17
174 D. Tiraki
cells (Shi et al. 2014b). Further in 2016, once again Shi et al. reported the mecha-
nism of these HAV vmiRNAs wherein they proposed that these vmiRNAs serve as
a self-mediated feedback regulator during HAV infection by attenuating the accu-
mulation of viral genomic RNAs (Shi et al. 2016).
Several cellular miRNAs are known to regulate viral pathogenesis by altering the
host mRNA/gene expression. Several other cellular miRNAs involved in the host
immune system are modulated by the viruses thereby affecting the course of infec-
tion. In the section below, we shed light upon the effect of viruses on cellular miR-
NAs and cellular miRNAs that regulate the viral life cycle in several human viral
infections.
12.6.2.1 Hepatitis B
HBV infection can cause acute or chronic hepatitis B, liver cirrhosis, and hepatocel-
lular carcinoma (Kiyosawa et al. 1990). In an attempt to identify host miRNAs
involved in HBV replication, both miRNAs inhibiting and promoting HBV replica-
tion have been identified.
12.6.2.2 Hepatitis C
HCV infection disrupts multiple pathways regulated by miRNAs, like antigen pro-
cessing, cell cycle, immune response, proteasome, and lipid metabolism (Ura et al.
2009). miRNAs act either by controlling target gene expression or by directly tar-
geting the HCV genome (Kerr et al. 2011; Pedersen et al. 2007; Peng et al. 2009).
did not lower miR-122 in HCV-infected patients (Köberle et al. 2013). Hepatic
miR-122 expression levels were significantly increased in HCV-3-infected patients
(Oliveira et al. 2016). Recently, van der Ree et al. demonstrated miravirsen dosing
decreased miR-122 levels in CHC patients (van der Ree et al. 2016). Insufficient
clinical data is available and hence more studies are required to understand the regu-
latory role of miR-122 in various states of HCV infection. Also, the results for the
clinical trials for miravirsen are awaited.
miRNAs like miR-141, -192, -491, and -215 are reported to facilitate HCV rep-
lication (Banaudha et al. 2011; Ishida et al. 2011). miR-199a-5p facilitated HCV
replication by regulating pro-survival pathways through PI3K/Akt, Ras/ERK, and
Wnt/β-catenin (Wang et al. 2015). Recently, Bandiera et al. reported facilitation of
HCV infection by upregulating miR-146a-5p in hepatocytes (Bandiera et al. 2016).
12.6.2.3 Hepatitis A
A few studies have reported the involvement of cellular miRNAs in HAV infec-
tion. Profiling and characterization of cellular miRNAs were carried out in HAV
infected human fibroblasts (Shi et al. 2016). The alteration of miRNA profile was
suggested to be related to the establishment of persistent HAV infection (Shi et al.
2016). In another study, Weseslindtner et al. have shown upregulation of miR-
106a and -197 in patients with acute viral hepatitis (caused by HAV, HBV, HCV,
and HEV). This report, however, does not differentiate between these etiologies
(Weseslindtner et al. 2016).
12.6.2.4 Hepatitis E
Cheng et al. showed downregulation of miR-221 and -222 in swine HEV-4 ORF3
expressing HEK 293 cells by regulating the p27kip1 expression (Cheng et al. 2013).
Another study by Trehanpati et al. reported for the first time, the role of miRNA
signatures in predicting ALF in HE during pregnancy. They demonstrated that the
interaction of distinct miRNAs in particular immune pathways was responsible for
the diverse outcomes of HE infection including inflammatory responses, liver fail-
ure, or death (Trehanpati et al. 2017). As mentioned in the earlier section,
12 MicroRNAs and Their Role in Viral Infection 177
Weseslindtner et al. have reported the upregulation of miR-106a, -122, and -197 in
patients with acute viral hepatitis (caused by HAV, HBV, HCV, and HEV) with no
differentiation in the different etiologies (Weseslindtner et al. 2016). Recently, posi-
tive regulation of Hepatitis E virus replication by interacting with the RdRp region
of the viral genome has been reported by Haldipur et al. (2018).
Several other miRNAs like miR-302c, -449b, -125a/b, -136, -194, -483-3p, -132,
and -26a have been shown to target cytokine production (Chen et al. 2017; Gui et al.
2015; Buggele et al. 2013; Hsu et al. 2017; Zhao et al. 2015; Wang et al. 2017;
Maemura et al. 2018; Lagos et al. 2010; Gao et al. 2017).
12.7 Conclusion
References
Ahluwalia JK et al (2008) Human cellular microRNA hsa-miR-29a interferes with viral nef protein
expression and HIV-1 replication. Retrovirology 5:117
Akamatsu S et al (2015) Differences in serum microRNA profiles in hepatitis B and C virus infec-
tion. J Infect 70(3):273–287
Ameres SL et al (2007) Molecular basis for target RNA recognition and cleavage by human
RISC. Cell 130(1):101–112
Axtell MJ et al (2011) Vive la différence: biogenesis and evolution of microRNAs in plants and
animals. Genome Biol 12(4):221
Bala S et al (2012) Circulating microRNAs in exosomes indicate hepatocyte injury and inflamma-
tion in alcoholic, drug-induced, and inflammatory liver diseases. Hepatology 56(5):1946–1957
Banaudha K et al (2011) MicroRNA silencing of tumor suppressor DLC-1 promotes efficient
hepatitis C virus replication in primary human hepatocytes. Hepatology 53(1):53–61
Bandiera S et al (2016) HCV-induced up-regulation of miR-146a-5p in hepatocytes promotes viral
infection and deregulates metabolic pathways associated with liver disease pathogenesis. J
Virol 90(14):6387–6400
Bandyopadhyay S et al (2011) Hepatitis C virus infection and hepatic stellate cell activation down-
regulate miR-29: miR-29 overexpression reduces hepatitis C viral abundance in culture. J
Infect Dis 203(12):1753–1762
Bartel DP (2009) MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233
Bernstein E et al (2001) Role for a bidentate ribonuclease in initiation step of RNA interference.
Nature 409(6818):363–366
Bhanja Chowdhury J et al (2012) Hepatitis C virus infection modulates expression of interferon
stimulatory gene IFITM1 by upregulating miR-130a. J Virol 86(18):10221–10225
Bogerd H et al (2014) Replication of many human viruses is refractory to inhibition by endog-
enous cellular microRNAs. J Virol 88(14):8065–8076
Bohnsack MT et al (2004) Exportin 5 is a RanGTP-dependent dsRNA-binding protein that medi-
ates nuclear export of pre-miRNAs. RNA 10(2):185–191
180 D. Tiraki
Buggele WA et al (2012) Influenza A virus infection of human respiratory cells induces primary
microRNA expression. J Biol Chem 287:31027–31040
Buggele WA et al (2013) Small RNA profiling of influenza A virus-infected cells identifies miR-
449b as a regulator of histone deacetylase 1 and interferon beta. PLoS One 8:e76560
Cai X et al (2004) Human microRNAs are processed from capped, polyadenylated transcripts that
can also function as mRNAs. RNA 10(12):1957–1966
Cai X et al (2006) Human papillomavirus genotype 31 does not express detectable microRNA
levels during latent or productive virus replication. J Virol 80:10890–10893
Chang J et al (2008) Liver-specific microRNA miR-122 enhances replication of hepatitis C virus
in nonhepatic cells. J Virol 82(16):8215–8223
Chen X et al (2008) Characterization of microRNAs in serum: a novel class of biomarkers for
diagnosis of cancer and or diseases. Cell Res 18(10):997–1006
Chen Y et al (2011) A liver-specific microRNA binds to a highly conserved RNA sequence of
hepatitis B virus and negatively regulates viral gene expression and replication. FASEB J
25(12):4511–4521
Chen J et al (2015) Activation of TLR2 and TLR6 by dengue NS1 protein and its implications in
the immunopathogenesis of dengue virus infection. PLoS Pathog 11:e1005053
Chen X et al (2017) MicroRNA-302a suppresses influenza A virus-stimulated interferon regula-
tory factor-5 expression and cytokine storm induction. J Biol Chem 292:21291–21303
Cheng J-C et al (2012) Let-7b is a novel regulator of hepatitis C virusreplication. Cell Mol Life
Sci 69(15):2621–2633
Cheng Y et al (2013) Identification of miR-221 and -222 as important regulators in genotype IV
swine hepatitis E virus ORF3-expressing HEK. Virus Genes 47:49–55
Chou W et al (2016) MicroRNA let-7g cooperates with interferon/ribavirin to repress hepatitis C
virus replication. J Mol Med 94(3):311–320
Corcoran DL et al (2009) Features of mammalian microRNA promoters emerge from polymerase
II chromatin immunoprecipitation data. PLoS One 4(4):e5279
Cullen BR (2006) Viruses and microRNAs. Nat Genet 38:S25–S30
Dai X et al (2014) Modulation of HBV replication by microRNA-15b through targeting hepatocyte
nuclear factor 1α. Nucleic Acids Res 42(10):6578–6590
Deng M et al (2016) Hepatitis B virus mRNAs functionally sequester let-7a and enhance hepato-
cellular carcinoma. Cancer Lett 383(1):62–72
Deng Y et al (2017a) MicroRNA-146a induction during influenza H3N2 virus infection targets and
regulates TRAF6 levels in human nasal epithelial cells (hNECs). Exp Cell Res 352:184–192
Deng W et al (2017b) MicroRNA-125b-5p mediates post-transcriptional regulation of hepatitis B
virus replication via LIN28B/let-7 axis. RNA Biol 14(10):1389–1398
Denli AM et al (2004) Processing of primary microRNAs by microprocessor complex. Nature
432(7014):231–235
Doench JG, Sharp PA (2004) Specificity of microRNA target selection in translational repression.
Genes Dev 18(5):504–511
Dong C et al (2017) Modulation of influenza A virus replication by microRNA-9 through targeting
MCPIP1. J Med Virol 89:41–48
Escalera-Cueto M et al (2015) Let-7c overexpression inhibits dengue virus replication in human
hepatoma Huh-7 cells. Virus Res 196:105–112
Fan CG et al (2011) miR-122 inhibits viral replication and cell proliferation in hepatitis B virus-
related hepatocellular carcinoma and targets NDRG3. Oncol Rep 26(5):1281–1286
Flores O et al (2013) Mutational inactivation of herpes simplex virus 1 microRNAs identifies viral
mRNA targets and reveals phenotypic effects in culture. J Virol 87(12):6589–6603
Friedman RC et al (2008) Most mammalian mRNAs are conserved targets of microRNAs. Genome
Res 19(1):92–105
Fu Y-R et al (2014) miR-21 attenuates human cytomegalovirus replication in neural cells by target-
ing Cdc25a. J Virol 89:1070–1082
Fukuhara T et al (2012) Expression of microRNA miR-122 facilitates an efficient replication in
nonhepatic cells upon infection with hepatitis C virus. J Virol 86(15):7918–7933
12 MicroRNAs and Their Role in Viral Infection 181
Jangra RK et al (2010) Regulation of hepatitis C virus translation and infectious virus production
by microRNA miR-122. J Virol 84(13):6615–6625
Ji X et al (2009) Plasma miR-208 as a biomarker of myocardial injury. Clin Chem 55(11):1944–1949
Jiao X et al (2017) Serum and exosomal miR-122 and miR-199a as a biomarker to predict thera-
peutic efficacy of hepatitis C patients. J Med Virol 89(9):1597–1605
Jin J et al (2013) MicroRNA-501 promotes HBV replication by targeting HBXIP. Biochem
Biophys Res Commun 430(4):1228–1233
Jopling CL et al (2005) Modulation of hepatitis C virus RNA abundance by a liver-specific
microRNA. Science 1:1577–1581
Jopling CL et al (2008) Position-dependent function for a tandem MicroRNA miR-122-binding
site located in hepatitis C virus RNA genome. Cell Host Microbe 4(1):77–85
Kakumani PK et al (2013) Role of RNA interference (RNAi) in dengue virus replication and iden-
tification of NS4B as an RNAi suppressor. J Virol 87:8870–8883
Kakumani PK et al (2016) Role of human GRP75 in miRNA mediated regulation of dengue virus
replication. Gene 586:7–11
Kamo Y et al (2014) Significance of micro RNA-122 in chronic hepatitis C patients with serotype
1 on interferon therapy. Hepatol Res 45(1):88–96
Kaul D et al (2009) HIV-1 genome-encoded hiv1-mir-H1 impairs cellular responses to infection.
Mol Cell Biochem 323(1–2):143–148
Kerr TA et al (2011) MicroRNAs and liver disease. Transl Res 157(4):241–252
Ketting RF et al (2001) Dicer functions in RNA interference and in synsis of small RNA involved
in developmental timing in C. elegans. Genes Dev 15(20):2654–2659
Khongnomnan K et al (2015) Human miR-3145 inhibits influenza A viruses replication by target-
ing and silencing viral PB1 gene. Exp Biol Med 240:1630–1639
Khvorova A et al (2003) Functional siRNAs and miRNAs exhibit strand bias. Cell 115(2):209–216
Kim VN et al (2009) Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol 10(2):126–139
Kiyosawa K et al (1990) Interrelationship of blood transfusion, non-a, non-B hepatitis and hepa-
tocellular carcinoma: analysis by detection of antibody to hepatitis C virus. Hepatology 12(4
Pt 1):671–675
Köberle V et al (2013) Serum microRNA-122 kinetics in patients with chronic hepatitis C virus
infection during antiviral therapy. J Viral Hepat 20(8):530–535
Kohno T et al (2014) Human microRNA hsa-miR-1231 suppresses hepatitis B virus replication by
targeting core mRNA. J Viral Hepat 21(9):e89–e97
Lagos D et al (2010) miR-132 regulates antiviral innate immunity through suppression of the p300
transcriptional co-activator. Nat Cell Biol 12:513–U232
Laterza OF et al (2009) Plasma MicroRNAs as sensitive and specific biomarkers of tissue injury.
Clin Chem 55(11):1977–1983
Lawrie CH et al (2008) Detection of elevated levels of tumour-associated microRNAs in serum of
patients with diffuse large B-cell lymphoma. Br J Haematol 141(5):672–675
Lecellier C-H et al (2005) A cellular microRNA mediates antiviral defense in human cells. Science
308(5721):557–560
Lee Y et al (2002) MicroRNA maturation: stepwise processing and subcellular localization.
EMBO J 21(17):4663–4670
Lee Y et al (2003) nuclear RNase III Drosha initiates microRNA processing. Nature
425(6956):415–419
Lee Y et al (2004) MicroRNA genes are transcribed by RNA polymerase II. EMBO J
23(20):4051–4060
Li D et al (2011) MicroRNA-99a inhibits hepatocellular carcinoma growth and correlates with
prognosis of patients with hepatocellular carcinoma. J Biol Chem 286(42):36677–36685
Lin Y et al (2017) microRNA-99 family modulates hepatitis B virus replication by promoting
IGF-1R/PI3K/Akt/mTOR/ULK1 signaling-induced autophagy. Cell Microbiol 19(5):e12709
Lytle JR et al (2007) Target mRNAs are repressed as efficiently by microRNA-binding sites in 5’
UTR as in 3’ UTR. PNAS 104(23):9667–9672
12 MicroRNAs and Their Role in Viral Infection 183
Ma YJ et al (2012) Cellular microRNA let-7c inhibits M1 protein expression of the H1N1 influ-
enza A virus in infected human lung epithelial cells. J Cell Mol Med 16:2539–2546
Machlin ES et al (2011) Masking 5′ terminal nucleotides of hepatitis C virus genome by an uncon-
ventional microRNA-target RNA complex. PNAS 108(8):3193–3198
Maemura T et al (2018) Lung-derived exosomal miR-483-3p regulates the innate immune response
to influenza virus infection. J Infect Dis 217(9):1372–1382
Martin G et al (2007) Prediction and validation of microRNA targets in animal genomes. J Biosci
32(6):1049–1052
Mitchell PS et al (2008) Circulating microRNAs as stable blood-based markers for cancer detec-
tion. PNAS 105(30):10513–10518
Mo M-H et al (2012) Cell-free circulating miRNA biomarkers in cancer. J Cancer 3:432–448
Moran J et al (2015) Circulating levels of miR-150 are associated with poorer outcomes of A/
H1N1 infection. Exp Mol Pathol 99:253–261
Mourelatos Z et al (2002) miRNPs: a novel class of ribonucleoproteins containing numerous
microRNAs. Genes Dev 16(6):720–728
Mukherjee A et al (2014) Transcriptional suppression of miR-181c by hepatitis C virus enhances
homeobox A1 expression. J Virol 88(14):7929–7940
Murakami Y et al (2009) Regulation of hepatitis C virus genome replication by miR-199a∗. J
Hepatol 50(3):453–460
Narbus CM et al (2011) HepG2 cells expressing miR-122 support entire hepatitis C virus life
cycle. J Virol 85(22):12087–12092
Nathans R et al (2009) Cellular microRNA and P bodies modulate host-HIV-1 interactions. Mol
Cell 34:696–709
Nogales-Gadea G et al (2014) Analysis of serum miRNA profiles of myasnia gravis patients. PLoS
One 9(3):1–11
Noorbakhsh F et al (2010) MicroRNA profiling reveals new aspects of HIV neurodegeneration:
caspase-6 regulates astrocyte survival. FASEB J 24:1799–1812
Oliveira KG et al (2016) Increased hepatic expression of miRNA-122 in patients infected with
HCV genotype 3. Med Microbiol Immunol 205(2):111–117
Omoto S et al (2004) HIV-1 nef suppression by virally encoded microRNA. Retrovirology 1:44
Othumpangat S et al (2017) Upregulation of miRNA- 4776 in influenza virus infected bronchial
epithelial cells is associated with downregulation of NFKBIB and increased viral survival.
Viruses 9(5):E94
Ouellet DL et al (2008) Identification of functional microRNAs released through asymmetrical
processing of HIV-1 TAR element. Nucleic Acids Res 36:2353–2365
Pang PS et al (2012) Structural map of a MicroRNA-122:hepatitis C virus complex. J Virol
86(2):1250–1254
Pedersen IM et al (2007) Interferon modulation of cellular microRNAs as an antiviral mechanism.
Nature 449(7164):919–922
Peng X et al (2009) Computational identi025ication of hepatitis C virus associated microRNA-
mRNA regulatory modules in human livers. BMC Genomics 10(1):373
Peng F et al (2016) Identification of microRNAs in throat swab as the biomarkers for diagnosis of
influenza. Int J Med Sci 13:77–84
Pfeffer S et al (2004) Identification of virusencoded microRNAs. Science 304:734–736
Pfeffer S et al (2005) Identification of microRNAs of the herpesvirus family. Nat Methods
2:269–276
Potenza N et al (2011) Human microRNA hsa-miR-125a-5p interferes with expression of hepatitis
B virus surface antigen. Nucleic Acids Res 39(12):5157–5163
Qian J et al (2010) Modulation of miR-122 on persistently Borna disease virus infected human
oligodendroglial cells. Antivir Res 87(2):249–256
Qiu L et al (2010) MiR-122-induced down-regulation of HO-1 negatively affects miR-122-
mediated suppression of HBV. Biochem Biophys Res Commun 398(4):771–777
Qiu D et al (2017) Expression of microRNA let-7a positively correlates with hepatitis B virus
replication in hepatocellular carcinoma tissues. Exp Biol Med 242(9):939–944
184 D. Tiraki
Abstract
Immunodeficiency viruses infect the host and primarily affect the immune sys-
tem of an organism, i.e., host. Till date Human Immunodeficiency Virus (HIV),
Simian Immunodeficiency Virus (SIV), Feline Immunodeficiency Virus (FIV),
Bovine Immunodeficiency Virus (BIV), and Dog Immunodeficiency Virus (DIV)
were reported in the literature. These viruses belong to phylum—Incertae sedis,
family—Retroviridae, genus—Lentivirus, and order—Ortervirales. The review
discusses about evolution, distribution, and diversity of immunodeficiency
viruses, which helps in understanding the biology of HIV and how to develop a
vaccine to the most harmful and dreadful diseases.
Keywords
Bovine immunodeficiency virus (BIV) · Dog immunodeficiency virus (DIV) ·
Feline immunodeficiency virus (FIV) · Human immunodeficiency virus (HIV) ·
Immunodeficiency virus · Simian immunodeficiency virus (SIV)
Viruses that infect the host and affect the immune system of the host upon infec-
tion are generally known as immunodeficiency viruses (IV). The host then acquires
a disease known as Acquired Immunodeficiency Disease Syndrome (AIDS).
These immunodeficiency viruses belong to phylum—Incertae sedis, family—
Retroviridae, genus—Lentivirus, and order—Ortervirales. The different types of
HIV is a retrovirus that infects human and affects the immune system of the human
upon infection (Fig. 13.1). The host then acquires a disease known as
AIDS. Worldwide statistics as of 2017 for HIV and AIDS are mentioned in
Table 13.1. In 1983, HIV called HIV1 was isolated by “… researchers at the Pasteur
Institute in France …” which was known to cause AIDS (Barré-Sinoussi et al.
1983). In 1986 “… HIV-2, was isolated from AIDS patients in West Africa …”
(Clavel et al. 1986).
Two types of HIVs exist, HIV1 and HIV2; among them, HIV1 was discovered
first and HIV2 was identified later (Fig. 13.2). HIV1 is distributed worldwide
(Fig. 13.3), whereas HIV2 is observed mainly in western Africa (Vidal et al.
2000). The reason for widespread HIV1 can due to the ancestor of HIV1, which
might have mutated at a much faster rate and traveled along with Homo sapiens
population generating diversity in HIV1 (Vidal et al. 2000; Sharp and Hahn 2008).
HIV1 group might have stemmed from strain SIVcpz of SIV (Keele et al. 2006),
whereas HIV2 group might have stemmed from strain SIVsmm of SIV (Gao et al.
1992, 1994). HIV1 is further classified into group M (main), group O (outlier),
group N (non-M/non-O), and group P. Group M is further subdivided into ten
subtypes A–K, CRF’s (Vidal et al. 2000; Sharp and Hahn 2008). Lemey et al.
(2004) classified HIV2 into groups A and B. Further, HIV2 was classified
Table 13.1 Worldwide statistics as of 2017 for HIV and AIDS (Global Statistics 2017)
S. no. Parameter Statistics
1 People living with HIV 36.9 million (31.1–43.9 million)
2 People on antiretroviral therapy 21.7 million (19.1−22.6 million)
3 People newly infected with HIV 1.8 million (1.4–2.4 million)
4 People died with HIV infection 940,000 (670,000–1.3 million)
5 People living with HIV since epidemic 77.3 million (59.9–100 million)
6 People died with HIV infection since 35.4 million (25.0–49.9 million)
epidemic
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 189
Fig. 13.1 (a) HIV infects humans and causes disease known as AIDS, (b) SIV infects nonhuman
primates and causes disease known as SAIDS, (c) FIV infects cats and causes disease known as
FAIDS, and (d) BIV infects cattle and causes disease known as BAIDS
Fig. 13.2 Classification of HIV into two major types HIV1 and HIV2
HIV1 advances into the host cells using two molecules of HIV envelope (glycopro-
tein—gp120 and gp41). This ingress of glycoproteins stimulates intracellular signal
cascades and facilitates replication of virus. The glycoproteins gp120 and gp41
form spikes on the surface of virion’s. The protein gp120 binds to the CD4+ receptor
and attaches to the host cell membrane. A marked decrease in CD4+ T cells (both
activated and memory) is the characteristic feature of infection and disease. The
virus then interacts with receptors (CCR5, CXCR4) leading to structural changes of
the protein. The viral and the cellular membranes of the host are fused and form a
viral pore. This allows the transfer of the viral core into the cytoplasm of the host.
After the core disassembles, the RNA is reverse transcribed into DNA by the virus’
with the help of enzyme reverse transcriptase. The DNA is then integrated into the
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 191
Fig. 13.3 Worldwide representation of HIV1 and its subtypes across the continents (Source:
Castro-Nallar et al. (2012))
gene rich and transcriptionally active domains of the host’s genome with the help of
the viral protein integrase, DNA repair enzymes of the host and LEDGF/p75 (lens
epithelium-derived growth factor—integrase binding host factor). Once the host
cell is transformed into a potential virus producer, transcription of viral genetic
material occurs. Later viral proteins are transported and assembled at the cell mem-
brane with the help of vesicular sorting pathway (ESCRT-I, -II, and -III). TSG101
domain, a short sequence motif in p6 of Gag I is used for protein sorting. Cleavage
of the Gag-Pol polyprotein by the viral protease produces mature infectious virions
…” (Naif 2013).
Table 13.2 Major types of SIVs and their corresponding nonhuman primate hosts infected with
SIV
S. Nonhuman primate S. Nonhuman primate
no. SIV type infected with SIV no. SIV type infected with SIV
1 SIVcpz Chimpanzee 19 SIVsun Sun-tailed monkey
2 SIVgor Gorillas 20 SIVprg Preuss’s guenon
3 SIVagm African green 21 SIVwrc Western red
monkeys colobus
4 SIVsmm Sooty mangabeys 22 SIVolc Olive colobus
5 SIVrcm Red-capped 23 SIVkrc Kibale red colobus
mangabeys
6 SIVgsn Greater spot-nosed 24 SIVtrc Tshuapa red
monkeys colobus
7 SIVmus Mustached guenons 25 SIVsyk Sykes’ monkey
8 SIVmon Mona monkeys 26 SIVdeb De Brazza’s
monkey
9 SIVagi Agile mangabey 27 SIVden Dent’s mona
monkey
10 SIVmnd 2 Mandrill 28 SIVwol Wolf’s mona
monkey
11 SIVdrl Drill monkey 29 SIVgsn/SIVmon/ Northern talapoin
SIVmus 1/SIVmus 2
clade, SIVtal
12 SIVmac Rhesus macaque 30 SIVasc Red-tailed guenon
13 SIVmne Pig-tailed macaque 31 SIVbkm Black mangabey
14 SIVstm Stump-tailed 32 SIVreg [formerly Red-eared guenon
macaque SIVery]
15 SIVsab, Grivet monkey 33 SIVblu Blue monkey
SIVver,
SIVgri
16 SIVtan Tantalus monkey 34 SIVcol Colobus guereza
17 SIVmnd 1 Mandrill 35 SIVkcol 1 Black-and-white
colobus
18 SIVlho L’hoest’s monkey 36 SIVkcol 2, SIVblc Black colobus
(formerly SIVbcm)
et al. 2010). Primate’s infection with SIV dated back to 14 Ma, if the virus and host
were coevolved then it dates to 85 Ma (Compton et al. 2013). The “non-human
primates” include species of gorilla, chimpanzees, and monkeys. The species of
gorilla are Gorilla gorilla (western lowland gorillas), and Gorilla beringei (eastern
Grauer’s gorillas). The species of western gorilla are subdivided into Gorilla gorilla
diehli (Cross River gorilla) and Gorilla gorilla gorilla (western lowland gorilla).
The eastern species are classified into Gorilla beringei graueri (Grauer’s gorilla),
Gorilla beringei beringei (mountain gorilla), and Gorilla beringei subspecies
(Bwindi gorilla). The chimpanzee species include Pan troglodytes verus (western
Africa), Pan troglodytes ellioti (Nigerian), Pan troglodytes troglodytes (central), and
Pan troglodytes schweinfurthii (eastern). The monkey species include African green
monkeys, sooty mangabeys, Cercocebus torquatus (red-capped mangabeys),
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 193
SIV infects CD4+ T cells and the SIV-infected cells undergo apoptosis within 1 day
of infection. The immune system of simians is not able to replace the cells at the
same pace leading to the deterioration of immune function. Subsequently, the host
acquires immunodeficiency syndrome leaving the animal susceptible to fatigue and
life-threatening coinfections (Fig. 13.4a). “… The interaction of SIV with the host’s
immune system triggers innate immune responses followed by virus-specific adap-
tive cellular and humoral immune responses. Rapidly occurring mutations lead to
immune evasion. Chronic immune activation contributes to the functional impair-
ment of the immune system. As the disease progresses, adaptive immune responses
are unsuccessful in containing the virus replication, and overt signs of a chronic
immune suppression become evident …” (Schmitz and Korioth-Schmitz 2013).
SIV infection leads to changes in mucosal tissues (cervicovaginal, gastrointestinal,
and penile tissues) and all organ systems (brain, lung, and heart) (Haase 2011). The
severity of the infection has an impact and functionally impairs the organs (Alammar
et al. 2011; Kelly et al. 2012). Successful infection of SIV also results in loss of
CD4+ T cells. SIV uses glycoproteins to bind CD4+ receptors of T cells and interacts
with co-receptor CXCR4 leading to conformational changes of the protein. “… The
viral and the cellular membranes of the host are fused allowing the transfer of the
SIV genome into the cytoplasm of the host. The RNA is reverse transcribed into
DNA by the virus with the help of enzyme reverse transcriptase. Then this DNA is
integrated into the host cell’s genome, transforming the host cell into a potential
virus producer. The virus then uses host cell’s replication machinery to transcribe its
DNA back into RNA. The copies of RNA are packed into virus particles of about
80–100 nm in diameter and bud off or free from the host cell infecting more cells.
SIV infections are non-pathogenic in their natural African simian primates.
However, if the virus infects an Asian or Indian rhesus macaque, these non-African
simian primates will develop simian AIDS (SAIDS). SIVs nef gene down-regulates
expression of CD3, CD4, and MHC class I and therefore do not induce immunode-
ficiency. Whereas, nef gene in HIV-1 lost its ability to down-regulate CD3, which
results in the immune activation and apoptosis …” (Swigut et al. 2004).
194 H. S. Vemuri et al.
Fig. 13.4 (a) Consequences and immunopathogenesis in simian when infected with a simian
immunodeficiency virus (SIV) (Source: Schmitz and Korioth-Schmitz (2013)). (b) Replication of
SIV in the host simian (Source: Encyclopaedia Britannica Inc. https://www.britannica.com/sci-
ence/SIV)
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 195
Feline Immunodeficiency Virus (FIV) infects cats and affects the immune system of
the cats upon infection (Fig. 13.1). The cat then acquires a disease known as Feline
Acquired Immunodeficiency Disease Syndrome (FAIDS). FIV was first isolated
from a cat exhibiting an immunodeficiency-like syndrome (Pedersen et al. 1987).
FIV is endemic in Felidae species (Biek et al. 2003, 2006; Brown et al. 1994;
Carpenter et al. 1996, 1998; Carpenter and O’Brien 1995; Driciru et al. 2006;
Hofmann-Lehmann et al. 1996; Munson et al. 2004; Olmsted et al. 1992; Troyer
et al. 2004; Troyer et al., 2005), from free-ranging lions (Pecon-Slattery et al. 2004)
to domestic cats (Olmsted et al. 1989). FIV occurs in Felis catus (domestic cats),
Puma concolor (pumas) (Langley et al. 1994), Panthera leo (lions), Otocolobus
manul (pallas cat) (Barr et al. 1997), Panthera pardus (puma leopard), Acinonyx
jubatus (cheetah), Leopardus pardalis (ocelot), Panthera onca (jaguar), Leopardus
weidii (margay), Leopardus tigrina (tigrina), large African lions, and Crocuta cro-
cuta (spotted hyaena) (Troyer et al. 2005). The different types of FIVs like FIVPle,
FIVFca, FIVPco, FIVOma, FIVLpa, FIVAju, FIVCcr, FIVPon, FIVLwe, and FIVLti were
reported in literature (Fig. 13.5). The major types of FIVs and their corresponding
felines infected with FIV are listed in Table 13.3.
Currently, isolates of FIV are classified into six subtypes (A, B, C, D, E, and F)
(Weaver 2010). Weaver (2010) performed a detailed phylogenetic analysis of FIV
and classified FIV into six subtypes (A, B, C, D, E, and F) (Fig. 13.4). Sodora et al.
(1994) initially classified FIV into three subtypes (A, B, and C). Sodora et al. (1994)
used phylogenetic analysis to classify FIV isolates based on FIV env (envelope
sequence). The geographical location of the subtype A is California and Europe;
subtype B is Japan and the central and eastern USA; and Subtype C is southwestern
Canada. Nishimura et al. (1998), later classified FIV into five subtypes (A, B, C, D,
Table 13.3 Major types of S. no. FIV type Felines infected with FIV
FIVs and their corresponding 1 FIVPle Panthera leo (lions)
felines infected with FIV
2 FIVFca Felis catus (domestic cats)
3 FIVPco Puma concolor (pumas)
4 FIVOma Otocolobus manul (pallas cat)
5 FIVPpa Panthera pardus (puma leopard)
6 FIVAju Acinonyx jubatus (cheetah)
7 FIVCcr Crocuta crocuta (spotted hyaena)
8 FIVLpa Leopardus pardalis (ocelot)
9 FIVPon Panthera onca (jaguar)
10 FIVLwe Leopardus weidii (margay)
11 FIVLti Leopardus tigrina (tigrina)
Fig. 13.6 Worldwide distribution of FIV and its subtypes across the continents (Source: Teixeira
et al. (2012))
and E) based on FIV env (envelope sequence). The geographical location of the
subtype A is California and Northern Europe; subtype B is central and eastern USA
and southern European countries; subtype C is California and British Columbia;
subtype D is Japan; and subtype E is Argentina. FIVPle has diverged into six sub-
types A–F, each with distinct geographic areas of endemicity (Brown et al. 1994;
Troyer et al. 2004; O’Brien et al. 2006). FIVFca has diverged into five subtypes, A–E
(Sodora et al. 1994; Kakinuma et al. 1995; Pecoraro et al. 1996), where subtypes A,
B, and C are widespread worldwide. Subtype D is found in Japan and Vietnam
(Kakinuma et al. 1995; Nakamura et al. 2003), whereas subtype E is only found in
Argentina (Pecon-Slattery et al. 2008). The four subtypes A–D are found in cat
populations from Japan (Nishimura et al. 1998; Kakinuma et al. 1995). FIVPco has
diverged into three subtypes A, B, and C (Pecon-Slattery et al. 2008) (Fig. 13.6).
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 197
FIV uses receptor CD9 (Poeschla and Looney 1998) for entry and then infects CD4+
T (Brown et al. 1991), FIV can also infect astroglial cells, CD8+ T lymphocytes,
macrophages, and kidney cells (Pedersen et al. 1989; Brunner and Pedersen 1989;
Phillips et al. 1990; Zenger et al. 1995). “… This leads to a significant drop in cells
which have critical roles in the immune system. Low levels of CD4+ and other
affected immune system cells cause the cat to be susceptible to opportunistic dis-
eases once the disease progresses to feline acquired immune deficiency syndrome
(FAIDS) …” (Bendinelli et al. 1995). Symptoms of immunodeficiency associated
with FIV are chronic lesions, opportunistic infections, neurological abnormalities,
and weight loss (Yamamoto et al. 2010).
CD134 is mostly present on T cells which are activated and binds to OX40
ligand, causing T cell activation, stimulation, proliferation, and apoptosis. The virus
enters the host’s cells using the glycoprotein env and interacts with surface recep-
tors. The glycoprotein SU binds to receptor CD134 of the host cell leading to con-
formational changes of protein SU. These changes facilitate interaction between SU
and the chemokine receptor CXCR4; and fuses viral membrane and cellular mem-
brane of the host (Hu 2012). “… This allows the transfer of the viral RNA into the
cytoplasm of the host. Then RNA is reverse transcribed and integrated into the
genome of the host through non-homologous recombination. Once viral RNA is
integrated into the host genome, the virus can be dormant in the asymptomatic stage
without being detected by the immune system of the host or can cause lysis of the
host cell …” (Lecollinet and Richardson 2008; Hartmann 2011).
Bovine Immunodeficiency Virus (BIV) infects cattle and affects the immune system
of the cattle upon infection (Fig. 13.1). The cattle then acquires a disease known as
Bovine Acquired Immunodeficiency Disease Syndrome (BAIDS). BIV strain “…
R-29 was originally isolated from an 8-year-old dairy cow suspected of having
bovine lymphosarcoma …” (Van Der Maaten et al. 1972a).
BIV is widespread in dairy and beef cattle in Australia (Forman et al. 1992), Canada
(McNab et al. 1994), France (Polack et al. 1996), Germany (Muluneh 1994), Italy
(Cavirani et al. 1998), Japan (Hirai et al. 1996; Meas et al. 1998), Korea (Cho et al.
1999), Netherlands (Horzinek et al. 1991a), New Zealand (Horner 1991), the UK
(Clayton 1994), the USA (Amborski et al. 1989; Cockerell et al. 1992; St. Cyr Coats
et al. 1994), and buffaloes in Pakistan (Meas et al. 2000). The prevalence of BIV in
dairy cattle is higher than beef cattle (Amborski et al. 1989; Cho et al. 1999). The
different BIV strains reported in literature are BIV R-29 (isolated from cow) (Van
198 H. S. Vemuri et al.
Der Maaten et al. 1972b), BIV-106, and BIV-127 (variants of BIV R-29) (Braun
et al. 1988; Garvey et al. 1990), BIV CR1 (BIV strain from Costa Rica (Hidalgo
et al. 1995), BIV FL112 (Hirari et al. 1996), and FL491 (Suarez et al. 1993) (strain
from Florida, USA).
BIV infects nondividing host cells and uses two phases—entry phase (first phase)
and replication phase (second phase) to replicate itself (Berkowitz et al. 2001). In
the first phase, glycoprotein of the virus envelope interacts and binds with the spe-
cific receptor of the cell. “… Then virus uses either of the ways, receptor mediated
endocytosis or viral envelope-cell membrane fusion to enter into the host cell. The
virus then dissembles in the cell and reverse-transcribes the RNA genome into DNA
with the help of enzyme reverse transcriptase (Gonda, 1992). The activity of reverse
transcriptase is higher at lower concentrations of Mn2+ ions when compared to Mg2+
ions (Horzinek et al. 1991b). The DNA is then transported to nucleus and is inte-
grated into the genome of the host cell. In the second phase, the integrated viral
DNA is transcribed and the transcript (viral mRNA) is transported and translated in
the cytoplasm ….” The translated viral structural proteins are then assembled into
virus particles and form a viral complex (virus buds) along with the viral RNA at the
plasma membrane. Then viral proteases process the viral buds and are released from
the cell in the mature stages, which are ready to infect another cell (Gonda 1992).
Dog Immunodeficiency Virus (DIV) is a retrovirus which infects dog and affects the
immune system of the dogs upon infection. The dog then acquires a disease known
as DAIDS. Safran et al. (1992) isolated canine immunodeficiency virus, (CIV)—
(canine lentivirus) from a leukemic dog. The ultrastructure and morphogenesis of
CIV is strikingly similar to that displayed by other lentiviruses, while the immuno-
logical relatedness close to any other lentivirus or oncovirus was not established.
These findings suggest that this canine retrovirus, fits in the subfamily lentivirus and
not related to other known members. Additional, investigations on DIV (CIV) are
essential to establish the biology, immunopathogenesis of virus; and how immuno-
deficiency is acquired by dogs.
Immunodeficiency viruses infect the host and primarily affect the immune system
of an organism i.e. host. HIV, SIV, FIV, BIV, and DIV were known to infect humans,
simians, cats, cattle, and dogs respectively. The ultrastructure and morphogenesis of
IVs like HIV, SIV, FIV, BIV, and DIV are strikingly similar. At the same time
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 199
immunological relatedness of the lentiviruses like HIV, SIV, FIV and BIV are close
with exception in DIV. Therefore, there is a need to establish the immunological
relatedness of the DIV. The similarities in the ultrastructure, morphogenesis and
immunological relatedness of lentivirus provide us with an opportunity for better
understanding of the immunodeficiency in different hosts. This information further
can be used to develop a vaccine to the most harmful and dreadful disease.
Acknowledgements HSV, SC, and NRRN are grateful to GITAM (Deemed to be University) for
providing necessary facilities to carry out the research work and for extending constant support.
Conflict of Interest Statement The authors declare that there is no potential con-
flict of interest.
References
Alammar L, Gama L, Clements JE (2011) Simian immunodeficiency virus infection in the
brain and lung leads to differential type I IFN signaling during acute infection. J Immunol
186:4008–4018
Amborski GF, Lo J, Seger CL (1989) Serological detection of multiple retroviral infections in
cattle: bovine leukemia virus, bovine syncytial virus and bovine visna virus. Vet Microbiol
20:247–253
Barr MC, Zou L, Long F, Hoose WA, Avery RJ (1997) Proviral organization and sequence analysis
of feline immunodeficiency virus isolated from a Pallas’ cat. Virology 228:84–91
Barré-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, Gruest J, Dauguet C, Axler-
Blin C, Vézinet-Brun F, Rouzioux C, Rozenbaum W, Montagnier L (1983) Isolation of a
T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome
(AIDS). Science 220:868–871
Bendinelli M, Pistello M, Lombardi S, Poli A, Garzelli C, Matteucci D, Ceccherini-Nelli L,
Malvaldi G, Tozzini F (1995) Feline immunodeficiency virus: an interesting model for AIDS
studies and an important cat pathogen. Clin Microbiol Rev 8:87–112
Berkowitz R, Ilves H, Lin WY, Eckert K, Coward A, Tamaki S, Veres G, Plavec I (2001)
Construction and molecular analysis of gene transfer systems derived from bovine immunode-
ficiency virus. J Virol 75(7):3371–3382
Biek R, Rodrigo AG, Holley D, Drummond A, Anderson CR Jr, Ross HA, Poss M (2003)
Epidemiology, genetic diversity, and evolution of endemic feline immunodeficiency virus in a
population of wild cougars. J Virol 77:9578–9589
Biek R, Ruth TK, Murphy KM, Anderson CR Jr, Johnson M, DeSimone R, Gray R, Hornocker
MG, Gillin CM, Poss M (2006) Factors associated with pathogen seroprevalence and infection
in Rocky Mountain cougars. J Wildl Dis 42:606–615
Braun MJ, Lahn S, Boyd AL, Kost TA, Nagashima K, Gonda MA (1988) Molecular cloning of
biologically active proviruses of bovine immunodeficiency-like virus. Virology 167:515–523
Brown EW, Yuhki N, Packer C, O’Brien SJ (1994) A lion lentivirus related to feline immunodefi-
ciency virus: epidemiologic and phylogenetic aspects. J Virol 68:5953–5968
200 H. S. Vemuri et al.
Brown WC, Bissey L, Logan KS, Pedersen NC, Elder JH, Collisson EW (1991) Feline immunode-
ficiency virus infects both CD4+ and CD8+ T lymphocytes. J Virol 65:3359–3364
Brunner D, Pedersen NC (1989) Infection of peritoneal macrophages in vitro and in vivo with
feline immunodeficiency virus. J Virol 63:5483–5488
Carpenter MA, Brown EW, Culver M, Johnson WE, Pecon-Slattery J, Brousset D, O’Brien SJ
(1996) Genetic and phylogenetic divergence of feline immunodeficiency virus in the puma
(Puma concolor). J Virol 70:6682–6693
Carpenter MA, Brown EW, MacDonald DW, O’Brien SJ (1998) Phylogeographic patterns of
feline immunodeficiency virus genetic diversity in the domestic cat. Virology 251:234–243
Carpenter MA, O’Brien SJ (1995) Coadaptation and immunodeficiency virus: lessons from the
Felidae. Curr Opin Genet Dev 5:739–745
Castro-Nallar E, Perez-Losada M, Burton GF, Crandall KA (2012) The evolution of HIV: infer-
ences using phylogenetics. Mol Phylogenet Evol 62:777–792
Cavirani S, Donofrio G, Chiocco D, Foni E, Martelli P, Allegri G, Cabassi CS, De Iaco B, Flammini
CF (1998) Seroprevalence to bovine immunodeficiency virus and lack of association with leu-
kocyte counts in Italian dairy cattle. Prev Vet Med 37:147–157
Cho KO, Meas S, Park NY, Kim YK, Endoh D, Lee SI, Ohashi K, Sugimoto C, Onuma M (1999)
Seroprevalence of bovine immunodeficiency virus in dairy and beef cattle herds in Korea. J Vet
Med Sci 61:549–551
Clavel F, Guetard D, Brun-Vezinet F, Chamaret S, Rey MA, Santos-Ferreira MO, Laurent
AG, Dauguet C, Katlama C, Rouzioux C, Klatzmann D, Champalimaud JL, Montagnier L
(1986) Isolation of a new human retrovirus from West African patients with AIDS. Science
233:343–346
Clayton J (1994) Spectre of AIDS haunts reports of sick cows. Nat (Lond) 367:585
Cockerell GL, Jensen WA, Rovnak J, Ennis WH, Gonda MA (1992) Seroprevalence of bovine
immunodeficiency-like virus and bovine leukemia virus in a dairy cattle herd. Vet Microbiol
31:109–116
Compton AA, Malik HS, Emerman M (2013) Host gene evolution traces the evolutionary history
of ancient primate lentiviruses. Philos Trans R Soc Lond Ser B Biol Sci 68(1626):20120496.
https://doi.org/10.1098/rstb.2012.0496
Damond F, Worobey M, Campa P, Farfara I, Colin G, Matheron S, Brun-Vézinet F, Robertson DL,
Simon F (2004) Identification of a highly divergent HIV type 2 and proposal for a change in
HIV type 2 classification. AIDS Res Hum Retrovir 20(6):666–672
Daniel MD, Letvin NL, King NW, Kannagi M, Sehgal PK, Hunt RD, Kanki PJ, Essex M,
Desrosiers RC (1985) Isolation of T-cell tropic HTLV-III-like retrovirus from macaques.
Science 228(4704):1201–1204
Driciru M, Siefert L, Prager KC, Dubovi E, Sande R, Princee F, Friday T, Munson L (2006) A
serosurvey of viral infections in lions (Panthera leo), from Queen Elizabeth National Park,
Uganda. J Wildl Dis 42:667–671
Forman AJ, Gibson CA, Rodwell BJ (1992) Serological evidence for the presence of bovine lenti-
virus infection in Australia. Aust Vet J 69:337
Gao F, Yue L, Robertson DL, Hill SC, Hui H, Biggar RJ, Neequaye AE, Whelan TM, Ho DD,
Shaw GM (1994) Genetic diversity of human immunodeficiency virus type 2: evidence for
distinct sequence subtypes with differences in virus biology. J Virol 68(11):7433–7447
Gao F, Yue L, White AT, Pappas PG, Barchue J, Hanson AP, Greene BM, Sharp PM, Shaw GM,
Hahn BH (1992) Human infection by genetically diverse SIVSM-related HIV-2 in west Africa.
Nature 358(6386):495–499
Garvey KJ, Obsrste MS, Esler JE, Braun MJ, Gonda MA (1990) Nucleotide sequence and genome
organization of biologically active proviruses of the bovine immunodeficiency-like virus.
Virology 175:391–409
Gilbert MTP, Rambaut A, Wlasiuk G, Spira TJ, Pitchenik AE, Worobey M (2007) The emergence
of HIV/ AIDS in the Americas and beyond. Proc Natl Acad Sci U S A 104:18566–18570
Global Statistics (2017). https://www.hiv.gov/hiv-basics/overview/data-and-trends/global-statis-
tics. Accessed 09 July 2017
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 201
McNab WB, Jacobs RM, Smith HE (1994) A serological survey for bovine immunodeficiency-
like virus in Ontario dairy cattle and associations between test results, production records and
management practices. Can J Vet Res 58:36–41
Meas S, Kabeya H, Ohashi K, Matsuki S, Mikami Y, Sugimoto C, Onuma M (1998) Seroprevalence
and field isolation of bovine immunodeficiency virus. J Vet Med Sci 60:1195–1202
Meas S, Seto J, Sugimoto C, Bakhsh M, Riaz M, Sato T, Neem K, Ohashi K, Onuma M (2000)
Infection of bovine immunodeficiency virus and bovine leukemia virus in water buffalo and
cattle populations in Pakistan. J Vet Med Sci 62:329–331
Muluneh A (1994) Seroprevalence of bovine immunodeficiency-virus (BIV) antibodies in the
cattle population in Germany. J Veterinary Med Ser B 41:679–684
Munson L, Marker L, Dubovi E, Spencer JA, Evermann JF, O’Brien SJ (2004) Serosurvey of viral
infections in free-ranging Namibian cheetahs (Acinonyx jubatus). J Wildl Dis 40:23–31
Naif HM (2013) Pathogenesis of HIV infection. Infect Dis Rep 5(Suppl 1):e6. https://doi.
org/10.4081/idr.2013.s1.e6
Nakamura K, Suzuki Y, Ikeo K, Ikeda Y, Sato E, Nguyen NTP, Gojobori T, Mikami T, Miyazawa T
(2003) Phylogenetic analysis of Vietnamese isolates of feline immunodeficiency virus: genetic
diversity of subtype C. Arch Virol 148:783–791
Nishimura Y, Goto Y, Pang H, Endo Y, Mizuno T, Momoi Y, Watari T, Tsujimoto H, Hasegawa
A (1998) Genetic heterogeneity of env gene of feline immunodeficiency virus obtained from
multiple districts in Japan. Virus Res 57:101–112
O’Brien SJ, Troyer JL, Roelke M, Marker L, Pecon-Slattery J (2006) Plagues and adaptation: les-
sons from the Felidae models for SARS and AIDS. Biol Conserv 131:255–267
Olmsted RA, Hirsch VM, Purcell RH, Johnson PR (1989) Nucleotide sequence analysis of feline
immunodeficiency virus: genome organization and relationship to other lentiviruses. Proc Natl
Acad Sci U S A 86:8088–8092
Olmsted RA, Langley R, Roelke ME, Goeken RM, Adger-Johnson D, Goff JP, Albert JP, Packer
C, Laurenson MK, Caro TM, Scheepers L, Wildt DE, Bush M, Martenson J, O’Brien SJ (1992)
Worldwide prevalence of lentivirus infection in wild feline species: epidemiologic and phylo-
genetic aspects. J Virol 66:6008–6018
Pecon-Slattery J, Pearks Wilkerson AJ, Murphy WJ, O’Brien SJ (2004) Phylogenetic assessment
of introns and SINEs within the Y chromosome using the cat family felidae as a species tree.
Mol Biol Evol 21(12):2299–2309
Pecon-Slattery J, Troyer JL, Johnson WE, O’Brien SJ (2008) Evolution of feline immunodefi-
ciency virus in Felidae: implications for human health and wildlife ecology. Vet Immunol
Immunopathol 123(1–2):32–44
Pecoraro MR, Tomonaga K, Miyazawa T, Kawaguchi Y, Sugita S, Tohya Y, Kai C, Etcheverrigaray
ME, Mikami T (1996) Genetic diversity of argentine isolates of feline immunodeficiency virus.
J Gen Virol 77:2031–2035
Pedersen NC, Ho EW, Brown ML, Yamamoto JK (1987) Isolation of a T-lymphotropic virus from
domestic cats with an immunodeficiency-like syndrome. Science 235:790–793
Pedersen NC, Yamamoto JK, Ishida T, Hansen H (1989) Feline immunodeficiency virus infection.
Vet Immunol Immunopathol 21:111–129
Pérez-Losada M, Jobes DV, Sinangil F, Crandall KA, Posada D, Berman PW (2010) Phylodynamics
of HIV-1 from a phase-III AIDS vaccine trial in North America. Mol Biol Evol 27:417–425
Phillips TR, Talbott RL, Lamont C, Muir S, Lovelace K, Elder JH (1990) Comparison of two host
cell range variants of feline immunodeficiency virus. J Virol 64:4605–4613
Plantier J-C, Leoz M, Dickerson JE, De Oliveira F, Cordonnier F, Lemee V, Damond F, Robertson
DL, Simon F (2009) A new human immunodeficiency virus derived from gorillas. Nat Med
15:871–872
Poeschla EM, Looney DJ (1998) CXCR4 is required by a nonprimate lentivirus: heterologous
expression of feline immunodeficiency virus in human, rodent, and feline cells. J Virol
72:6858–6866
13 Evolution, Distribution, and Diversity of Immunodeficiency Viruses 203
Abstract
Hepatitis B virus infection is a common cause of liver cirrhosis and hepatocel-
lular carcinoma leading to about one million deaths annually. The disease patho-
genesis is complex and incompletely understood, especially when it turns
chronic. Among various factors known to be responsible for the disease progres-
sion, the immune system has the most important role to play. Viral clearance is
largely T cells dependent, and progression to a chronic state is due to insufficient
T cells response, partly due to T-regulatory cells. In chronic infections, functions
of important immune cells like natural killer cells and dendritic cells are impaired,
which enables the viral persistence and promotes the pathogenesis in the host
body. Similarly, the virus is reported to modulate functions of monocytes, mac-
rophages, and Kupffer cells to promote an immune-tolerant local environment in
the liver. Though B cells are expected to be central to the humoral immune
response that clears the virus, its role in chronic HBV is still obscure. This chap-
ter would elaborate on the significant roles of immune cells in pathogenesis and
in clearance of the hepatitis B virus.
Keywords
Hepatitis B virus · Innate immunity · Adaptive immunity · T cells ·
Immunopathogenesis
Hepatitis B virus infection is the most common cause of liver pathologies like cir-
rhosis and hepatocellular carcinoma leading to about one million deaths annu-
ally and around 257 million people are chronically infected globally (WHO report
2017). Most adults clear an acute viral infection without any symptoms, but 5–10%
of them become chronic patients due to persisting presence of virus, while most
neonates born to mothers with an active infection develop chronic hepatitis B infec-
tion. The infection could lead to a range of pathologies depending on the virus and
host immune system interplay, from acute self-contained infection to chronic hepa-
titis, leading to cirrhosis, hepatocellular carcinoma (HCC), and death of the patient.
Most acute infections are mild and asymptomatic with only a small proportion
developing fatigue, nausea, jaundice, and rarely, acute liver failure. In chronic infec-
tions, the disease manifests itself to variable degree in each patient depending on
various factors. Some patients remain asymptomatic or have nonspecific symptoms;
some show symptoms similar to that in acute infection, while more severe cases
have a more morbid prognosis with a 50% 5-year survival rate (Liang 2009).
Current treatment includes pegylated-interferon (IFN)-α and five nucleos(t)ide
analogs (NAs). Though each of these drugs show some benefit in controlling the
virus and symptoms, they do not completely clear the virus. Antiviral resistance and
side effects limit usage of these drugs. Hence, therapy is advised for only those who
are likely to develop serious complications (Koumbi 2015).
Fig. 14.2 Schematic representation of the Hepatitis B virus life cyce (Mohd-Ismail et al. 2019)
208 P. Sinha and P. Sahu
HBV reaches high titers in the blood and this is the major route of transmission. On
infection there is minimal immune response and hence the virus spreads rapidly in
the hepatocytes. Immune activation within the first few weeks leads to inflammation
in the liver and antiviral response. Cytotoxic T cells are the major cells responsible
for killing infected hepatocytes as well as arresting viral replication by secretion of
antiviral cytokines like INF-γ and TNF-α. Extensive cell death in the liver may
result in some symptoms, indicative of liver inflammation and damage. Appearance
of antibodies against the viral surface antigen (HBsAb) in serum is a major indicator
of clearance of the virus from the circulation.
As mentioned earlier, 5–10% of adults and most HBV-infected infants are unable
to fully clear the virus and develop a chronic HBV infection. Insufficient or
exhausted T cell response is believed to be the primary reason for this persistent
infection. Low levels of viral DNA and particles are detected beyond 6 months of
infection and this is accompanied by occasional flares of viral–immune activity.
This continued cycle of inflammation and cell damage, over the years, leads to
development of liver cirrhosis and hepatocellular carcinoma in many patients. The
status of the disease in a chronic patient can be categorized as immune-tolerant,
immune-active, or immune-inactive carrier phase, depending upon the viral activity
and host immune response (Trepo et al. 2014).
The course of the disease and its outcome is variable in chronic HBV patients
making it tricky to manage. Hence, extensive studies have been done to fully under-
stand the immune pathogenesis of HBV infection and identify possible targets to
help manage the infection. The narrow host range of HBV makes it challenging to
study. Most of the understanding of the HBV life cycle comes from animal models,
i.e., chimpanzees and Tupaia Belangeri (Tree shrew) which can be infected by HBV
as well as closely related viruses like Duck-HBV and Woodchuck-HBV in their
respective host. Many studies are also done on transgenic and humanized mice.
Viral biology is also studied in vitro, on either primary human hepatocytes or NTCP
expressing transgenic cell lines. Major gaps still exist in the understanding of the
pathogenesis and involvement of immune cells in HBV infection (Lamontagne
et al. 2016).
14.4 T Cells
T lymphocytes are the major effector cells involved in adaptive immune response to
viruses. A robust T cell response clears the infection in acutely infected HBV
patients, while weak and inadequate T cell response is believed to result in chronic
HBV infection. In a mouse model of HBV infection, the results strongly show that
CD4+ T cells (T helper cells) serve as major regulators of the adaptive immune
response and the CD8+ T cells (cytotoxic T cells or CTLs) being the main effector
cells for HBV clearance along with supporting functions of NK cells and interfer-
ons (Yang et al. 2010).
14 Role of Immune Cells in Hepatitis B Infection 209
CD4+ T helper cells are involved in early priming and development of CD8+ cyto-
toxic T cell function as well as B cell function of production of antibodies to clear
free viral particles (Sant and McMichael 2012). Th1 CD4+ cells have been found to
increase during hepatic inflammatory activity and in active phases of chronic HBV
infection (Jiang et al. 2002), but not during peak viremia; rather they are required
early in the infection for priming of CD8+ T cells for successfully clearing the infec-
tion. This was supported by another report (Fisicaro et al. 2009) where they found
that CD4+ and CD8+ T cell responses are found to peak, early in the infection, with
IFN-γ producing CD8+ cells found at higher frequencies at the height of immune
response. Hence, even though CD4+ T cells may not be direct effector cells for viral
clearance, they play an important role by early induction of CD8+ T cells. The CD4+
T cells also exert their immunological function by producing cytokines like
Interferon (IFN)-γ and Tumor Necrosis Factor (TNF)-α to control the infection and
regulate other immune cells (Li et al. 2016a). Chronic HBV infection is character-
ized by T cell exhaustion and inadequacy of CD4+ T cells function may be another
contributing factor to the effector CD8+ T cell exhaustion in chronic viral infections
(Ye et al. 2015). Indeed, inhibitory molecule PD-1 (programmed cell death protein
1) has been found to be upregulated in CD4+ T cells in chronic HBV infection
(Raziorrouh et al. 2014), hence this could be a contributing factor for the dysfunc-
tion of the T cell compartment.
Early reports show that strong HBV-specific CD8+ T cell response correlates with
viral clearance during acute infection. The highest frequency of peripheral HBV-
specific CD8+ T cells are found in the acute phase of HBV infection; however, a
transient attenuation of the CD8+ proliferation (Maini et al. 1999) and functions,
coinciding with increase in the immunosuppressive cytokine, IL-10 during peak
viremia is noted (Dunn et al. 2009). An effective CD8+ response is robust, poly-
clonal and multi-antigen specific, while such a response is undetectable in chronic
patients (Rehermann et al. 1995; Stelma et al. 2017). These are the main effector
cells that clear the HBV infection by both cytolytic and non-cytolytic mechanisms,
as observed, in chimpanzees (Thimme et al. 2003).
The non-cytolytic mechanism of viral inhibition, mediated by IFN-γ and TNF-α,
inhibits viral replication and clears viral intermediates from infected cells (Guidotti
et al. 1996) without liver damage (Maini et al. 2000) and is the predominant mode
of viral clearance in an effective response, while significant cytolytic activity is
observed by nonspecific but activated lymphoid cells (Phillips et al. 2010).
The cytotoxic T cell response (CTL) is weaker and less specific in chronic HBV
infections with dampening of not only HBV-specific but global T cells functions
(Park et al. 2015). The HBV-specific CD8+ T cells show dysfunction correlating
with viral levels and they can be detected in patients with lower viremia while rarely
210 P. Sinha and P. Sahu
found in patients with higher viral load. HBV-specific CTLs, specific for the core
antigen (HBcAg), is associated with viral control, while those specific for envelop
and polymerase epitopes are more frequent in higher viremia patients, implying that
the core specific CD8+ cells are therapeutically more relevant (Webster et al. 2004).
Also, HBV mutational escape and evolution is observed during progression of the
chronic infection and is believed to be due to the selection pressure from HBV core
specific CTLs (Zhang et al. 2018); however, complete understanding of viral escape
from CD8+ immune response is still incomplete (Lumley et al. 2018).
The virus-specific CD8+ cells found in circulation of chronic patients show an
exhausted phenotype with impaired proliferation, cytokine secretion, expression of
inhibitory molecules like PD-1 (Boni et al. 2007), cytotoxic T lymphocyte antigen-4
(CTLA-4) (Schurich et al. 2011), T cell immunoglobulin domain and mucin domain-
containing molecule-3 (Tim-3) (Nebbia et al. 2012; Wu et al. 2012) and CD244
(Raziorrouh et al. 2010), which lead to their deletion on interacting with their respec-
tive ligands. This exhausted phenotype is a stable form of dysfunction and cannot be
reversed by changing the immune environment (Wang et al. 2018). However, their
functionality could be improved in vitro, by blocking these inhibitory molecules and
receptors highlighting important immunotherapeutic targets in chronic HBV infec-
tion. Blocking of PD-1 with antibodies has especially shown promise by increasing
IFN-γ producing CTLs in the liver in mice (Maier et al. 2007), and improved viral
control in combination with nucleoside analog and DNA vaccine therapy in wood-
chuks models (Liu et al. 2014b). Therapeutic efficacy in woodchuck model showed
no significant liver injury in acute models and good viral control in infected animals
expressing PD-1 on CD8+ T cells; however, combination treatment with additional
antiviral agents was recommended to achieve a robust and more consistent response
for complete immune clearance (Balsitis et al. 2018). Nivolumab, a PD-1 blocking
monoclonal antibody has been approved for therapeutic use in many forms of cancer
and has recently been approved in the USA for hepatocellular carcinoma caused by
various afflictions including HBV infection (Highleyman 2017).
Successful antiviral treatment with pegylated-IFN-α-2b is accompanied with
increase in number of circulating HBV-core or envelop-specific CD8+ T cells, along
with increased Th1-type cytokines (IL-12, TNF-α, IFN-γ) (Chen et al. 2010). T
cells with TCRs (T cell Receptor) which have high avidity for HBV antigens hold
lot of potential for adoptive transfer and treatment, and such TCRs have also been
cloned and studied in vitro for possible applications in therapy (Wisskirchen et al.
2017). Adoptive transfer of HBV-specific CD4+ and CD8+ T cells along with immu-
notherapies to prevent T cell exhaustion has been proposed for treatment of chronic
viral infection as well as cancer (Kamphorst and Ahmed 2013).
Regulatory T cells or Tregs are a set of naturally occurring and inducible CD4+
CD25+ and FoxP3 expressing T cells with a basic role of controlling hyperactivity
of effector T cells. Even though their main role is to prevent autoimmune reactions
14 Role of Immune Cells in Hepatitis B Infection 211
The T-helper-17 cells are a newly identified lineage of the CD4+ T helper cells that
predominantly produce IL-17, IL-21, IL-22 and play an important role in protection
against extracellular pathogens as well as in inflammation and autoimmunity
(Harrington et al. 2005; Park et al. 2005; Bettelli et al. 2007; Ouyang et al. 2008).
The Th17 cells in circulation, as well as intrahepatic, increase with disease progres-
sion and correlate with viral load, serum ALT, and histopathology index and this
may be through activation of mDCs, monocytes, and increased production of pro-
inflammatory cytokines in chronic patients (Zhang et al. 2010a). IL-17(a) activates
hepatic stellate cells to express fibrogenic factors thereby leading to fibrosis and
cirrhosis in an inflammatory environment in the liver (Tan et al. 2013), and this is
suggested to be a major mechanism of cirrhosis in severe cases of chronic HBV
infection (Sun et al. 2012). TH17 and IL-17 levels are found to be higher in non-
survivors and decrease in circulation of recovering patients (Yang et al. 2013).
Th17 and Treg cells play opposite roles of mediating and restraining inflamma-
tion and a lower Treg/Th17 ratio indicates extensive liver injury and fibrosis in
chronic HBV patients (Li et al. 2012). The serum IL-17 levels and frequency of
Th17 cells correlate with ALT levels while nTregs correlate positively with HBV
DNA load and inversely with Th17 frequencies (Yang et al. 2016). TGF-β/IL-17
ratio have also been found to markedly increase in decompensated cirrhosis patients
and non-survivors, and hence both these ratios can help in monitoring and prognosis
of HBV-associated cirrhotic patients (Yu et al. 2014). On administration of NA anti-
viral therapy, the Treg/Th17 ratio declined in complete responders due to increased
Th17 cells and IL-17 and decreased Treg and their cytokines, correlating with
decrease in HBV DNA and ALT levels (Yu et al. 2013). Cell-based therapy using
autologous bone marrow mesenchymal stem cells, post antiviral therapy in HBV-
associated cirrhotic patients, showed improved liver function and an increase in the
Treg/Th17 ratio (Xu et al. 2014a).
with a peptide from HBcAg and coculture with autologous T cells, showed increase
in percentage of induced CTLs (Wu et al. 2010).
The B cells do not seem to be activated in chronic HBV patients as seen by the
proportion of CD86+ B cells in their blood (Zhao et al. 2015). However, effective
treatment can activate the B cell subsets. The B cell subsets remodeling, from naïve
and post-germinal to transitional and plasmablasts strongly correlated with
decrease in serum HBsAg, as observed in PEG-IFNα treated patients (Aspord et al.
2016). In chronic HBV patients, an exhausted or defective subset of B cells has
also been recently characterized. It has been found that HBsAg-specific B cells
persist in circulation; however, these show an atypical Memory B cell phenotype
(atMBC) which is a functionally defective subset enhanced in conditions of con-
tinuous antigen exposure. These cells had enhanced expression of inhibitory mol-
ecules like PD-1, were prone to apoptosis, and perhaps diminished ability to
differentiate to antibody producing plasma cells and attenuated signaling and cyto-
kine production. These at MBC were found to preferentially accumulate in the
infected liver and it is possible that the highly inflammatory microenvironment
promotes the development of the defective phenotype. PD-1 blockade was found to
partially rescue the premature apoptosis and impaired antibody and cytokine pro-
duction (Burton et al. 2018). Similar subset of impaired B cells was also defined in
another study, where these cells from chronic patients were found to express inhib-
itory receptors of FcRL family and downregulate genes responsible for antigen
presentation. Treatment of B cells from healthy subjects, with HBV core particles,
resulted in expression of the inhibitory molecules and this altered phenotype, high-
lighting another immunosuppressive effect of HBV particles on immune cells
(Poonia et al. 2018).
A regulatory subset of B cells which predominantly produces IL-10 was also
identified. These were increased in patients, and their frequency and IL-10 levels
correlated temporally with liver inflammation. These cells were found to suppress
HBV-specific CD8+ T cell response in a IL-10 dependent manner and hence play an
important role in controlling liver inflammation or in establishment of a tolerogenic
environment (Das et al. 2012). This subset was further defined as Bregs and found
to be elevated in immune-active chronic HBV patients, which correlated with the
serum IL-10, ALT, and AST levels. Also, Bregs and IL-10 levels were higher in
inactive carrier group than healthy controls. The expression of TLR2 is elevated and
TLR9 is reduced on Bregs of chronic HBV patients compared to healthy controls;
however, its significance is yet to be elucidated (Wang et al. 2017).
Macrophages, Kupffer cells, dendritic cells, and hepatocytes would be few of the
first cells to recognize and respond to HBV, through their Toll-like receptors (Ma
et al. 2018). Monocytes mediate host antimicrobial defense by migrating to breached
tissues in response to chemokines and depending upon interaction with the
214 P. Sinha and P. Sahu
pathogen, differentiate into macrophages and dendritic cells, along with secretion of
immune modulators. Many viruses are known to preferentially infect monocytes,
express viral proteins in the cell, and trigger their rapid differentiation to dendritic
cells (Hou et al. 2012). Early reports claim that HBV DNA is found in monocytes
of chronically infected patients (Yoffe et al. 1986; Guang 1992). As found from
in vitro experiments, recombinant HBsAg binds preferentially to B cells and mono-
cytes (Pontisso et al. 1991) especially by interacting and binding to CD14 receptor
(Vanlandschoot et al. 2002). Monocytes from chronic HBV patients have been
found to be the only antigen presenting cells to contain intracellular HBsAg, which
when differentiated ex vivo, was able to cross present and stimulate expansion of
HBV-specific T cells (Gehring et al. 2013). However, literature regarding the effect
of such interaction of monocytes with different subviral particles of HBV has been
ambiguous.
HBcAg induces production of pro-inflammatory cytokines IL-6, IL-12, and
TNF-α in monocytic cell line THP-1, by signaling through TLR2 (Cooper et al.
2005). However, there is a reduction of surface expression TLR2 accompanied by
reduction of secretion of TNF-α on interaction with HBeAg (Riordan et al. 2006;
Visvanathan et al. 2007) and IL-12 on interaction with HBsAg with monocytes
(Wang et al. 2013). TLR8 is another important virus-sensing receptor expressed
strongly on monocytes but found to be reduced in monocytes from chronic HBV
patients with a concomitant reduction in production of antiviral cytokines, also cor-
relating with reduced response to interferon therapy (Deng et al. 2017). Also, while
some in vitro studies report an immunosuppressive response of the monocytes on
interaction with HbsAg, where in they secrete IL-10 and TNF-α and suppress func-
tions of pDCs (Shi et al. 2012), other similar study observe a robust secretion of
inflammatory cytokines IL-6 and TNF by monocytes on treatment with HBsAg,
though the serum level of these cytokines in chronic HBV patients did not correlate
serum HBV and HBsAg levels (Boltjes et al. 2014). Hence, more studies are
required to delineate the exact effect of the monocyte–virus interaction and role of
these cells in the disease pathogenesis.
Pro-inflammatory CD16+ subset of monocytes have also been implicated to con-
tribute to liver injury in immune-active patients since these increase in circulation
and in liver of these patients, correlating with their serum ALT and extent of liver
damage (Zhang et al. 2011). Kupffer cells (KCs), the liver tissue resident macro-
phages are also expected to have a significant role in the liver microenvironment
even though there is sparse information about their role in HBV immunity or patho-
genesis. Intrahepatic KCs from chronic HBV patients have been found to be HBsAg
positive and more activated than controls (Boltjes et al. 2015). In a mouse model of
HBV infection, KCs were found to secrete immunosuppressive IL-10 which could
be induced by viral particles to favor viral persistence (Xu et al. 2014a). On the
other hand, KCs and lymphoid cells from chronic patients are found to express
Fas-L (Tang et al. 2003) while HBsAg sensitizes liver cells to Fas mediated apopto-
sis. Hence, KCs may play in important role in clearance of infected cells, but at the
same time, cause extensive cell death and injury in an infected liver.
14 Role of Immune Cells in Hepatitis B Infection 215
The dendritic cells (DCs) are classical antigen presenting cells and their role is
important for antigen presentation and activation of T cells. Two major subtypes of
DCs are plasmacytoid DC (pDC) and myeloid DC (mDC) based on their lineage of
origin. These differ by expression of certain cell surface markers, with pDCs having
a predominant role in antiviral response by producing type I interferons (Kadowaki
2009).
HBV DNA and RNA has been reported to be found in DCs enriched and cultured
from chronic HBV patient’s blood and these DCs showed impaired functioning like
reduced ability to stimulate T cells and reduced secretion of IL-12 (Arima et al.
2003). mDCs show reduction in maturation and secretion of TNF-α while pDCs
produce less interferon-α in in vitro experiments (van der Molen et al. 2004). DCs
generated from patient monocytes in vitro (moDCs) show impaired T cell stimula-
tory function and lower IL-12 secretion, which could be restored with antiviral
treatment (Beckebaum et al. 2003). Similar functional impairment along with the
presence of viral DNA and viral particles within the cells was observed in pDCs
from chronic patients (Beckebaum et al. 2002). Even though DCs take up the viral
particles, they do not support the viral replication or subsequent steps within the
cells (Untergasser et al. 2006). These cells reduce in circulation and enrich in the
liver of immune-active chronic HBV patients (Zhang et al. 2007) while antiviral
treatment (with adefovir) that reduces the viral load shows some restoration of func-
tion as well as number of circulating DCs in the circulation, concomitant with
decreased inflammation in the liver (Van der Molen et al. 2006).
The functional impairment of DCs on interaction with viral particles is expected
to contribute to the inadequate immune response and persistence of virus in chronic
HBV infection. Reviving DC function in chronic infections would be one of the
ways to overcome the tolerogenic environment in chronic infections. It has been
observed in one study that monocytes retained a depot of HBsAg and on differenti-
ating them into DCs in vitro, they were able to crosspresent the antigen to T cells
(Gehring et al. 2013). Hence an idea of using in vitro activated autologous DCs as
vaccine in chronic patients has been explored with some success in achieving
immune clearance in these patients (Chen et al. 2005; Luo et al. 2010). On treating
MoDCs from patient’s PBMCs, with HBV sub-viral particles like HBsAg and
HBcAg, they were found to have strong capacity to stimulate T cell proliferation
and produce IL-12, showing some potential for autologous therapy for chronic
HBV patients (Shi et al. 2007). Hence, MoDCs have been considered to help boost
HBV-specific T cell response in chronic HBV patients with hepatocellular carci-
noma (HCC).
Studies have also been done to understand the interaction of the virus with DCs
and mechanism of the dysfunction. Dendritic cells would be one of the first cells to
sense the virus through its Toll-like receptor (Ma et al. 2018). It has been found that
HBV can downregulate the expression of TL9 and the subsequent IFN-α production
from pDCs (Vincent et al. 2011) which is the case in chronic HBV patients (Xu
et al. 2012). Targeting activation of Toll-like receptors, specially use of TLR7 ligand
216 P. Sinha and P. Sahu
GS-9620, to elicit an antiviral response is an attractive idea and has been tested with
some success in Woodchuk HBV model (Menne et al. 2011) and chimpanzee model
(Lanford et al. 2013); however, it was not as effective in patients (Janssen et al.
2018). Another approach was the use of nanoparticles containing HBV-CpG and
HBsAg which would activate TLR9, and was used with success in mouse in elicit-
ing an enhanced CD4+ T cell response (Lv et al. 2014). TLR 4 and 9 agonists are
already used as adjuvants for HBV vaccines while agonists for TLR 3, 7, and 8
show potential and need to be further investigated for use in therapeutics (Ma et al.
2018).
Natural killer (NK) cells are from the lymphoid lineage, and are involved mainly in
innate immune response against intracellular pathogens and abnormal cells. They
exert their antiviral activity by recognition and killing of infected cells and secretion
of antiviral cytokines like IFN-γ and TNF-α. They express CD56 and can be catego-
rized as two subsets, i.e., CD56bright, which have a predominant function of antiviral
cytokine secretion and CD56dim, which are more involved in killing target cells (Wu
et al. 2015). NK cells also promote release of antiviral cytokines like IFN-α and
IL-6 from pDCs. IFN-α in turn upregulates cyotoxic activity of NK cells and IL-6
promotes B cell differentiation (Della Chiesa et al. 2006). NK cells are the most
abundant innate immune cells in the liver (Faure-Dupuy et al. 2017) and are one of
the first cells to respond to HBV infection in patients which would allow a timely
adaptive immune response to be induced (Fisicaro et al. 2009).
Natural killer cells have been found to increase in number post-peak viremia in
acute HBV infection, where in a transient reduction of NK cells and T cell responses
coincided with a surge in IL-10 immunosuppressive cytokine (Dunn et al. 2009).
After peak viremia, an increase in TRAIL (TNF-related apoptosis-inducing
ligand)-expressing CD56bright NK cells is observed and is believed to be responsible
for clearance of the infected cells (Stelma et al. 2017) while these are also respon-
sible for elimination of HBV-specific CD8+ T cells (Peppa et al. 2013) and CD4+ T
cells (Ghosh et al. 2016) which show an exhausted phenotype and express death
receptors in chronic infections. Patients who clear an acute infection have fully
functional and activated NK cells which correlate with serum ALT levels and
inversely with serum viral load, while NK cells from chronic HBV patients pro-
duce IL-10 elicited by HBV-induced monocytes (Li et al. 2017), have diminished
IFN-γ secretion but enhanced cytolytic activity (Zhao et al. 2012), correlating with
liver histopathology and serum ALT levels especially in patients in the immune-
active phase (Zhang et al. 2010a, b). This skew in the NK cell function, correlated
with serum viral load and antiviral therapy partially restored normal NK cell func-
tioning and interferon production by these cells (Tjwa et al. 2011; Stelma et al.
2015).
Major liver damage is caused by the increased cytolytic activity of NK cells in
chronic HBV infection. Hence, it has been proposed to block NKG2D receptor of
14 Role of Immune Cells in Hepatitis B Infection 217
NK cells to alleviate the severe liver damage caused by these cells (Huang et al.
2013). PD-1/PD-L1 blockade is another popular potential strategy which would
restore IFN-γ production of NK cells, along with alleviating T cell exhaustion.
Further study to understand NK cell function in HBV infection will reveal more
therapeutic targets for treatment of HBV infection.
14.9 Neutrophils
Neutrophils are the most abundant immune cells which actively eliminate patho-
gens and target cells. However, over-activation can lead to extensive tissue damage,
as happens in hepatitis (Xu et al. 2014b). Reports of direct interaction of the virus
and neutrophils is scant, but these cells have a more significant role in the secondary
effects of the viral infection. Though one study noted the immunosuppressive effect
of recombinant HBeAg by suppression of respiratory burst in neutrophils, as well as
their chemotactic functions in vitro (Leu et al. 2014), most studies are focused
toward its role in the pathogenesis of injury in hepatitis.
In a mouse model of HBV infection, it was observed that transferring HBV-
specific cytotoxic T cells resulted in recruitment of nonspecific inflammatory cells
that caused liver pathology. In this setting, depletion of neutrophils reduced the
recruitment of antigen nonspecific inflammatory cells, hence ameliorating the
severity of liver damage, but did not affect antiviral activity of CTLs. It can be
inferred that the neutrophils have a role in the pathogenesis of the disease and may
serve as a target for immunotherapeutic approaches (Sitia et al. 2002). Concurrently,
blocking a serine protease like neutrophil elastase with an inhibitor, reduced serum
ALT levels, and the migration of inflammatory cells in the liver, post injection of
antigen-specific CTLs in a HBV mouse model, and hence administration of neutro-
phil elastase inhibitors may hold therapeutic potential against severe hepatitis (Takai
et al. 2005). The recruitment of neutrophils can be due to the expression of CXCL-8
by CTLs which recruits and elicits cytotoxic activity of neutrophils (Gehring et al.
2011) by interacting with the CXCR1 and CXCR2 on neutrophils (Nasser et al.
2009). Hence another approach could be to block CXCR1/CXCR2, which was
found to be increased in neutrophils in HBV-related acute-on-chronic liver failure
(ACLF) patients. Blocking CXCR1/CXCR2 had significantly reduced cell death in
in vitro experiments with hepatic cell lines (Khanam et al. 2017).
The neutrophil-to-lymphocyte ratio (NLR) has been widely accepted as reflec-
tion of inflammatory status of the patient before treatment and has been used in the
context of hepatitis B infection to predict acute-on-chronic liver failure in patients.
Low NLR indicates better prognosis, while higher NLR indicates a high short-term
mortality rate (Liu et al. 2014a). The NLR has been found to be a useful predictive
marker in patients with HBV-related decompensated cirrhosis (Zhang et al. 2016),
ACLF patients treated with artificial liver support system (Fan et al. 2017), in
patients receiving PEG interferon treatment (Le et al. 2017), and may be applied to
many other similar conditions.
218 P. Sinha and P. Sahu
Conflict of Interest The authors declare that they have no competing interests.
References
Aalaei-Andabili SH, Alavian SM (2012) Regulatory T cells are the most important determinant
factor of hepatitis B infection prognosis: a systematic review and meta-analysis. Vaccine
30(38):5595–5602
Alberti A, Diana S, Sculard GH, Eddleston AL, Williams R (1978) Detection of a new antibody
system reacting with Dane particles in hepatitis B virus infection. Br Med J 2(6144):1056–1058
Arima S, Akbar SM, Michitaka K, Horiike N, Nuriya H, Kohara M, Onji M (2003) Impaired func-
tion of antigen-presenting dendritic cells in patients with chronic hepatitis B: localization of
HBV DNA and HBV RNA in blood DC by in situ hybridization. Int J Mol Med 11(2):169–174
Aspord C, Bruder Costa J, Jacob MC, Dufeu-Duchesne T, Bertucci I, Pouget N, Brevot-Lutton
O, Zoulim F, Bourliere M, Plumas J, Leroy V, ANRS HB06 PEGAN study group (2016)
Remodelling of B-cell subsets in blood during pegylated IFNα-2a therapy in patients with
chronic hepatitis B infection. PLoS One 11(6):e0156200
Balsitis S, Gali V, Mason PJ, Chaniewski S, Levine SM, Wichroski MJ, Feulner M, Song Y,
Granaldi K, Loy JK, Thompson CM, Lesniak JA, Brockus C, Kishnani N, Menne S, Cockett
MI, Iyer R, Mason SW et al (2018) Safety and efficacy of anti-PD-L1 therapy in the wood-
chuck model of HBV infection. PLoS One 13(2):e0190058. https://doi.org/10.1371/journal.
pone.0190058
Beckebaum S, Cicinnati VR, Dworacki G, Muller-Berghau J, Stolz D, Harnaha J, Whiteside TL,
Thomson AW, Lu L, Fung JJ, Bonham AA (2002) Reduction in the circulating pDC1/pDC2
ratio and impaired function of ex vivo-generated DC1 in chronic hepatitis B infection. Clin
Immunol 104(2):138–150
Beckebaum S, Cicinnati VR, Zhang X, Ferencik S, Frilling A, Grosse-Wilde H, Broelsch CE,
Gerken G (2003) Hepatitis B virus-induced defect of monocyte-derived dendritic cells leads to
impaired T helper type 1 response in vitro: mechanisms for viral immune escape. Immunology
109(4):487–495
Belkaid Y (2007) Regulatory T cells and infection: a dangerous necessity. Nat Rev Immunol
7(11):875–888
220 P. Sinha and P. Sahu
Bertoletti A, Le Bert N (2018) Immunotherapy for chronic hepatitis B virus infection. Gut Liver
12(5):497–507
Bettelli E, Korn T, Kuchroo VK (2007) Th17: the third member of the effector T cell trilogy. Curr
Opin Immunol 19(6):652–657
Boltjes A, Groothuismink ZM, van Oord GW, Janssen HL, Woltman AM, Boonstra A (2014)
Monocytes from chronic HBV patients react in vitro to HBsAg and TLR by producing cyto-
kines irrespective of stage of disease. PLoS One 9(5):e97006
Boltjes A, van Montfoort N, Biesta PJ, Op den Brouw ML, Kwekkeboom J, van der Laan LJ,
Janssen HL, Boonstra A, Woltman AM (2015) Kupffer cells interact with hepatitis B surface
antigen in vivo and in vitro, leading to proinflammatory cytokine production and natural killer
cell function. J Infect Dis 211(8):1268–1278
Boni C, Fisicaro P, Valdatta C, Amadei B, Di Vincenzo P, Giuberti T, Laccabue D, Zerbini A,
Cavalli A, Missale G, Bertoletti A, Ferrari C (2007) Characterization of hepatitis B virus
(HBV)-specific T-cell dysfunction in chronic HBV infection. J Virol 81(8):4215–4225
Burton AR, Pallett LJ, McCoy LE, Suveizdyte K, Amin OE, Swadling L, Alberts E, Davidson BR,
Kennedy PT, Gill US, Mauri C, Blair PA, Pelletier N et al (2018) Circulating and intrahepatic
antiviral B cells are defective in hepatitis B. J Clin Invest 128(10):4588–4603
Cao T, Lazdina U, Desombere I, Vanlandschoot P, Milich DR, Sällberg M, Leroux-Roels G (2001)
Hepatitis B virus core antigen binds and activates naive human B cells in vivo: studies with a
human PBL-NOD/SCID mouse model. J Virol 75(14):6359–6366
Chen M, Li YG, Zhang DZ, Wang ZY, Zeng WQ, Shi XF, Guo Y, Guo SH et al (2005) Therapeutic
effect of autologous dendritic cell vaccine on patients with chronic hepatitis B: a clinical study.
World J Gastroenterol 11(12):1806–1808
Chen J, Wang Y, Wu XJ, Li J, Hou FQ, Wang GQ (2010) Pegylated interferon α-2b up-
regulates specific CD8+ T cells in patients with chronic hepatitis B. World J Gastroenterol
16(48):6145–6150
Cooper A, Tal G, Lider O, Shaul Y (2005) Cytokine induction by the hepatitis B virus capsid
in macrophages is facilitated by membrane heparan sulfate and involves TLR2. J Immunol
175(5):3165–3176
Das A, Ellis G, Pallant C, Lopes AR, Khanna P, Peppa D, Chen A, Blair P, Dusheiko G, Gill U,
Kennedy PT, Brunetto M, Lampertico P, Mauri C et al (2012) IL-10-producing regulatory B
cells in the pathogenesis of chronic hepatitis B virus infection. J Immunol 189(8):3925–3935
Della Chiesa M, Romagnani C, Thiel A, Moretta L, Moretta A (2006) Multidirectional interactions
are bridging human NK cells with plasmacytoid and monocyte-derived dendritic cells during
innate immune responses. Blood 108(12):3851–3858
Deng G, Ge J, Liu C, Pang J, Huang Z, Peng J, Sun J, Hou J, Zhang X (2017) Impaired expression
and function of TLR8 in chronic HBV infection and its association with treatment responses
during peg-IFN-α-2a antiviral therapy. Clin Res Hepatol Gastroenterol 41(4):386–398
Dunn C, Peppa D, Khanna P, Nebbia G, Jones M, Brendish N, Lascar RM, Brown D, Gilson RJ,
Tedder RJ, Dusheiko GM, Jacobs M, Klenerman P, Maini MK (2009) Temporal analysis of
early immune responses in patients with acute hepatitis B virus infection. Gastroenterology
137(4):1289–1300
Fan Z, EnQiang C, Yao DL, LiBo Y, Hong L, Lang B, Ping F et al (2017) Neutrophil-lymphocyte
ratio predicts short term mortality in patients with hepatitis B virus-related acute-on-chronic
liver failure treated with an artificial liver support system. PLoS One 12(4):e0175332
Faure-Dupuy S, Lucifora J, Durantel D (2017) Interplay between the hepatitis B virus and innate
immunity: from an understanding to the development of therapeutic concepts. Viruses 9(5):95
Feng IC, Koay LB, Sheu MJ, Kuo HT, Sun CS, Lee C, Chuang WL, Liao SK, Wang SL, Tang LY,
Cheng CJ, Tsai SL (2007) HBcAg-specific CD4+CD25+ regulatory T cells modulate immune
tolerance and acute exacerbation on the natural history of chronic hepatitis B virus infection. J
Biomed Sci 14(1):43–57
Fisicaro P, Valdatta C, Boni C, Massari M, Mori C, Zerbini A, Orlandini A, Sacchelli L, Missale
G, Ferrari C (2009) Early kinetics of innate and adaptive immune responses during hepatitis B
virus infection. Gut 58(7):974–982
14 Role of Immune Cells in Hepatitis B Infection 221
Fontaine H, Kahi S, Chazallon C, Bourgine M, Varaut A, Buffet C, Godon O, Meritet JF, Saïdi Y,
Michel ML, Scott-Algara D, Aboulker JP, Pol S, ANRS HB02 Study Group (2015) Anti-HBV
DNA vaccination does not prevent relapse after discontinuation of analogues in the treatment
of chronic hepatitis B: a randomised trial--ANRS HB02 VAC-ADN. Gut 64(1):139–147
Franzese O, Kennedy PT, Gehring AJ, Gotto J, Williams R, Maini MK, Bertoletti A (2005)
Modulation of the CD8+-T-cell response by CD4+ CD25+ regulatory T cells in patients with
hepatitis B virus infection. J Virol 79(6):3322–3328
Fu J, Xu D, Liu Z, Shi M, Zhao P, Fu B, Zhang Z, Yang H, Zhang H, Zhou C, Yao J, Jin L,
Wang H, Yang Y, Fu YX, Wang FS (2007) Increased regulatory T cells correlate with CD8
T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology
132(7):2328–2339
Ganem D, Prince AM (2004) Hepatitis B virus infection--natural history and clinical conse-
quences. N Engl J Med 350(11):1118–1129
Gehring AJ, Koh S, Chia A, Paramasivam K, Chew VS, Ho ZZ, Lee KH, Maini MK, Madhavan K,
Lim SG et al (2011) Licensing virus-specific T cells to secrete the neutrophil attracting chemo-
kine CXCL-8 during hepatitis B virus infection. PLoS One 6(8):e23330
Gehring AJ, Haniffa M, Kennedy PT, Ho ZZ, Boni C, Shin A, Banu N, Chia A, Lim SG, Ferrari
C, Ginhoux F et al (2013) Mobilizing monocytes to cross-present circulating viral antigen in
chronic infection. J Clin Invest 123(9):3766–3776
Ghosh S, Nandi M, Pal S, Mukhopadhyay D, Chakraborty BC, Khatun M, Bhowmick D, Mondal
RK, Das S, Das K, Ghosh R, Banerjee S, Santra A, Chatterjee M, Chowdhury A, Datta S (2016)
Natural killer cells contribute to hepatic injury and help in viral persistence during progres-
sion of hepatitis B e-antigen-negative chronic hepatitis B virus infection. Clin Microbiol Infect
22(8):733
Guang LX (1992) Hepatitis B virus infection in monocytes and their functional changes in patients
with chronic hepatitis B. Zhonghua Yi Xue Za Zhi 72(11):651–654. 701
Guidotti LG, Ishikawa T, Hobbs MV, Matzke B, Schreiber R, Chisari FV (1996) Intracellular inac-
tivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity 4(1):25–36
Han YF, Zhao J, Ma LY, Yin JH, Chang WJ, Zhang HW, Cao GW (2011) Factors predicting
occurrence and prognosis of hepatitis-B-virus-related hepatocellular carcinoma. World J
Gastroenterol 17(38):4258–4270
Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, Weaver CT (2005)
Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper
type 1 and 2 lineages. Nat Immunol 6(11):1123–1132
Highleyman L (2017, September 27) US FDA approves nivolumab immunotherapy for liver can-
cer. Retrieved from http://www.infohep.org/page/3175886
Hou W, Gibbs JS, Lu X, Brooke CB, Roy D, Modlin RL, Bennink JR et al (2012) Viral infec-
tion triggers rapid differentiation of human blood monocytes into dendritic cells. Blood
119(13):3128–3131
Huang M, Sun R, Wei H, Tian Z (2013) Simultaneous knockdown of multiple ligands of innate
receptor NKG2D prevents natural killer cell-mediated fulminant hepatitis in mice. Hepatology
57(1):277–288
Janssen HLA, Brunetto MR, Kim YJ, Ferrari C, Massetto B, Nguyen AH, Joshi A, Woo J, Lau AH,
Gaggar A, Subramanian GM, Yoshida EM, Ahn SH, Tsai NCS, Fung S, Gane EJ (2018) Safety,
efficacy and pharmacodynamics of vesatolimod (GS-9620) in virally suppressed patients with
chronic hepatitis B. J Hepatol 68(3):431–440
Jiang R, Feng X, Guo Y, Lu Q, Hou J, Luo K, Fu N (2002) Early kinetics of innate and adaptive
immune responses during hepatitis B virus infection. Chin Med J 115(3):422–424
Kadowaki N (2009) The divergence and interplay between pDC and mDC in humans. Front Biosci
(Landmark Ed) 14:808–817
Kalathil S, Lugade AA, Miller A, Iyer R, Thanavala Y (2013) Higher frequencies of GARP(+)
CTLA-4(+)Foxp3(+) T regulatory cells and myeloid-derived suppressor cells in hepato-
cellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res
73(8):2435–2444
222 P. Sinha and P. Sahu
Kalathil SG, Lugade AA, Miller A, Iyer R, Thanavala Y (2016) PD-1+ and Foxp3+ T cell reduc-
tion correlates with survival of HCC patients after sorafenib therapy. JCI Insight 1(11):e86182
Kamphorst AO, Ahmed R (2013) CD4 T-cell immunotherapy for chronic viral infections and can-
cer. Immunotherapy 5(9):975–987
Khanam A, Trehanpati N, Riese P, Rastogi A, Guzman CA, Sarin SK (2017) Blockade of neutro-
phil’s chemokine receptors CXCR1/2 abrogate liver damage in acute-on-chronic liver failure.
Front Immunol 8:464. https://doi.org/10.3389/fimmu.2017.00464
Koumbi L (2015) Current and future antiviral drug therapies of hepatitis B chronic infection.
World J Hepatol 7(8):1030–1040
Lamontagne RJ, Bagga S, Bouchard MJ (2016) Hepatitis B virus molecular biology and pathogen-
esis. Hepatoma Res 2:163–186
Lanford RE, Guerra B, Chavez D, Giavedoni L, Hodara VL, Brasky KM, Fosdick A, Frey CR,
Zheng J, Wolfgang G, Halcomb RL, Tumas DB (2013) GS-9620, an oral agonist of Toll-like
receptor-7, induces prolonged suppression of hepatitis B virus in chronically infected chimpan-
zees. Gastroenterology 144(7):1508–1517
Le PH, Liang KH, Chang ML, Hsu CW, Chen YC, Lin CL, Lin WR, Lai MW et al (2017) Clinical
predictors for neutrophil-to-lymphocyte ratio changes in patients with chronic hepatitis B
receiving peginterferon treatment. In Vivo 31(4):723–729
Leu C, Lu YC, Peng WL, Chu HT, Hu CP (2014) The hepatitis B virus e antigen suppresses
the respiratory burst and mobility of human monocytes and neutrophils. Immunobiology
219(11):880–887
Li J, Qiu SJ, She WM, Wang FP, Gao H, Li L, Tu CT, Wang JY, Shen XZ et al (2012) Significance
of the balance between regulatory T (Treg) and T helper 17 (Th17) cells during hepatitis B
virus related liver fibrosis. PLoS One 7(6):e39307
Li X, Liu X, Tian L, Chen Y (2016a) Cytokine-mediated immunopathogenesis of hepatitis B virus
infections. Clin Rev Allergy Immunol 50(1):41–54
Li W, Han J, Wu H (2016b) Regulatory T-cells promote hepatitis B virus infection and hepatocel-
lular carcinoma progression. Chronic Dis Transl Med 2(2):67–80. https://doi.org/10.1016/j.
cdtm.2016.09.001
Li H, Zhai N, Wang Z, Song H, Yang Y, Cui A, Li T, Wang G, Niu J, Crispe IN, Su L et al (2017)
Regulatory NK cells mediated between immunosuppressive monocytes and dysfunctional T
cells in chronic HBV infection. Gut 67(11):2035–2044
Liang TJ (2009) Hepatitis B: the virus and disease. Hepatology 49(5 Suppl):S13–S21. https://doi.
org/10.1002/hep.22881
Liu H, Zhang H, Wan G, Sang Y, Chang Y, Wang X, Zeng H (2014a) Neutrophil-lymphocyte ratio:
a novel predictor for short-term prognosis in acute-on-chronic hepatitis B liver failure. J Viral
Hepat 21(7):499–507
Liu J, Zhang E, Ma Z, Wu W, Kosinska A, Zhang X, Möller I, Seiz P, Glebe D, Wang B, Yang
D, Lu M, Roggendorf M (2014b) Enhancing virus-specific immunity in vivo by combining
therapeutic vaccination and PD-L1 blockade in chronic hepadnaviral infection. PLoS Pathog
10(1):e1003856
Lumley SF, McNaughton AL, Klenerman P, Lythgoe KA, Matthews PC (2018) Hepatitis B virus
adaptation to the CD8+ T cell response: consequences for host and pathogen. Front Immunol
9:1561
Luo J, Li J, Chen RL, Nie L, Huang J, Liu ZW, Luo L, Yan XJ (2010) Autologus dendritic cell
vaccine for chronic hepatitis B carriers: a pilot, open label, clinical trial in human volunteers.
Vaccine 28(13):2497–2504
Lv S, Wang J, Dou S, Yang X, Ni X, Sun R, Tian Z, Wei H (2014) Nanoparticles encapsulat-
ing hepatitis B virus cytosine-phosphate-guanosine induce therapeutic immunity against HBV
infection. Hepatology 59:385–394
Ma Z, Cao Q, Xiong Y, Zhang E, Lu M (2018) Interaction between hepatitis B virus and toll-like
receptors: current status and potential therapeutic use for chronic hepatitis B. Vaccine 6(1):6
14 Role of Immune Cells in Hepatitis B Infection 223
of CD244 in chronic hepatitis B infection and its inhibitory potential on virus-specific CD8+
T-cell function. Hepatology 52(6):1934–1947
Raziorrouh B, Heeg M, Kurktschiev P, Schraut W, Zachoval R, Wendtner C et al (2014) Inhibitory
phenotype of HBV-specific CD4+ T-cells is characterized by high PD-1 expression but
absent coregulation of multiple inhibitory molecules. PLoS One 9(8):e105703. https://doi.
org/10.1371/journal.pone.0105703
Rehermann B, Fowler P, Sidney J, Person J, Redeker A, Brown M, Moss B, Sette A et al (1995)
The cytotoxic T lymphocyte response to multiple hepatitis B virus polymerase epitopes during
and after acute viral hepatitis. J Exp Med 181(3):1047–1058
Riordan SM, Skinner N, Kurtovic J, Locarnini S, Visvanathan K (2006) Reduced expression of
toll-like receptor 2 on peripheral monocytes in patients with chronic hepatitis B. Clin Vaccine
Immunol 13(8):972–974
Sant AJ, McMichael A (2012) Revealing the role of CD4(+) T cells in viral immunity. J Exp Med
209(8):1391–1395
Schurich A, Khanna P, Lopes AR, Han KJ, Peppa D, Micco L, Nebbia G, Kennedy PT, Geretti
AM, Dusheiko G, Maini MK (2011) Role of the coinhibitory receptor cytotoxic T lymphocyte
antigen-4 on apoptosis-Prone CD8 T cells in persistent hepatitis B virus infection. Hepatology
53(5):1494–1503. https://doi.org/10.1002/hep.24249
Shi M, Qian S, Chen WW, Zhang H, Zhang B, Tang ZR, Zhang Z et al (2007) Hepatitis B virus
(HBV) antigen-pulsed monocyte-derived dendritic cells from HBV-associated hepatocellular
carcinoma patients significantly enhance specific T cell responses in vitro. Clin Exp Immunol
147(2):277–286
Shi B, Ren G, Hu Y, Wang S, Zhang Z, Yuan Z (2012) HBsAg inhibits IFN-α production in plasma-
cytoid dendritic cells through TNF-α and IL-10 induction in monocytes. PLoS One 7(9):e44900
Shrivastava S, TrehanPati N, Patra S, Kottilil S, Pande C, Trivedi SS, Sarin SK (2013) Increased
regulatory T cells and impaired functions of circulating CD8 T lymphocytes is associated with
viral persistence in Hepatitis B virus-positive newborns. J Viral Hepat 20(8):582–591
Sitia G, Isogawa M, Kakimi K, Wieland SF, Chisari FV, Guidotti LG (2002) Depletion of neu-
trophils blocks the recruitment of antigen-nonspecific cells into the liver without affecting the
antiviral activity of hepatitis B virus-specific cytotoxic T lymphocytes. Proc Natl Acad Sci U
S A 99(21):13717–13722
Stelma F, de Niet A, Tempelmans Plat-Sinnige MJ, Jansen L, Takkenberg RB, Reesink HW,
Kootstra NA, van Leeuwen EM (2015) Natural killer cell characteristics in patients with
chronic hepatitis B virus (HBV) infection are associated with HBV surface antigen clear-
ance after combination treatment with pegylated interferon alfa-2a and adefovir. J Infect Dis
212(7):1042–1051
Stelma F, Willemse SB, Erken R, de Niet A, Sinnige MJ, van Dort K, Zaaijer HL, van Leeuwen
E, Kootstra NA, Reesink HW (2017) Dynamics of the immune response in acute hepatitis B
infection. Open Forum Infect Dis 4(4):ofx231. https://doi.org/10.1093/ofid/ofx231
Stoop JN, van der Molen RG, Baan CC, van der Laan LJ, Kuipers EJ, Kusters JG, Janssen HL
(2005) Regulatory T cells contribute to the impaired immune response in patients with chronic
hepatitis B virus infection. Hepatology 41(4):771–778
Stoop JN, Claassen MA, Woltman AM, Binda RS, Kuipers EJ, Janssen HL, van der Molen RG,
Boonstra A (2008) Intrahepatic regulatory T cells are phenotypically distinct from their periph-
eral counterparts in chronic HBV patients. Clin Immunol Soc 129(3):419–427
Stross L, Günther J, Gasteiger G, Asen T, Graf S, Aichler M, Esposito I, Busch DH, Knolle P,
Sparwasser T, Protzer U (2012) Foxp3+ regulatory T cells protect the liver from immune dam-
age and compromise virus control during acute experimental hepatitis B virus infection in
mice. Hepatology 56(3):873–883
Sun HQ, Zhang JY, Zhang H, Zou ZS, Wang FS, Jia JH (2012) Increased Th17 cells contrib-
ute to disease progression in patients with HBV-associated liver cirrhosis. J Viral Hepat
19(6):396–403
Takai S, Kimura K, Nagaki M, Satake S, Kakimi K, Moriwaki H (2005) Blockade of neutrophil elas-
tase attenuates severe liver injury in hepatitis B transgenic mice. J Virol 79(24):15142–15150
14 Role of Immune Cells in Hepatitis B Infection 225
Tan Z, Qian X, Jiang R, Liu Q, Wang Y, Chen C, Wang X, Ryffel B, Sun B (2013) IL-17A plays
a critical role in the pathogenesis of liver fibrosis through hepatic stellate cell activation. J
Immunol 191(4):1835–1844
Tang TJ, Kwekkeboom J, Laman JD, Niesters HG, Zondervan PE, de Man RA, Schalm SW,
Janssen HL (2003) Regulatory T cells in HBV and HCV liver diseases: implication of regula-
tory T lymphocytes in the control of immune response. J Viral Hepat 10(3):159–167
Thimme R, Wieland S, Steiger C, Ghrayeb J, Reimann KA, Purcell RH, Chisari FV (2003) CD8(+)
T cells mediate viral clearance and disease pathogenesis during acute hepatitis B virus infec-
tion. J Virol 77(1):68–76
Tjwa ET, van Oord GW, Hegmans JP, Janssen HL, Woltman AM (2011) Viral load reduction
improves activation and function of natural killer cells in patients with chronic hepatitis B. J
Hepatol 54(2):209–218
Trepo C, Chan HLY, Lok A (2014) Hepatitis B virus infection. Lancet 384:2053–2063
Untergasser A, Zedler U, Langenkamp A, Hösel M, Quasdorff M, Esser K, Dienes HP,
Tappertzhofen B, Kolanus W, Protzer U (2006) Dendritic cells take up viral antigens but do not
support the early steps of hepatitis B virus infection. Hepatology 43(3):539–547
van der Molen RG, Sprengers D, Binda RS, de Jong EC, Niesters HG, Kusters JG, Kwekkeboom
J, Janssen HL (2004) Functional impairment of myeloid and plasmacytoid dendritic cells of
patients with chronic hepatitis B. Hepatology 40(3):738–746
Van der Molen RG, Sprengers D, Biesta PJ, Kusters JG, Janssen HL (2006) Favorable effect of
adefovir on the number and functionality of myeloid dendritic cells of patients with chronic
HBV. Hepatology 44(4):907–914
Vanlandschoot P, Roobrouck A, Van Houtte F, Leroux-Roels G (2002) Recombinant HBsAg, an
apoptotic-like lipoprotein, interferes with the LPS-induced activation of ERK-1/2 and JNK-
1/2 in monocytes. Biochem Biophys Res Commun 297(3):486–491
Vincent IE, Zannetti C, Lucifora J, Norder H, Protzer U, Hainaut P, Zoulim F, Tommasino M,
Trépo C, Hasan U et al (2011) Hepatitis B virus impairs TLR9 expression and function in
plasmacytoid dendritic cells. PLoS One 6(10):e26315
Visvanathan K, Skinner NA, Thompson AJ, Riordan SM, Sozzi V, Edwards R, Rodgers S, Kurtovic
J, Chang J, Lewin S, Desmond P, Locarnini S (2007) Regulation of toll-like receptor-2 expres-
sion in chronic hepatitis B by the precore protein. Hepatology 45(1):102–110
Wang S, Chen Z, Hu C, Qian F, Cheng Y, Wu M, Shi B, Chen J, Hu Y, Yuan Z (2013) Hepatitis B
virus surface antigen selectively inhibits TLR2 ligand-induced IL-12 production in monocytes/
macrophages by interfering with JNK activation. J Immunol 190(10):5142–5151
Wang G, Liu Y, Huang R, Jia B, Su R, Sun Z, Tian C, Xiong Y, Xia J, Yan X, Zhang Z, Wu C (2017)
Characteristics of regulatory B cells in patients with chronic hepatitis B virus infection in dif-
ferent immune phases. Discov Med 23(128):295–304
Wang Q, Pan W, Liu Y, Luo J, Zhu D, Lu Y, Feng X, Yang X, Dittmer U, Lu M, Yang D et al
(2018) Hepatitis B virus-specific CD8+ T cells maintain functional exhaustion after antigen
reexposure in an acute activation immune environment. Front Immunol 9:219. https://doi.
org/10.3389/fimmu.2018.00219
Webster GJ, Reignat S, Brown D, Ogg GS, Jones L, Seneviratne SL, Williams R, Dusheiko G,
Bertoletti A (2004) Longitudinal analysis of CD8+ T cells specific for structural and nonstruc-
tural hepatitis B virus proteins in patients with chronic hepatitis B: implications for immuno-
therapy. J Virol 78(11):5707–5719
WHO (2017, April) Global hepatitis report. Retrieved from http://www.who.int/hepatitis/
publications/global-hepatitis-report2017/en/
Wisskirchen K, Metzger K, Schreiber S, Asen T, Weigand L, Dargel C, Witter K, Kieback E, Sprinzl
MF, Uckert W, Schiemann M, Busch DH, Krackhardt AM et al (2017) Isolation and functional
characterization of hepatitis B virus-specific T-cell receptors as new tools for experimental and
clinical use. PLoS One 12(8):e0182936. https://doi.org/10.1371/journal.pone.0182936
Wu C, Liu Y, Zhao Q, Chen G, Chen J, Yan X, Zhou YH et al (2010) Soluble CD40 ligand-activated
human peripheral B cells as surrogated antigen presenting cells: a preliminary approach for
anti-HBV immunotherapy. Virol J 7:370. https://doi.org/10.1186/1743-422X-7-370
226 P. Sinha and P. Sahu
Zhang JY, Zou ZS, Huang A, Zhang Z, Fu JL, Xu XS, Chen LM, Li BS, Wang FS (2011) Hyper-
activated pro-inflammatory CD16 monocytes correlate with the severity of liver injury and
fibrosis in patients with chronic hepatitis B. PLoS One 6(3):e17484
Zhang H, Sun Q, Mao W, Fan J, Ye B (2016) Neutrophil-to-lymphocyte ratio predicts early
mortality in patients with HBV-related decompensated cirrhosis. Gastroenterol Res Pract
2016:4394650
Zhang Y, Wu Y, Deng M, Xu D, Li X, Xu Z, Hu J, Zhang H, Liu K, Zhao Y, Gao F, Bi S, Gao GF,
Zhao J, Liu WJ, Meng S (2018) CD8+ T-cell response-associated evolution of hepatitis B virus
core protein and disease progress. J Virol 92(17):e02120-17
Zhao J, Li Y, Jin L, Zhang S, Fa R, Sun Y, Zhou C, Shang Q, Li W, Zhang Z, Wang FS (2012)
Natural killer cells are characterized by the concomitantly increased interferon-γ and cytotox-
icity in acute resolved hepatitis B patients. PLoS One 7(11):e49135
Zhao PW, Ma L, Ji HF, Yu L, Feng JY, Wang J, Liu MY et al (2015) The expression of TLR-9,
CD86, and CD95 phenotypes in circulating B cells of patients with chronic viral hepatitis B or
C before and after antiviral therapy. Mediat Inflamm 2015:762709
Significance of RNA Sensors
in Activating Immune System 15
in Emerging Viral Diseases
Abstract
Viruses are known for their utilization of the host machinery to aid their replica-
tion within the infected cells. The viral nucleic acid, their intermittent forms con-
tain pathogen-associated molecular patterns (PAMP) which enable the pattern
recognition receptors (PRR) to distinguish them from self and mount a response
against them. Highly robust targets for the PRRs are the cell entry points and
phases of viral replication. RNA-sensing PRR can be classified as endosomal
PRR and cytosolic PRR. Both enveloped and non-enveloped viruses utilize endo-
somal compartments to undergo proteolytic cleavage or pH-dependent conforma-
tional changes to ensure membrane fusion. Thus, monitoring the endosomal
compartments in order to restrict the virus from penetrating into the cytoplasm is
a key antiviral strategy. Endosomal compartments are guarded by Toll-like recep-
tors (TLRs) which are the earliest discovered PRRs. TLR3, 7, and 8 specifically
cater to viral RNA sensing. They can detect dsRNA and stable stem structures of
ssRNA. Cytosolic PRRs include RIG-I like receptors (RLR) and nucleotide-bind-
ing oligomerization domain-containing (NOD)—like receptors (NLR). RLRs are
cytosolic helicases which detect viral RNA, ds RNA, short 5′ppp RNA, and RNase
L cleaved RNA. Retinoic acid-inducible gene I product (RIG-I), melanoma differ-
entiation-associated antigen 5 (MDA5), and laboratory of genetics and physiology
2 (LGP2) are aspartate-glutamate-any amino acid-aspartate/histidine (DExD/H)-
box helicases belonging to the SF2 superfamily. Other RNA helicases like
SNRNP200 which belongs to the Ski-2 superfamily, or DDX60 belonging to the
Ski-2-like helicase family are also involved in viral RNA sensing. NLRs like
NLRP3 and NOD2 are cytosolic RNA-sensing PRRs. In addition to these, RNA-
binding proteins like RNase L, protein kinase R (PKR), and interferon-inducible
P. Nair (*)
NMIMS School of Science, Mumbai, India
S. U. Sapre
National Institute of Virology, Pune, India
transmembrane protein (IFIT) are also present which play pertinent roles to
enhance the antiviral immunity during a viral infection. Recognition of PAMPs or
DAMPs by PRRs activate various signaling cascades which ultimately triggers the
transcription of type I/III IFN, production of various pro-inflammatory cytokines,
and various other genes that can ensure an intracellular antiviral state which will
aid in containing the viral infection. Furthermore, these interferons can lead to the
expression of multiple interferon-stimulated genes (ISGs) which induce antiviral
activities controlling the life cycle of the virus, restricting its replication, and
transmission to the surrounding cells. This chapter will elaborate on these RNA-
sensing PRRs, their structures, agonists, mechanism of activation, and response
mounted against viral infection.
Keywords
Host machinery RNA sensors · Viral diseases · Immune activation
15.1 Introduction
Viruses are known for their utilization of the host machinery to aid their replication
within the infected cells. The viral nucleic acid, its intermittent forms contain
pathogen-associated molecular patterns (PAMP) which enable the pattern recogni-
tion receptors (PRR) to distinguish them from self and mount a response against
them. Highly robust targets for the PRRs are the cell entry points and phases of viral
replication. RNA-sensing PRR can be classified as endosomal PRR and cytosolic
PRR. Both enveloped and non-enveloped viruses utilize endosomal compartments
to undergo proteolytic cleavage or pH-dependent conformational changes to ensure
membrane fusion. Thus, monitoring the endosomal compartments in order to restrict
the virus from penetrating into the cytoplasm is a key antiviral strategy. Endosomal
compartments are guarded by Toll-like receptors (TLRs) which are the earliest dis-
covered PRRs. TLR3, 7, and 8 specifically cater to viral RNA sensing. They can
detect dsRNA and stable stem structures of ssRNA.
Cytosolic PRRs include RIG-I like receptors (RLR) and nucleotide-binding
oligomerization domain-containing (NOD)—like receptors (NLR). RLRs are cyto-
solic helicases which detect viral RNA, ds RNA, short 5′ppp RNA and RNase L
cleaved RNA. Retinoic acid-inducible gene I product (RIG-I), melanoma
differentiation-associated antigen 5 (MDA5), and laboratory of genetics and physi-
ology 2 (LGP2) are aspartate-glutamate-any amino acid-aspartate/histidine (DExD/
H)-box helicases belonging to the SF2 superfamily. Other RNA helicases like
SNRNP200 which belongs to the Ski-2 superfamily, or DDX60 belonging to the
Ski-2-like helicase family are also involved in viral RNA sensing. NLRs like NLRP3
and NOD2 are cytosolic RNA-sensing PRRs. In addition to these, RNA-binding
proteins like RNase L, protein kinase R (PKR), and interferon-inducible transmem-
brane protein (IFIT) are also present which play pertinent roles to enhance the anti-
viral immunity during a viral infection.
15 Significance of RNA Sensors in Activating Immune System in Emerging Viral… 231
Innate immunity provides the first line of defence against the invading pathogens. It
exerts its role by distinguishing between self and non-self, subsequently initiating a
host response against the invading pathogen. Innate immunity is triggered by the
identification of PAMPs (pathogen-associated molecular patterns) via the PRRs
(pattern recognition receptors). PRRs are germline-encoded proteins expressed by a
variety of cells which can carry out surveillance to detect any pathogenic invasions.
PRRs have been classified into various families based on protein domain homology;
Toll-like receptors (TLRs), RIG-I like receptors (RLRs), NOD-like receptors
(NLRs), C-type lectin receptors (CLRs), AIM2-like receptors (ALRs), and cytosol
DNA sensing PRR cyclic GMP-AMP synthase (cGAS). PRRs not only survey the
PAMPS but also the DAMPS (danger-associated molecular patterns), which are
typically the cellular products produced as a response to cell stress (Anafu et al.
2013; Chen et al. 2017; Chow et al. 2018; Said et al. 2018).
Recognition of PAMPs or DAMPs by PRRs leads to transcription of genes involved
in pro-inflammatory responses. The responses mounted against a viral infection,
upregulates type I and III interferons, pro-inflammatory chemokines, and various
other genes that can ensure an intracellular antiviral state in order to contain the infec-
tion. Furthermore, these interferons can lead to the expression of multiple interferon-
stimulated genes (ISGs) which induce antiviral activities controlling the life cycle of
the virus, restricting its replication and transmission to the surrounding cells.
Viruses are known for their utilization of the host machinery to aid their replica-
tion within the infected cells. The viral nucleic acid, their intermittent forms contain
PAMPs which enable the PRRs to distinguish them from self and mount a response
against them. Highly robust targets for the PRRs are the cell entry points and phases
of viral replication. Both enveloped and non-enveloped viruses utilize endosomal
compartments to undergo proteolytic cleavage or pH-dependent conformational
changes to ensure membrane fusion (Said et al. 2018). Thus, monitoring the endo-
somal compartments in order to restrict the virus from penetrating into the cyto-
plasm is a key antiviral strategy. Endosomal compartments are guarded by Toll-like
receptors (TLRs) which are the earliest discovered PRRs. TLR3, 7, and 8 specifi-
cally cater to viral RNA sensing. Cytosolic PRRs include RIG-I, MDA5, LGP2,
NLRP3, and NOD2. RNA sensing is also carried out by other RNA helicases like
232 P. Nair and S. U. Sapre
TLRs are a very important and earliest detected type of PRRs. They comprise of 11
genes on the human genome. Majority of the TLRs are associated with the plasma
membrane. However, the TLRs associated with RNA sensing namely, TLR3, 7, and
8 are present in the endosomal compartment. TLRs are type I transmembrane pro-
teins which consists of N-terminal ectodomain or extracellular leucine-rich domain
(ECD), middle transmembrane domain (TM), and C-terminal cytoplasmic Toll/IL-1
receptor (TIR) domain. Leucine-rich regions (LRRs) in the ECD are attached into a
horseshoe-shaped solenoid structure. α-helices of each LRR forms into the convex
structure of the solenoid and the β sheets assemble into the concave surfaces of the
solenoid structure. It is a unique property of the TLRs to bind their agonists to the
lateral convex surface instead of concave surface.
The formation of M-shaped dimer or multimer is needed for all TLR activation,
so that the C-terminal regions of the two TLR ECDs are brought into proximity. It
in turn causes the multimerization of cytoplasmic TIR domains, which will recruit
downstream adaptors TRIF or MyD88 through homotypic interaction, further form-
ing signaling complex called signalosome and activating downstream transcription
factors like NF-κB which induces pro-inflammatory cytokines and interferon regu-
latory factor (IRF) which in turn induces type I interferon.
Further, there are positive regulators to this pathway like S100A9 which acts dur-
ing the early stages of TLR3 activation by easing the maturation of TLR3-containing
early endosomes into late endosomes, etc. Similarly, there are negative regulators
such as Rho proteins that decrease the production of pro-inflammatory cytokines
upon TLR3 triggering. The pathway is also regulated via post-translational modifi-
cations.TLR3 has been implicated in pathogenesis of HCV, infection, HSV-1, HBV,
etc. This makes TLR3 an interesting candidate to prod into antiviral therapies.
The first line of cytosolic RNA sensors include the PRR families RLRs and
NLRs. RLRs consists of cytosolic helicases: RIG-I, MDA5, and LGP2; other
DEXD/H-box helicases like DDX3, DDX60, and SNRP20 (Chow et al. 2018;
Dang et al. 2018).
234 P. Nair and S. U. Sapre
Cytosolic helicases are ubiquitously expressed in low levels in most cell types.
They belong to a family of aspartate-glutamate-any amino acid-aspartate/histi-
dine (DExD/H)-box helicases (SF2 superfamily). RLRs are also known as double-
stranded RNA (dsRNA)-dependent ATPases. Their function includes sensing the
viral RNA in the cytosol, binding to the non-self RNA stably, and imposing the
innate immune response against the target RNA. Three members widely studied
are the retinoic acid-inducible gene I product (RIG-I), melanoma differentiation-
associated antigen 5 (MDA5), and laboratory of genetics and physiology 2
(LGP2).
15 Significance of RNA Sensors in Activating Immune System in Emerging Viral… 235
15.4.1.1 RIG-I
When disengaged, the RIG-I CARDs and RD are bound to the helicase region and
this leads to autoinhibition (Chow et al. 2018; Dang et al. 2018; Pichlmair et al.
2006). During viral replication the CTD/RD detects and associates with the viral
RNA along with the helicase, exposing the CARDS. These CARDs are no longer
autorepressed; the RLRs oligomerize and further associate with the mitochondrial
antiviral signaling adaptor called MAVS (also referred to as IPS-1/VISA/cardif).
MAVS are located at the cytosolic face of the outer mitochondrial membrane, and
this mitochondrial association is necessary for initiating further signaling events.
On association the MAVS undergo a prion-like aggregation. This leads to the
recruitment of E3 ubiquitin ligases and the downstream effector proteins TNF
receptor-associated factor 2 (TRAF2), TRAF3, and TRAF6 which assembles into
an active “signalosome.” The resulting cascade leads to the phosphorylation and
nuclear translocation of key innate immune transcription factors IRF3 and IRF7.
Further, it leads to the activation of NF-κB to drive the expression of type I and III
interferons, innate immune genes, pro-inflammatory cytokines, and chemokines
that will aid in containing the virus infection.
The RD domain is not only critical for RNA sensing but also confers it with the
displayed selectivity. RIG-I can recognize 5′ppp dsRNA, 5′pp dsRNA, pU/UC
genomic RNA, AU-rich 3 UTR, RNase L cleavage products and circular viral RNA
(Chow et al. 2018; Said et al. 2018; Dang et al. 2018; Liu and Gale Jr 2011;
Yoneyama and Fujita 2007; Zou et al. 2009). The distinguishing between self and
non-self RNA is based on the 5′-7-methylguanosine cap, which is subject to
2′-O-methylation.
15.4.1.2 MDA5
RIG-I and MDA5 being homologues showcase the same overall domain structures.
The RD domain however recognizes long dsRNA which under normal circum-
stances is absent in an uninfected cell. The presence of this PAMP signifies vial
invasion which activates MDA5 and stimulates a cytokine response similar to the
RIG-I. MDA5 binds to the long dsRNA replicative intermediates generated by
picornavirus, consisting of its positive-sense genome annealed to the negative-sense
antigenome, AU-rich motifs and RNase L cleavage products (Said et al. 2018; Dang
et al. 2018; Bruns et al. 2014; Rodriguez et al. 2014).
236 P. Nair and S. U. Sapre
On binding with the target RNA it undergoes a conformational change and the 2
CARD domains are exposed. The 2 CARDS assemble into a stable tetramer due to
its binding with the dsRNA and they form a filament structure. They therefore are
independent on the polyubiquitin chain binding; the tetramer itself nucleates the
MAVS, which in turn activates TRAF3/TBK1/IKKe/IRF3 and TRAF6/IKK/NF-jB,
which drive IFN and pro-inflammatory cytokine expression, respectively.
15.4.1.3 LGP2
Although LGP2 is homologues to RIG-I and MDA5, it lacks the CARD domain
which restricts it from binding to the MAVS. However, LGP2 is shown to bind to
the termini of blunt-ended dsRNA of different lengths with high affinity, forming
complexes with 2:1 stoichiometry. LGP2 has been both implicated as a positive and
negative effector of RIG-I and MDA5 response (Said et al. 2018; Jensen and
Thomsen 2012; Bruns et al. 2014; Rodriguez et al. 2014). Studies have shown that
CTDs of LGP2 and RIG-I are analogous because of which LGP2 CTD interacts
with RIG-I to abolish its ability to initiate antiviral signaling. RIG-I also undergoes
attenuation of viral sensing. LGP2 has also been believed to utilize its ATP depen-
dent/RNA helicase activity to increase the interaction of nucleic acid PAMPS for
improved antiviral RLR signaling. LGP2 has also been implicated for its activity
against DICER protein to maintain the cytosolic PAMPs in intact condition for
detection by the cytosolic RNA sensors. Therefore, LGP2 overall is a modulator of
the innate immune response to a viral infection and not a sensor of PAMPs. LGP2
does not initiate antiviral gene expression.
amino terminal of SNRP200 binds to the vRNA, perinuclear relocation occurs and
it aids as an adaptor o trigger IRF3 signaling.
15.4.2 NLRs
Multiple RNA-binding proteins are also involved directly or indirectly in the pro-
cess of vRNA sensing.
Fig. 15.2 Structure, activation, and mechanism of action of cytosolic PRRs—RIG-I, MDA5, and
LGP2. Overview of endosomal and cytosolic RNA sensors
substrates to RNase L. The cleavage products are utilized for increasing the activa-
tion of RIG-I/MDA5. These are being studied in order to use them as targets for
antiviral therapies (Sarkar 2014).
(continued)
239
Endosomal RNA sensors Cytosolic RNA sensors RNA-binding proteins
240
Acknowledgments Authors are grateful to NIV Pune and NMIMS Pune for the support extended.
Conflict of Interest The authors declare that they have no competing interests.
References
Anafu AA, Bowen CH, Chin CR, Brass AL, Holm GH (2013) Interferon inducible transmembrane
protein 3 (IFITM3) restricts reovirus cell entry. J Biol Chem 288(24):17261–17271
Bruns AM, Leser GP, Lamb RA, Horvath CM (2014) The innate immune sensor LGP2 activates
antiviral signaling by regulating MDA5-RNA interaction and filament assembly. Mol Cell
55(5):771–781
Chakrabarti A, Banerjee S, Franchi L, Loo Y-M, Gale M Jr, Núñez G et al (2015) RNase L acti-
vates the NLRP3 inflammasome during viral infections. Cell Host Microbe 17(4):466–477
Chen N, Xia P, Li S, Zhang T, Wang TT, Zhu J (2017) RNA sensors of the innate immune system
and their detection of pathogens. IUBMB Life 69(5):297–304
Chow KT, Gale M Jr, Loo Y-M (2018) RIG-I and other RNA sensors in antiviral immunity. Annu
Rev Immunol 36:667–694
Crozat K, Beutler B (2004) TLR7: a new sensor of viral infection. Proc Natl Acad Sci 101(18):
6835–6836
Dang W, Xu L, Yin Y, Chen S, Wang W, Hakim MS et al (2018) IRF-1, RIG-I and MDA5 display
potent antiviral activities against norovirus coordinately induced by different types of interfer-
ons. Antivir Res 155:48–59
Dauber B, Wolff T (2009) Activation of the antiviral kinase PKR and viral countermeasures.
Viruses 1(3):523–544
Diamond MS (2014) IFIT1: a dual sensor and effector molecule that detects non-2′-O methylated
viral RNA and inhibits its translation. Cytokine Growth Factor Rev 25(5):543–550
Gantier MP, Williams BR (2011) Making sense of viral RNA sensing. Mol Ther 19(9):1578–1581
Gil J, Esteban M (2000) Induction of apoptosis by the dsRNA-dependent protein kinase (PKR):
mechanism of action. Apoptosis 5(2):107–114
Hewson CA, Jardine A, Edwards MR, Laza-Stanca V, Johnston SL (2005) Toll-like receptor 3
is induced by and mediates antiviral activity against rhinovirus infection of human bronchial
epithelial cells. J Virol 79(19):12273–12279
Jensen S, Thomsen AR (2012) Sensing of RNA viruses–a review on innate immune receptors
involved in recognizing RNA virus invasion. J Virol 86(6):2900–2910
Jurk M, Heil F, Vollmer J, Schetter C, Krieg AM, Wagner H et al (2002) Human TLR7 or TLR8
independently confer responsiveness to the antiviral compound R-848. Nat Immunol 3(6):499
Liu HM, Gale M Jr (2011) ZAPS electrifies RIG-I signaling. Nat Immunol 12(1):11
Malathi K, Dong B, Gale M Jr, Silverman RH (2007) Small self-RNA generated by RNase L
amplifies antiviral innate immunity. Nature 448(7155):816
Patel MC, Shirey KA, Pletneva LM, Boukhvalova MS, Garzino-Demo A, Vogel SN et al (2014)
Novel drugs targeting Toll-like receptors for antiviral therapy. Futur Virol 9(9):811–829
Pichlmair A, Schulz O, Tan CP, Näslund TI, Liljeström P, Weber F et al (2006) RIG-I-mediated anti-
viral responses to single-stranded RNA bearing 5′-phosphates. Science 314(5801):997–1001
Rodriguez KR, Bruns AM, Horvath CM (2014) MDA5 and LGP2: accomplices and antagonists of
antiviral signal transduction. J Virol 88(15):8194–8200
Said EA, Tremblay N, Al-Balushi MS, Al-Jabri AA, Lamarre D (2018) Viruses seen by our cells:
the role of viral RNA sensors. J Immunol Res 2018:9480497
Sarkar SN (2014) Could boosting the oligoadenylate synthetase-like pathway bring a new era of
antiviral therapy? Futur Virol 9(12):1011–1014
242 P. Nair and S. U. Sapre
Silverman RH (2007) Viral encounters with 2′,5′-oligoadenylate synthetase and RNase L during
the interferon antiviral response. J Virol 81(23):12720–12729
Thomas A, Laxton C, Rodman J, Myangar N, Horscroft N, Parkinson T (2007) Investigating
Toll-like receptor agonists for potential to treat hepatitis C virus infection. Antimicrob Agents
Chemother 51(8):2969–2978
Vladimer GI, Górna MW, Superti-Furga G (2014) IFITs: emerging roles as key anti-viral proteins.
Front Immunol 5:94
Yoneyama M, Fujita T (2007) Function of RIG-I-like receptors in antiviral innate immunity. J Biol
Chem 282(21):15315–15318
Zhu R, Zhang Y-B, Zhang Q-Y, Gui J-F (2008) Functional domains and the antiviral effect of
the double-stranded RNA-dependent protein kinase PKR from Paralichthys olivaceus. J Virol
82(14):6889–6901
Zou J, Chang M, Nie P, Secombes CJ (2009) Origin and evolution of the RIG-I like RNA helicase
gene family. BMC Evol Biol 9(1):85(Continued)
Potentiality of DNA Sensors
in Activating Immune System 16
in Emerging Viral Infectious Diseases
Abstract
Viruses are obligatory intracellular parasites and hijack the host cell machinery
to make more identical copies of it and continue self-propagation. They attach
and replicate in the susceptible and permissive hosts and host derived cell lines.
They enter the cells either through direct attachment, receptor-mediated endocy-
tosis, or phagocytosis. Hence, to thwart the invasion by viruses, hosts have devel-
oped immunity in ascending stages—intrinsic, innate and adaptive immunity. A
robust intrinsic and innate immune response governs an effective adaptive
immune response, should that be needed. Both enveloped as well as non-
enveloped viruses are subject to distinct types of DNA sensors, subject to their
site of replication. DNA sensors of viral PAMPs can be classified into three
types, based on the location of their PAMPs in the host cellular compartment viz.
cell surface, cytoplasmic and nuclear. The host cell membrane both, surface as
well as intra cellular, is continuously monitored for the non-host, pathogenic
components or PAMPs. Among the intracellular sensors of the viral genome,
there are two types—essentially due to the two types of major viral genomes i.e.
RNA and DNA sensors. The cytosolic DNA sensors include AIM2, IFI16, cGAS,
RNA Pol III, DNA-PK, DDX9, DHX36, DDX41, DDX60, DAI, LRRFIP1,
HMGB, ABCF1 and MRE11. PYHIN family of sensors include AIM2, IFI16,
IFIX and MNDA. Another recently discovered family of sensor called stimulator
of interferon (IFN) genes (STING), specifically houses on the endoplasmic retic-
ulum (ER) and functions in association with its upstream sensor, cGAS. Some
DNA sensors shuttle between the cytosol and nucleus pre- and post-extraneous
DNA binding. These include IFI16, IFIX, RNA Pol III, etc. There is no exclusive
nuclear DNA sensor. Many enzymes known to be present in the cells for their
S. U. Sapre
National Institute of Virology, Pune, India
P. Nair (*)
NMIMS School of Science, Mumbai, India
Keywords
Viral DNA sensors · Immune responses · Immune activation
16.1 Introduction
Viral PAMPS
Cellular
Soluble (blood)
ssRNA of dsRNA of dsDNA of
Mucosal surfaces viruses viruses viruses
(sectretions)
Cell-associated molecules are mostly restricted to the cells of the immune sys-
tem like phagocytes—Macrophages and Neutrophils; antigen presenting cells
(APCs)—Dendritic cells; cells that form the obstacle between the internal milieu
and the external environment—epithelial cells; as well as other cells like mast cells
and tissue resident cells.
Pathogens like viruses, bacteria, fungi and protozoa can establish themselves
within any compartment of the cell viz. cytosol, nucleus, endosomes or on the sur-
face of the cell, tissue, etc. The innate immune system efficiently tackles the micro-
bial invasion in all these compartments by activating the signal transduction
pathways downstream of the recognition molecules which ultimately promote the
pro-inflammatory and anti-microbe activity.
Broadly though, the pattern recognition receptors can be either cell-associated or
soluble.
The cell-associated receptors include:
1. Pentraxins
2. Collectins
3. Ficolins
4. Complement
246 S. U. Sapre and P. Nair
The scope of discussion on each of the above PRRs is wide. However, since this
chapter specifically deals with the DNA-sensing molecules, further discussion
would be restricted to the viral DNA sensors.
There are several ways by which DNA can be present in the cell cytoplasm.
These routes include (but are not limited to):
Site of DNA
PRR Cell types sensing Response References
Toll-like receptor (TLR) family
TLR9 pDCs Endosomes Type I IFN (Hemmi et al. 2000;
Latz et al. 2004, 2007)
PYHIN family
AIM2 Macrophages, Cytoplasm IL-1β, IL-18 (Hornung et al. 2009;
DCs Fernandes-Alnemri
et al. 2009;
Burckstummer et al.
2009; Roberts et al.
2009)
IFI16 Macrophages, Cytoplasm, IFN-β, CXCL10, (Unterholzner et al.
endothelial cells nucleus IL-6, IL-1β 2010; Horan et al. 2013;
Kerur et al. 2011)
IFIX Macrophages Nucleus IL-1β, IL-18 (Diner et al. 2015a)
MNDA Less explored
STING activator family
cGAS L929, THP-1, Cytoplasm IFN-β (Sun et al. 2013)
HEK293
DNAse family
DNAse II Ubiquitous Lysosomes DNA degradation (Okabe et al. 2005)
TREX1 Ubiquitous Cytoplasm-ER Degradation of (Stetson et al. 2008;
associated DNA elements Yang et al. 2007)
derived from
endogenous
retroviruses
RNAse family
16 Potentiality of DNA Sensors in Activating Immune System in Emerging Viral… 247
Site of DNA
PRR Cell types sensing Response References
RNA Pol EBV+ B cell, Cytoplasm, IFN-β (Ablasser et al. 2009;
III macrophage, nucleus Chiu et al. 2009)
cell line
Protein kinase (PK) family
DNA-PK 293T, MEFs Cytoplasm IFN-λ1, IFN-β, (Zhang et al. 2011a;
IL-6 Ferguson et al. 2012)
DExD/H-box helicase family
DDX9 pDCs Cytoplasm TNF-α (Kim et al. 2010)
DHX36 pDCs Cytoplasm IFN-α (Kim et al. 2010)
DDX41 DCs Cytoplasm IFN-α, β (Zhang et al. 2011b)
DDX60 HeLa cells Cytoplasm IFN-β, CXCL10 (Miyashita et al. 2011)
Others/less characterised family
DAI Fibroblasts Cytoplasm IFN-β, necrosis (Takaoka et al. 2007;
Upton et al. 2012)
LRRFIP1 Less studied Cytoplasm Type I IFN (Sabbah et al. 2009;
HMGB Cytoplasm induction Yanai et al. 2009; Yang
et al. 2010)
ABCF1 Cytoplasm Less known
MRE11 MEFs, DCs Cytoplasm IFN-β, CXCL10, (Kondo et al. 2013)
IL-6
16.3.1 Structure
TLRs belong to the type I integral membrane glycoproteins embedded in the surface
membrane or in the endosomal membrane. They possess an extracellular (or intra-
endosomal) region—characteristic leucine-rich repeats (LRRs) which are sur-
rounded by cysteine-rich motifs which essentially bind ligand(s), a transmembrane
region, and an intracellular (or cytosolic) region—known as the Toll/IL-1 receptor
(TIR) involved as a part of the cytoplasmic tail. There are about 18–26 copies of
LRRs which vary in different TLRs. Each LRR of TLR protein is composed of
around 20–25 amino acids and multiple such LRRs make up a typical question mark
hook shaped protein scaffold which adapts for ligand binding. Both, the concave as
well as convex surfaces, are involved in ligand binding. The N-terminus of a TLR is
at its LRR end, whereas the C-terminus is at its TIR end.
16.3.2 Function
There are nine different functional TLRs in humans (TLR1-9). The function of
TLR-10 is unknown. TLRs always act as dimers. Some of them form heterodimers
like TLR-1 and TLR-2, whereas others form homodimers like TLR-3, TLR-4,
248 S. U. Sapre and P. Nair
TLR-5, TLR-5, TLR-6, TLR-7, TLR-8, TLR-9 and TLR-10. TLR-1, 2, 4, 5 and 6
are expressed on the surface of the membrane, whereas TLR-3, 7, 8 and 9 are
expressed inside the endosomal membrane. TLR-4 utilises accessory proteins MD2,
LPS-binding protein (LBP) and CD14.
PAMPs
TLR activation
Ligand binding to the TLR brings about dimerization of the respective TLRs
involved. This brings about the cytoplasmic tails to come in proximity. TIR domain
containing adaptor proteins are now recruited, which bind the TIR domains in the
cytoplasmic tails. Further, this brings about recruitment and activation of distinct
protein kinases, which activate the transcription factors. Nuclear factor κ-B (NF-
κB), activation protein-1 (AP-1), interferon response factor 3 (IRF3) and IRF7 are
the major transcription factors which are activated by TLR signalling pathways.
NF-κB and AP-1 are responsible for the expression of genes encoding inflammatory
response molecules, including (but not limited to) inflammatory cytokines, endothe-
lial adhesion molecules and chemokines. IRF3 and IRF7 stimulate the production
type I interferons (IFNs), which are central to antiviral innate immune responses.
Different TLRs use different combination of adaptors and signalling intermediates
and thus mediate unique downstream effects. TLR9 uses the MyD88-dependent,
TRIF-independent pathway and activates both NF-κB and IRFs. Hence, they induce
both inflammatory and antiviral responses.
Two protein domains of MyD88 allow it to function as an adaptor protein: a TIR
domain at its carboxy terminus that associates with the TIR domains of the TLR
16 Potentiality of DNA Sensors in Activating Immune System in Emerging Viral… 249
cytoplasmic tails and a death domain at its amino terminus, which associates with
the death domains present in other intracellular signalling proteins. It is worth-
while to note that both the domains of MyD88 are essential for signalling. The
MyD88 death domain recruits and activates IL-1 receptor associated kinase
(IRAK4) and IRAK1, which are both serine-threonine protein kinases. The com-
plex involving IRAK, TIR of cytoplasmic tails and MyD88 executes two func-
tions—executing the enzymes that produce a signalling scaffold, employing the
scaffold to recruit other molecules which are then phosphorylated by the IRAKs.
The formation of signalling scaffold is a multi-step process: The IRAK complex
brings in the enzyme tumour necrosis factor receptor-associated factor 6 (TRAF6),
which functions as an E3 ubiquitin ligase in association with TRIKA1 (composed
of UBC13, which is an E2 ubiquitin ligase with a cofactor for Uve1A). TRAF6 and
UBC13 together have the function of polyubiquitination. This polyubiquitin has
linkages between lysine63 of pervious subunit and C terminus of the next, leading
to K63 linkages. The same process can be initiated on vivid proteins, including
TRAF6 or the multi-ubiquitin chains can exist independently as free linear chains
which can be extended to form polyubiquitin chains, which can bind to other sig-
nalling proteins. Further, the scaffold brings in signalling complex that is made up
of TAB1, TAB2-ubiquitin binding adaptor molecules, and TAK1-a serine-threo-
nine kinase. IRAK complex phosphorylates TAK1, which propagates signalling by
activation of certain MAPKs like c-Jun terminal kinase (JNK) and MAPK14 (p38
MAPK). This brings about downstream activation of AP-1 family of transcription
factors which transcribe cytokine genes.
TAK1 additionally phosphorylates and activates the IκB kinase (IKK) complex.
IKKα, IKKβ and IKKγ constitute the IKK complex (the IKKγ is also known as
NF-Κb essential modifier or NEMO). NEMO binds to polyubiquitin chains, which
leads to IKK complex close to TAK1. TAK1 then activates IKKβ by its phosphory-
lation. IKKβ then phosphorylates inhibitor of κB (IκB), which is a cytoplasmic
protein that natively binds to the transcription factor NF-κB. IκB is made up of two
subunits—p50 and p65. The binding of IκB prevents the movement of NF-κB to the
nucleus from cytoplasm. Post phosphorylation of IKK, the IκB is released from the
functional subunit of NF-κB, which leads to translocation of NF-κB to nucleus,
where it leads to transcription of genes for pro-inflammatory cytokines like TNF-α,
IL-1β and IL-6. It is noteworthy that the effect of TLR activation varies depending
on the cell type in which it occurs.
The nucleic acid sensing TLRs, including TLR9 activate IRF family of proteins.
Natively present in the cytoplasm, they only get activated upon phosphorylation of
serine and threonine residues in their C terminal. Upon activation, they move to the
nucleus to act as transcription factors. Among all the IRFs, IRF3 and IRF7 are par-
ticularly important for TLR signalling and expression of antiviral type I IFNs. For
TLR9 signalling in plasmacytoid dendritic cells, MyD88 exclusively is used as an
adaptor protein. The TIR domain of MyD88 employs IRAK1/IRAK4 complex as
described earlier. However, the IRAK complex carries out a different function
beyond recruiting TRAFs which generates a signalling scaffold. In these cells,
IRAK1 can also interact with IRF7, which is highly expressed by plasmacytoid
250 S. U. Sapre and P. Nair
dendritic cells. This enables IRAK1 to phosphorylate IRF7 which leads to induction
of type I IFNs (Table 16.1).
There are 4 PYHIN proteins in humans, and 13 in mice. Two human proteins in this
class, namely AIM2 and IFN-γ inducible (IFI16) have been predicted based on the
studies so far, to be necessary for different DNA-modulated immune responses and
perhaps, also function as DNA sensors (Hornung et al. 2009; Roberts et al. 2009;
16 Potentiality of DNA Sensors in Activating Immune System in Emerging Viral… 251
Unterholzner et al. 2010). PYHIN family of proteins can also activate the formation
of inflammasome.
The PYHIN family proteins contain an N-terminal pyrin (PY) domain and an H
inversion (HIN) domain
16.4.1.1 AIM2
Absent in melanoma (AIM2) is one such example of a PYHIN family of proteins.
The HIN region of AIM2 recognises dsDNA genome and triggers caspase 1 activa-
tion. This occurs through the simultaneous interaction of pyrin domain with
ASC. Primarily AIM2 is involved in the inflammasome formation (Schattgen and
Fitzgerald 2011). Microbial DNA is one such PAMP. It is usually located in the
cytosol and is key to responses in vitro to vaccinia virus (Janeway). However, it
ubiquitously oligomerises upon stimulation. The downstream aggregates so formed
recruit and activate a protease-caspase-1 that leads to the maturation of pro-
inflammatory cytokines IL-1β and IL-18 which ultimately culminates into a pro-
grammed cell death, termed ‘pyroptosis’ (Bergsbaken et al. 2009; Miao et al. 2011).
16.4.1.2 IFI16
Interferon-inducible protein 16 (IFI16) was recently characterised as the first viral
DNA sensor to function within the nucleus. It is a member of PYHIN protein fam-
ily (Diner et al. 2015a; Schattgen and Fitzgerald 2011). IFI16 has two HIN
domains. At its C terminus, it has two HIN200 domains which bind to DNA. At its
N-terminal, is a pyrin (PY) domain which mediates homotypic interactions within
the molecules as well as cooperative assembly of small subunits of IFI16 (Li et al.
2013; Morrone et al. 2014). This binding is sequence-independent manner (Jin
et al. 2012). It primarily functions in the nucleus, where it is located. It recognises
viral dsDNA.
The function of IFI16 depends on the type of cell in which it functions. In the
immune cells, IFI16 binds to cytosolic viral DNA, engaging sting and induces IFN
production (Unterholzner et al. 2010; Horan et al. 2013; Jakobsen et al. 2013).
However, in non-immune cells, IFI16 majorly functions in the nucleus by localising
252 S. U. Sapre and P. Nair
there (Diner et al. 2015a; Li et al. 2013, 2012; Orzalli et al. 2012). A plausible expla-
nation for this is that the there exists a multi-partite nuclear localisation signal on
IFI16 and is necessary for its transit between nucleus and cytoplasm (Li et al. 2012).
Especially in case of herpesvirus infections, both IFI16 and STING are required for
inducing the expression of IFN and IFN-stimulated genes (Orzalli et al. 2012). The
differences in IFI16 DNA-sensing are possibly due to the cell type-dependent pro-
cess (Diner et al. 2015b). Prompt responses are required in case of immune cells,
and thus, DNA-sensing components localised in the cytoplasm makes more sense,
whereas, in non-immune cells, the IFI16 might play some housekeeping functions
but additionally may respond to the successful viral infections in the nucleus.
Additionally, IFI16 also mounts inflammatory and apoptotic responses to foreign
DNA through inflammasome—a multiprotein assembly (Kerur et al. 2011; Johnson
et al. 2013; Ansari et al. 2013; Singh et al. 2013; Monroe et al. 2014). In reality, the
evidence that IFI16 elicits both, type I IFN response as well as inflammation is con-
tradictory, since type I IFNs are known to exhibit anti-inflammatory effect
(Theofilopoulos et al. 2005; Billiau 2006; Guarda et al. 2011). Recently, IFI16 was
shown to play a direct role in inhibiting the formation of both, AIM2 and NLR fam-
ily inflammasomes (Veeranki et al. 2011). Hence, it is quite possible that the role of
IFI16 in inflammasome responses is cell type dependent and other yet unknown
factors do play a role too. Recent evidences suggest that IFI16 and cGAS may func-
tion in harmony to execute immune signalling to nuclear foreign DNA (Orzalli et al.
2015). This also suggests that IFI16 is a dominant nuclear DNA sensor, whereas
cGAS has auxiliary functions like stabilising the IFI16 to enable or prolong signal
efficiency.
16.4.1.3 IFIX
Some of the PYHIN family members, namely, IFIX and MNDA also majorly local-
ise in the nucleus. Yet, their functions were not known in the early phase and were
suspected to have some immunological function. IFIX associates with antiviral fac-
tors and its expression is inversely associated with the capacity of herpesvirus rep-
lication (Diner et al. 2015a) as it binds to the DNA of the virus and remains localised
in the nucleus. It binds DNA substrates in a sequence-dependent fashion and leads
to type I IFN response (Diner et al. 2015a). IFIX expression, like IFI16, is depen-
dent on the type of cell and tissue, thus making it likely that its function varies from
one cell type to other (Ding et al. 2004; Haque et al. 2014).
viral pathogens. Stimulator of IFN genes (STING) is one such pathway vital to the
mechanism of dsDNA-induced stimulation of type I IFN responses. If a viral
dsDNA happens to exist in the cytosol, post entry and uncoating, it activates the
enzyme cGAS (cyclic GMP-AMP synthase) that generates cyclic guanosine
monophosphate-adenosine monophosphate (cGAMP), a signalling molecule. cGAS
contains a protein motif as a part of the nucelotidyltransferase (NTase) family of
enzymes, including adenylate cyclase and distinct DNA polymerases (Janeway).
cGAS has an affinity for DNA and readily attaches to the cytosolic DNA. This inter-
action stimulates the cGAS enzymatic activity that leads to generation of cGAMP
from GTP and ATP in the cytoplasm. cGAMP binds to both the subunits of STING
dimer and activates STING signalling.
It activates TANK binding kinase 1 (TBK1) through interaction with
cGAMP. TBK1 phosphorylates and activates a transcription factor IRF3 which
induces expression of type I IFN genes. Besides these, the STING is also known to
respond to other cytosolic DNA sensors like DAI and IFI16. Additionally, STING
induces autophagy. In the innate immune system, autophagy is a potential mecha-
nism of delivering cytosolic microbes to lysosome where they are acted upon by the
proteolytic enzymes.
It is quite interesting to know that STING, MAVS and TRIF, all have similar
amino acid sequence motif at their carboxy termini
16.6.1 DNAse II
Cells possess DNAses, which degrade the unwanted DNA in the compartment it is
not expected to be present. DNAse II is restricted to lysosomes which digests patho-
genic and by-products of dead cells which enter this compartment (Okabe et al.
2005). Originally, this mechanism has evolved for the degradation of the cells
undergoing programmed cell death, i.e. apoptosis, which are usually taken up by
macrophages. In cells which lack DNAse II activity, DNA may stimulate aberrant
responses by entering cytoplasm, consequently stimulating the cytosolic DNA sen-
sors (Okabe et al. 2005).
16.6.2 TREX1
Yet another cellular DNAse is three primer repair endonuclease 1(TREX1) that is
present in association with the endoplasmic reticulum (ER) in the cytoplasm. Under
housekeeping conditions, basal amount of DNA tends to accumulate in the cytosol.
TREX1 regularly degrades such DNA in the cytosol.
254 S. U. Sapre and P. Nair
Another putative cytosolic DNA sensor discovered is RNA Polymerase III (PolIII).
It uses AT-rich and herpesvirus DNA as a template to produce 5′triphosphate RNAs.
RNAs so produced get detected by the cytosolic RNA sensors like RIG-I (Ablasser
et al. 2009; Chiu et al. 2009). However, the broad mechanism being the Pol III as a
potential DNA senor remains to be completely defined.
Some proteins which are usually involved in DNA damage repair also serve as DNA
sensors. DNA-PK (DNA-dependent protein kinase which is composed of Ku70,
Ku80 and DNA-PKc).
Both, RNA, as well as DNA helicases constitute the DExD/H-box (DDX) protein
family which has it characteristic DExD/H-box domain. DDXs are multi-functional
and control the gene induction at multiple points that includes signal transduction
pathways, gene promoters, mRNA splicing, and translation regulation. Several
DNA sensors have been identified in the DDX family (Kim et al. 2010; Zhang et al.
2011b; Yoneyama et al. 2004; Schroder et al. 2008).
DHX9 and DHX36 are the other two DExD/H-box helicases which bind to CpG
DNA and interact with MyD88 (Kim et al. 2010). The inflammatory response and
type I interferon response depend partly on DHX9 and DHX36, respectively.
16.9.2 DDX41
directly bind the cyclic di-nucleotides (CDNs) and thus, indirectly, the IFN response
induced was DDX41 dependent (Parvatiyar et al. 2012)
16.9.3 DDX60
It is a novice antiviral factor among the pre-existing list of DExD/H box helicases
and functions in association with RIG-I, MDA5 and LGP2 to induce the type I IFN
response (Miyashita et al. 2011).
There are many unknown and known but less characterised candidate DNA sensors.
They might play some vital role in other cellular processes, but their role as a DNA
sensor is yet to be confirmed. Very less is known regarding their mechanism of rec-
ognition and signalling, or their in vivo function.
16.10.1 DAI
Another protein is DAI (a.k.a. ZBP1) (Takaoka et al. 2007). The peculiarity of this
protein is that the response of DAI as a DNA sensor is decided by the cell type
(Unterholzner et al. 2010; Upton et al. 2012, 2010; DeFilippis et al. 2010; Ishii
et al. 2008). It interacts with the dsDNA and drives type I IFN response (DeFilippis
et al. 2010).
16.10.2 LRRFIPI
It senses the DNA present in the cytoplasm and phosphorylates β-catenin. Β-catenin
translocates to the nucleus to induce IFN-β production.
16.10.3 HMGB
Three subtypes of HMGB, viz. HMGB1, HMGB2 and HMGB3 are known to
respond to cytosolic DNA. Primarily, ABCF1 binds to the cytosolic DNA. This
complex then binds to HMGB2 and IFI16 to stimulate further innate immune
response (Yanai et al. 2009; Yang et al. 2010; Lee et al. 2013; Goubau et al. 2013)
16.10.4 ABCF1
16.10.5 MRE11
There are numerous mechanisms through which the DNA sensors in the host cells
can detect the viral genomic components and trigger a cascade of downstream reac-
tions. Some of them have been discussed below:
16.11.1 Herpesviridae
Till date, the role of DNA sensors has been best studied extensively using herpesvi-
ruses, HSV, in particular (Paludan et al. 2011). Quite evidently, currently known-
well, established, as well as prospective DNA sensors have been shown to counteract
herpesvirus infections. The very primary DNA sensor was TLR9, followed by the
contemporary DNA sensors. To state a few, the HSV-1 infection in human origin
primary MDMs (monocyte-derived macrophages) induces the production of pro-
inflammatory cytokines like IL-1β, which imitates the response post HSV-1 infec-
tion along IFI16 axis (Horan et al. 2013). However, this response is altered when the
cell type differs. HSV-1 infection of primary murine dendritic cells (DCs) invokes a
type I IFN response with the protein DDX41 involved (Zhang et al. 2011b). IRF3 is
known to drive the type I IFN response as well. DAI drives IRF3 activation as well.
However, contrastingly, DAI (or ZBP1) is shown to play a central role in inducing
necroptosis, post infection with MCMV (Upton et al. 2012).
As discussed earlier, STING is central to all the DNA induced antiviral responses.
This coincides with the study which demonstrated that STING deficiency in mice
causes increased susceptibility to HSV-1 infection (Ishikawa et al. 2009). While it is
not known if the cell-specific response applies to all the cell-lines, the above docu-
mented work irrefutably states that the response to herpesvirus infections in primary
cells is cell type specific.
16.11.2 Retroviridae
HIV is the most widely studied retrovirus for known reasons on its complex patho-
genesis, drug resistance and co-evolution with the host. As is known, HIV replica-
tion proceeds through and RNA–DNA double-stranded intermediate in the process
of its RNA being converted to a dsDNA form. Hence, both, RNA as well as DNA
sensors, play a crucial role in the detection of retroviral genome upon target cell
entry (Solis et al. 2011; Berg et al. 2012; Doitsh et al. 2010; Yan et al. 2010). A
recent study, intended to understand the type I IFN response against HIV proved
16 Potentiality of DNA Sensors in Activating Immune System in Emerging Viral… 257
that ssDNA, rather than dsDNA form of HIV, is more potent at inducing the type I
IFN response. TREX1 is a 3′5′-exonuclease that degrades the unintegrated cyto-
solic cDNA, making it unavailable for sensing to the DNA sensors. Eliminating the
expression of TREX1 leads to upregulation in type I IFN response and was more so
in case of ssRNA intermediate of HIV, rather than dsDNA form. Undoubtedly, thus,
cell does employ the intracellular sensors of DNA that culminates in type I IFN
response. Although the definite sensors involved remain to be defined to date, they
sure function through a STING-TBK1-IRF3 axis.
Other virus families and the respective DNA sensors have been summarised in
Table 16.2
16.11.3 M
echanisms Employed By Viruses to Evade DNA
Sensors
Viral genomes are subject to recognition by DNA sensors only if they happen to
expose their characteristic features, leading to the consequent recognition as non-
self genomic entity by the host DNA sensing mechanisms. However, myriad viruses
evade and subvert the host immune responses so as to render these mechanisms of
detecting the viral components ineffective.
One such strategy is to simply obscure their genome and replication intermedi-
ates involving potential ligand to the DNA sensors:
Viruses belonging to the Herpesviridae family inhibit DNA sensors like DAI,
DHX9 and IFI16 responses.
Table 16.2 Human nuclear replicating DNA viruses and implicated DNA sensors
Virus Implicated DNA sensors
Adenovirus C (types 1, 2, 5 and cGAS, TLR9
6)
Hepatitis B virus AIM2, cGAS
Cytomegalovirus AIM2, DAI/ZBP1, IFI16, TLR7, TLR9
Epstein–Barr virus RNA Pol III, IFI16, TLR9
Herpes simplex virus type I cGAS, DAI/ZBP1, DDX41, DDX60, DHX9, DHX36,
DNA-PK, RNA Pol III, IFI16, TLR9, IFIX
Herpes simplex virus type II DNA-PK, TLR9
Herpesvirus 8 (Kaposi sarcoma- IFI16
associated virus)
Varicella zoster virus NLRP3, TLR9
Papillomavirus (>170 types) TLR9
258 S. U. Sapre and P. Nair
3. Proteases of viral origin like the NS2B3 protease, coronavirus papain-like prote-
ases subvert STING in a direct manner (Aguirre et al. 2012; Sun et al. 2012; Yu
et al. 2012).
Acknowledgements Authors are grateful to NIV Pune and NMIMS Pune for the support
extended.
Conflict of Interest The authors declare that they have no competing interests.
References
Ablasser A et al (2009) RIG-I-dependent sensing of poly(dA:dT) through the induction of an RNA
polymerase III-transcribed RNA intermediate. Nat Immunol 10(10):1065–1072
Aguirre S et al (2012) DENV inhibits type I IFN production in infected cells by cleaving human
STING. PLoS Pathog 8(10):e1002934
Ansari MA et al (2013) Constitutive interferon-inducible protein 16-inflammasome activation dur-
ing Epstein-Barr virus latency I, II, and III in B and epithelial cells. J Virol 87(15):8606–8623
Berg RK et al (2012) Genomic HIV RNA induces innate immune responses through RIG-I-
dependent sensing of secondary-structured RNA. PLoS One 7(1):e29291
Bergsbaken T, Fink SL, Cookson BT (2009) Pyroptosis: host cell death and inflammation. Nat Rev
Microbiol 7(2):99–109
Billiau A (2006) Anti-inflammatory properties of Type I interferons. Antivir Res 71(2-3):108–116
Burckstummer T et al (2009) An orthogonal proteomic-genomic screen identifies AIM2 as a cyto-
plasmic DNA sensor for the inflammasome. Nat Immunol 10(3):266–272
Chiu YH, Macmillan JB, Chen ZJ (2009) RNA polymerase III detects cytosolic DNA and induces
type I interferons through the RIG-I pathway. Cell 138(3):576–591
Cristea IM et al (2010) Human cytomegalovirus pUL83 stimulates activity of the viral immediate-
early promoter through its interaction with the cellular IFI16 protein. J Virol 84(15):7803–7814
DeFilippis VR et al (2010) Human cytomegalovirus induces the interferon response via the DNA
sensor ZBP1. J Virol 84(1):585–598
Diner BA et al (2015a) The functional interactome of PYHIN immune regulators reveals IFIX is a
sensor of viral DNA. Mol Syst Biol 11(1):787
Diner BA, Lum KK, Cristea IM (2015b) The emerging role of nuclear viral DNA sensors. J Biol
Chem 290(44):26412–26421
Ding Y et al (2004) Antitumor activity of IFIX, a novel interferon-inducible HIN-200 gene, in
breast cancer. Oncogene 23(26):4556–4566
Doitsh G et al (2010) Abortive HIV infection mediates CD4 T cell depletion and inflammation in
human lymphoid tissue. Cell 143(5):789–801
Ferguson BJ et al (2012) DNA-PK is a DNA sensor for IRF-3-dependent innate immunity. elife
1:e00047
Fernandes-Alnemri T et al (2009) AIM2 activates the inflammasome and cell death in response to
cytoplasmic DNA. Nature 458(7237):509–513
Goubau D, Deddouche S, Reis e Sousa C (2013) Cytosolic sensing of viruses. Immunity 38(5):
855–869
Guarda G et al (2011) Type I interferon inhibits interleukin-1 production and inflammasome acti-
vation. Immunity 34(2):213–223
16 Potentiality of DNA Sensors in Activating Immune System in Emerging Viral… 259
Haque A et al (2014) Mouse pyrin and HIN domain family member 1 (pyhin1) protein posi-
tively regulates LPS-induced IFN-beta and NO production in macrophages. Innate Immun
20(1):40–48
Hemmi H et al (2000) A toll-like receptor recognizes bacterial DNA. Nature 408(6813):740–745
Horan KA et al (2013) Proteasomal degradation of herpes simplex virus capsids in macrophages
releases DNA to the cytosol for recognition by DNA sensors. J Immunol 190(5):2311–2319
Hornung V et al (2009) AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating
inflammasome with ASC. Nature 458(7237):514–518
Ishii KJ et al (2008) TANK-binding kinase-1 delineates innate and adaptive immune responses to
DNA vaccines. Nature 451(7179):725–729
Ishikawa H, Ma Z, Barber GN (2009) STING regulates intracellular DNA-mediated, type I
interferon-dependent innate immunity. Nature 461(7265):788–792
Jakobsen MR et al (2013) IFI16 senses DNA forms of the lentiviral replication cycle and controls
HIV-1 replication. Proc Natl Acad Sci U S A 110(48):E4571–E4580
Jin T et al (2012) Structures of the HIN domain: DNA complexes reveal ligand binding and acti-
vation mechanisms of the AIM2 inflammasome and IFI16 receptor. Immunity 36(4):561–571
Johnson KE, Chikoti L, Chandran B (2013) Herpes simplex virus 1 infection induces activation
and subsequent inhibition of the IFI16 and NLRP3 inflammasomes. J Virol 87(9):5005–5018
Kerur N et al (2011) IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in
response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe 9(5):363–375
Kim T et al (2010) Aspartate-glutamate-alanine-histidine box motif (DEAH)/RNA helicase A heli-
cases sense microbial DNA in human plasmacytoid dendritic cells. Proc Natl Acad Sci U S A
107(34):15181–15186
Kondo T et al (2013) DNA damage sensor MRE11 recognizes cytosolic double-stranded DNA
and induces type I interferon by regulating STING trafficking. Proc Natl Acad Sci U S A
110(8):2969–2974
Latz E et al (2004) TLR9 signals after translocating from the ER to CpG DNA in the lysosome.
Nat Immunol 5(2):190–198
Latz E et al (2007) Ligand-induced conformational changes allosterically activate Toll-like recep-
tor 9. Nat Immunol 8(7):772–779
Lee MN et al (2013) Identification of regulators of the innate immune response to cytosolic DNA
and retroviral infection by an integrative approach. Nat Immunol 14(2):179–185
Li T et al (2012) Acetylation modulates cellular distribution and DNA sensing ability of interferon-
inducible protein IFI16. Proc Natl Acad Sci U S A 109(26):10558–10563
Li T, Chen J, Cristea IM (2013) Human cytomegalovirus tegument protein pUL83 inhibits IFI16-
mediated DNA sensing for immune evasion. Cell Host Microbe 14(5):591–599
Miao EA, Rajan JV, Aderem A (2011) Caspase-1-induced pyroptotic cell death. Immunol Rev
243(1):206–214
Miyashita M et al (2011) DDX60, a DEXD/H box helicase, is a novel antiviral factor promoting
RIG-I-like receptor-mediated signaling. Mol Cell Biol 31(18):3802–3819
Monroe KM et al (2014) IFI16 DNA sensor is required for death of lymphoid CD4 T cells abort-
ively infected with HIV. Science 343(6169):428–432
Morrone SR et al (2014) Cooperative assembly of IFI16 filaments on dsDNA provides insights
into host defense strategy. Proc Natl Acad Sci U S A 111(1):E62–E71
Okabe Y et al (2005) Toll-like receptor-independent gene induction program activated by mam-
malian DNA escaped from apoptotic DNA degradation. J Exp Med 202(10):1333–1339
Orzalli MH, DeLuca NA, Knipe DM (2012) Nuclear IFI16 induction of IRF-3 signaling during
herpesviral infection and degradation of IFI16 by the viral ICP0 protein. Proc Natl Acad Sci U
S A 109(44):E3008–E3017
Orzalli MH et al (2015) cGAS-mediated stabilization of IFI16 promotes innate signaling during
herpes simplex virus infection. Proc Natl Acad Sci U S A 112(14):E1773–E1781
Paludan SR et al (2011) Recognition of herpesviruses by the innate immune system. Nat Rev
Immunol 11(2):143–154
260 S. U. Sapre and P. Nair
Parvatiyar K et al (2012) The helicase DDX41 recognizes the bacterial secondary messengers
cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune response. Nat
Immunol 13(12):1155–1161
Roberts TL et al (2009) HIN-200 proteins regulate caspase activation in response to foreign cyto-
plasmic DNA. Science 323(5917):1057–1060
Sabbah A et al (2009) Activation of innate immune antiviral responses by Nod2. Nat Immunol
10(10):1073–1080
Schattgen SA, Fitzgerald KA (2011) The PYHIN protein family as mediators of host defenses.
Immunol Rev 243(1):109–118
Schroder M, Baran M, Bowie AG (2008) Viral targeting of DEAD box protein 3 reveals its role in
TBK1/IKKepsilon-mediated IRF activation. EMBO J 27(15):2147–2157
Singh VV et al (2013) Kaposi's sarcoma-associated herpesvirus latency in endothelial and B cells
activates gamma interferon-inducible protein 16-mediated inflammasomes. J Virol 87(8):
4417–4431
Solis M et al (2011) RIG-I-mediated antiviral signaling is inhibited in HIV-1 infection by a
protease-mediated sequestration of RIG-I. J Virol 85(3):1224–1236
Stetson DB et al (2008) Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell 134(4):587–598
Sun L et al (2012) Coronavirus papain-like proteases negatively regulate antiviral innate immune
response through disruption of STING-mediated signaling. PLoS One 7(2):e30802
Sun L et al (2013) Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I
interferon pathway. Science 339(6121):786–791
Takaoka A et al (2007) DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate
immune response. Nature 448(7152):501–505
Theofilopoulos AN et al (2005) Type I interferons (alpha/beta) in immunity and autoimmunity.
Annu Rev Immunol 23:307–336
Unterholzner L et al (2010) IFI16 is an innate immune sensor for intracellular DNA. Nat Immunol
11(11):997–1004
Upton JW, Kaiser WJ, Mocarski ES (2010) Virus inhibition of RIP3-dependent necrosis. Cell Host
Microbe 7(4):302–313
Upton JW, Kaiser WJ, Mocarski ES (2012) DAI/ZBP1/DLM-1 complexes with RIP3 to mediate
virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell
Host Microbe 11(3):290–297
Veeranki S et al (2011) IFI16 protein mediates the anti-inflammatory actions of the type-I interfer-
ons through suppression of activation of caspase-1 by inflammasomes. PLoS One 6(10):e27040
Yan N et al (2010) The cytosolic exonuclease TREX1 inhibits the innate immune response to
human immunodeficiency virus type 1. Nat Immunol 11(11):1005–1013
Yanai H et al (2009) HMGB proteins function as universal sentinels for nucleic-acid-mediated
innate immune responses. Nature 462(7269):99–103
Yang YG, Lindahl T, Barnes DE (2007) Trex1 exonuclease degrades ssDNA to prevent chronic
checkpoint activation and autoimmune disease. Cell 131(5):873–886
Yang P et al (2010) The cytosolic nucleic acid sensor LRRFIP1 mediates the production of type I
interferon via a beta-catenin-dependent pathway. Nat Immunol 11(6):487–494
Yoneyama M et al (2004) The RNA helicase RIG-I has an essential function in double-stranded
RNA-induced innate antiviral responses. Nat Immunol 5(7):730–737
Yu CY et al (2012) Dengue virus targets the adaptor protein MITA to subvert host innate immunity.
PLoS Pathog 8(6):e1002780
Zhang X et al (2011a) Cutting edge: Ku70 is a novel cytosolic DNA sensor that induces type III
rather than type I IFN. J Immunol 186(8):4541–4545
Zhang Z et al (2011b) The helicase DDX41 senses intracellular DNA mediated by the adaptor
STING in dendritic cells. Nat Immunol 12(10):959–965
Advanced Immunotechnological
Methods for Detection and Diagnosis 17
of Viral Infections: Current Applications
and Future Challenges
Abstract
Keywords
RNA and DNA viral diseases · Diagnostics · Accuracy · Specificity · Sensitivity
17.1 Introduction
Viruses are obligate intracellular parasites and their detection and identification is
an imperative component clinically. Viral infectious signify a major portion in pub-
lic health perspectives with thousands of deaths annually. Notwithstanding from
highly contagious infections and serious pandemics to common influenza episodes,
clinical diagnosis of viral infection often relies on early detection. Therefore, effec-
tual detection of viruses is indispensable aid to avert transmission, initiate befitting
therapy, and scrutinize response to treatment which leads to effective disease con-
trol and management (Souf 2016).
Viral diagnosis is a dynamic process. Although prophylaxis is better than cure,
an accurate diagnosis of any virus fundamental to the infection is equally vital.
Generally, diagnostic tests are categorized into three groups: direct detection, iso-
lation of virus, and serology tests. Direct examination methods are merely faster
and examined directly for presence of virus particles, virus antigen, or viral
nucleic acids. Immunofluorescence assay is extensively used for rapid identifica-
tion of virus infections through detection of virus antigen or virus-specific anti-
bodies in clinical specimens. Viral diagnosis is a crucial revolution and renders
more accessible and makes it potential to standardize the recently developed diag-
nostic methods. Serology essentially comprises bulk of work of any virology
laboratory. A serological diagnosis can be identified by increasing titers of anti-
body among acute and convalescent stages of infection, or by the detection of
Immunoglobulin M (IgM).
The diagnosis of viral infections were enhanced noticeably all through 1990s,
with the onset of highly sensitive methods, viz. enzyme-linked immunosorbent
assay (ELISA) and PCR are superseded for this reason. Nevertheless, during the last
20 years this technique is being utilized, due to its unique nature, making diagnosis
probable through visualization of virus. Then after, introduction of ELISA then
revolutionized viral diagnosis by simplifying detection and shortening the time
(Torrance and Jones 1981). Molecular diagnostic tools for viral diagnosis has trialed
speedy advancements in last few years (Hayden and Persing 2001), and revolution-
ized diagnosis of infectious diseases, particularly viral diseases. The use of amplifi-
cation techniques, viz. PCR, RT-PCR, or NASBA (nucleic acid sequence-based
amplification) (Van Belkum and Niesters 1995) for detection of virus, genotyping,
and quantification have several advantages such as high reproducibility and sensi-
tivity, including broad dynamic range (Ebner et al. 2005, 2006). Several molecular
diagnostic techniques were recently swapped by fully automated devices that use
less time, maneuver smaller volumes of liquids, and provide quantified results with
improved accuracy. The current review emphasizes the advanced immunotech-
niques, how the specific characteristics of diagnostic methods revolutionized field
of viral diagnosis clinically from a decade.
17 Advanced Immunotechnological Methods for Detection and Diagnosis of Viral… 263
17.2.1.1 Microscopy
Transmission electron microscopy (TEM) is a merely imaging technique that per-
mits direct image of viruses attributable to its nanoscale resolution. This is a classi-
cal technique owing to its advantage of direct visualization of virus. Amid the 1960s
and 1990s, TEM contributed a breakthrough and doled out as a diagnostic tool for
recognizing numerous virus unswervingly in any biological samples. For that rea-
son, in modern years, role of TEM in diagnosis of anonymous infectious agents in
specific viral outbreaks or transmission clusters shifted from regular use to an initial
screening test. TEM is also considered an important method for assessment of viral
safety in biopharmaceutical products. Application of TEM understands the unknown
viruses for which there is an imminent risk of contamination, and TEM will undeni-
ably be functional for documenting the subsistence of viruses or virus-like particles
in these cells and the products derivative from them (Roingeard et al. 2019).
sampling methods. The sensitivity and specificity of this assay was comparable to
serum-based ELISAs. This salivary assay could improve our understanding of the
ecology and natural history of HEV (Pisanic et al. 2017).
Diagnosing ZIKV remains a great challenge, as detection of viral RNA is only
possible merely few days after onset of symptoms. Conversely, novel high-
throughput image-based fluorescent neutralization method for identification of
ZIKV was thoroughly evaluated and developed which reported higher sensitivity
than Plaque reduction neutralization test (PRNT) and MAC-ELISA, respectively.
This test might employ for clinical diagnosis, clinical trials, and confirmation and
seroprevalence studies of ZIKV infection (Koishi et al. 2018). In one of the recent
studies, detection of serum HEV antigen (Ag) is deemed to be sensitive and prom-
ising biomarker for HEV antigen diagnosis with HEV RNA in both acute and
chronic genotypes. Strikingly an antigen assay was recently evaluated for diagnos-
ing HEV genotypes with higher sensitivity than commercial anti-HEV IgM and
HEV RNA ELISA tests (Zhang et al. 2019). Nonetheless, recent studies on respira-
tory syncytial virus (RSV) developed Luciferase Immunoprecipitation Systems
(LIPS) assay to detect IgG Antibodies against Human RSV G-Glycoprotein.
Moreover, Human RSV G-Glycoprotein also acts as biomarker for natural expo-
sure or immunization. RSV genes encoding native and mutated G (mG) proteins
from subgroups A and B strains were cloned, expressed as luciferase-tagged pro-
teins, and experimented separately to spot anti-RSV-G specific IgG antibodies
employing a high-throughput luciferase immunoprecipitation system (LIPS-G). It
was pertinent to note that RSV monoclonal antibodies and polyclonal antisera
explicitly bound in LIPS-GA and/or -GB assays (Crim et al. 2019).
The diagnosis of (ZIKV) and dengue virus (DENV) infections against viral
envelope protein and nonstructural proteins (NS) was developed using flavivirus
multiplex microsphere immunoassay (MIA). However MIA could not differentiate
more recent from past infections, which represents a key diagnostic challenge;
therefore, in a most recent report an immunoglobulin G (IgG)-based avidity assay
was developed for its diagnostic performance to accurately differentiate between
recent ZIKA and past dengue virus infections. This assay was found useful in
patients with high risk of ZIKA complications, viz. pregnant women and monitor-
ing immune responses in vaccine trials (Furuya et al. 2019). Consecutively to
develop serological diagnosis of ZIKV-IgA and ZIKV-IgG, avidity assays were
evaluated to characterize ZIKA infections in want of viremia. These assay facili-
tated construed low avidity of IgG and IgA results, enhanced the serological diag-
nosis of ZIKV (Amaro et al. 2019). In another study, homologous proteins of diverse
flaviviruses exhibited high degrees of sequence uniqueness, mainly within sub-
groups. This led to prevalent immunological cross-reactivity. Therefore, a propor-
tional deconvolution of complex B cell responses against ZIKV and other flavivirus
were deliberated by screening with a microarray chip-based high-resolution sero-
logical analysis primed from overlapping peptides covering the whole amino acid
sequence of ZIKV genomic polyprotein was developed. Additionally with advent of
this assay several infections, viz. dengue, yellow fever, tick-borne encephalitis, and
West Nile viruses shall be diagnosed (Hansen et al. 2019).
17 Advanced Immunotechnological Methods for Detection and Diagnosis of Viral… 265
qRT-PCR and TCID50 (Qadiri et al. 2019). Development and evaluation of a new
one-step, real-time RT-PCR assay was developed for detecting latest H9N2 influ-
enza viruses competent of causing human infection. The sensitivity of one-step,
real-time RT-PCR assay was generally determined to be used in vitro transcribed
RNA, devoid of any cross-reactivity against RNA from H1–15 subtypes of influ-
enza viruses and other viral respiratory pathogens with no nonspecific reactions
(Saito et al. 2019).
Table 17.1 Advanced immunotechnological techniques for detection and diagnosis of viral
infections
Application to diagnosis of emerging
S. No. Techniques viral infections
1. Serological tests Diagnostic approach for chronic
hepatitis C infection, ZIKV, RSV,
DENV, tick-borne encephalitis, and
West Nile viruses
2. Enzyme assays (fluorescence polarization Identification of structural proteins of
immune assay (FPIA), microparticle immune Hendra and Nipah virus, Crimean–
assay (MEIA), chemiluminescent (CLIA) Congo hemorrhagic fever virus
(CCHFV), chikungunya
3. PCR-based immunodetection (RT-RPA, Detection of Ebola virus, Rabies virus,
RT-LAMP, RNA-ISH, RT-PCR, Trioplex Middle East respiratory syndrome
real-time PCR) coronavirus, hemorrhagic septicemia
virus, influenza viruses
4. Mass spectrometry (MS/MS) Prognosis of DENV antigen, ZIKV
5. miRNA-based immunodetection Circulating microRNA biomarkers in
detection of arboviruses; hepatitis
infection
6. Next-generation sequencing (NGS) Provides more exact and precise time
estimates of infection, crucial for
HIV-1 surveillance
7. Metagenomics (panpathogen metagenomic Diagnostic standard techniques used in
sequencing assay) diagnosis and genetic analysis of
influenza and other clinical respiratory
viruses
8. Immunosensors (sandwich-type Diagnostic test for HBV, swine flu
electrochemiluminescence (ECL) (H1N1) infection, H7N9 virus with
immunosensor) high sensitivity
9. Microfluidic technology Used to test the convenience of
neutralizing antibodies to explore
impact on virus–cell interactions
10. CRISPR/Cas system Prognosis of coxsackievirus, hepatitis
B virus, Zika virus
11. Nanoparticles-based immunodetection Nanoparticle-based lateral flow
immunoassay as point-of-care
diagnostic tool for infectious agents
and diseases
The most recently developed diagnostic viral techniques are redesigning the field
of clinical virology, which could contribute to reduction in incidence of serious
infectious viral diseases. The foremost advantages of molecular techniques are its
higher sensitivity and specificity matched up with other diagnostic methods, viz.
serological assays and culture methods, as well as its rapidity and possibility of
automation. Nevertheless, the technological capabilities alone are inadequate if
not sustained by health promotion policies to boost the consciousness a propos the
significance of early detection of infectious viral diseases outbreak and its spread.
In conclusion, good quality diagnosis has a cost that only developed countries can
afford in regular practice so far, and this is delaying the execution of new-fangled
methods in developing world and in disease endemic areas. Conversely, it is antic-
ipated that asserted efforts may persist toward developing new high-quality tests
inexpensive in low-income countries, which would considerably reinforce disease
control strategies.
Acknowledgments The authors are grateful to their respective academic institutions for the sup-
port extended.
17 Advanced Immunotechnological Methods for Detection and Diagnosis of Viral… 273
Conflict of Interest The authors declare that they have no competing interests.
References
Afsahi S, Lerner MB, Goldstein JM, Lee J, Tang X, Bagarozzi DA Jr, Goldsmith BR (2018) Novel
graphene-based biosensor for early detection of Zika virus infection. Biosens Bioelectron
100:85–88
Amaro F, Sánchez-Seco MP, Vázquez A, Alves MJ, Zé-Zé L, Luz MT, De Ory F (2019) The
application and interpretation of IgG avidity and IgA ELISA tests to characterize Zika virus
infections. Viruses 11(2):179
Azhar EI (2018) Development of fluorescent reverse transcription loop-mediated isothermal
amplification (RT-LAMP) using quenching probes for the detection of the Middle East respira-
tory syndrome coronavirus. J Virol Methods 258:41–48
Babamiri B, Hallaj R, Salimi A (2018) Ultrasensitive electrochemiluminescence immunosensor
for determination of hepatitis B virus surface antigen using CdTe@ CdS-PAMAM dendrimer
as luminescent labels and Fe3O4 nanoparticles as magnetic beads. Sensors Actuators B Chem
254:551–560
Banerjee R, Jaiswal A (2018) Recent advances in nanoparticle-based lateral flow immunoassay as
a point-of-care diagnostic tool for infectious agents and diseases. Analyst 143(9):1970–1996
Carlisle LA, Turk T, Kusejko K, Metzner KJ, Leemann C, Schenkel C, Yerly S (2019) Viral diver-
sity from next-generation sequencing of HIV-1 samples provides precise estimates of infection
recency and time since infection. J Infect Dis 220(2):254–265
Chang YF, Wang WH, Hong YW, Yuan RY, Chen KH, Huang YW, Wang SF (2018) Simple strat-
egy for rapid and sensitive detection of Avian influenza A H7N9 virus based on intensity-
modulated SPR biosensor and new generated antibody. Anal Chem 90(3):1861–1869
Chen KF, Rothman RE, Ramachandran P, Blyn L, Sampath R, Ecker DJ, Gaydos CA (2011) Rapid
identification viruses from nasal pharyngeal aspirates in acute viral respiratory infections by
RT-PCR and electrospray ionization mass spectrometry. J Virol Methods 173(1):60–66
Chiu CY, Coffey LL, Murkey J, Symmes K, Sample HA, Wilson MR, Stryke D (2017) Diagnosis
of fatal human case of St. Louis encephalitis virus infection by metagenomic sequencing,
California, 2016. Emerg Infect Dis 23(10):1964
Crim RL, Kumari S, Jayanti P, Audet S, Kulkarni A, Beeler J (2019) Development of luciferase
immunoprecipitation systems (LIPS) assay to detect IgG antibodies against human respiratory
syncytial virus G-glycoprotein. Vaccine 7(1):16
Deyde VM, Sampath R, Garten RJ, Blair PJ, Myers CA, Massire C, Klimov AI (2010) Genomic
signature-based identification of influenza A viruses using RT-PCR/electro-spray ionization
mass spectrometry (ESI-MS) technology. PLoS One 5(10):e13293
Draz MS, Shafiee H (2018) Applications of gold nanoparticles in virus detection. Theranostics
8(7):1985
Duchesne L, Njouom R, Lissock F, Tamko-Mella GF, Rallier S, Poiteau L, Pawlotsky JM (2017)
HCV Ag quantification as a one-step procedure in diagnosing chronic hepatitis C infection in
Cameroon: the ANRS 12336 study. J Int AIDS Soc 20(1):21446
Duncombe TA, Tentori AM, Herr AE (2015) Microfluidics: reframing biological enquiry. Nat Rev
Mol Cell Biol 16(9):554–567
Ebner K, Suda M, Watzinger F, Lion T (2005) Molecular detection and quantitative analysis of
the entire spectrum of human adenoviruses by a two-reaction real-time PCR assay. J Clin
Microbiol 43:3049–3053
Ebner K, Rauch M, Preuner S, Lion T (2006) Typing of human adenoviruses in specimens from
immunosupressed patients by PCR-fragment length analysis and real-time quantitative PCR. J
Clin Microbiol 44:2808–2815
274 P. V. Bramhachari et al.
Emmerich P, Mika A, von Possel R, Rackow A, Liu Y, Schmitz H, Berisha L (2018) Sensitive and
specific detection of Crimean-Congo Hemorrhagic Fever Virus (CCHFV)—Specific IgM and
IgG antibodies in human sera using recombinant CCHFV nucleoprotein as antigen in μ-capture
and IgG immune complex (IC) ELISA tests. PLoS Negl Trop Dis 12(3):e0006366
Faye M, Dacheux L, Weidmann M, Diop SA, Loucoubar C, Bourhy H, Faye O (2017) Development
and validation of sensitive real-time RT-PCR assay for broad detection of rabies virus. J Virol
Methods 243:120–130
Fernandes JN, Dos Santos LM, Chouin-Carneiro T, Pavan MG, Garcia GA, David MR, Sikulu-
Lord MT (2018) Rapid, noninvasive detection of Zika virus in Aedes aegypti mosquitoes by
near-infrared spectroscopy. Sci Adv 4(5):eaat0496
Fischer K, Diederich S, Smith G, Reiche S, dos Reis VP, Stroh E, Balkema-Buschmann A (2018)
Indirect ELISA based on Hendra and Nipah virus proteins for the detection of henipavirus
specific antibodies in pigs. PLoS One 13(4):e0194385
Furuya AK, Hunt D, George KS, Dupuis AP, Kramer LD, Shi PY, Wong S (2019) Use of the
immunoglobulin G avidity assay to differentiate between recent Zika and past dengue virus
infections. Clin Sci 133(7):859–867
Gootenberg JS, Abudayyeh OO, Kellner MJ, Joung J, Collins JJ, Zhang F (2018) Multiplexed
and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science
360(6387):439–444
Hansen S, Hotop SK, Faye O, Ndiaye O, Böhlken-Fascher S, Pessôa R, Schmidt-Chanasit J (2019)
Diagnosing Zika virus infection against a background of other flaviviruses: studies in high
resolution serological analysis. Sci Rep 9(1):3648
Hayden R, Persing DH (2001) Diagnostic molecular microbiology review. Curr Clin Top Infect
Dis 21:323–348
Kaushik A, Yndart A, Kumar S, Jayant RD, Vashist A, Brown AN, Nair M (2018) A sensitive elec-
trochemical immunosensor for label-free detection of Zika-virus protein. Sci Rep 8(1):9700
Kim HS, Lee K, Kim SJ, Cho S, Shin HJ, Kim C, Kim JS (2018) Arrayed CRISPR screen with
image-based assay reliably uncovers host genes required for coxsackievirus infection. Genome
Res 28(6):859–868
Koishi AC, Suzukawa AA, Zanluca C, Camacho DE, Comach G, dos Santos CND (2018)
Development and evaluation of a novel high-throughput image-based fluorescent neutraliza-
tion test for detection of Zika virus infection. PLoS Negl Trop Dis 12(3):e0006342
Lewandowski K, Xu Y, Pullan ST, Lumley SF, Foster D, Sanderson N, Vipond R (2019)
Metagenomic nanopore sequencing of influenza virus direct from clinical respiratory samples.
bioRxiv, 676155
Makarova KS, Wolf YI, Alkhnbashi OS, Costa F, Shah SA, Saunders SJ, Horvath P et al (2015) An
updated evolutionary classification of CRISPR–Cas systems. Nat Rev Microbiol 13(11):722
Magro L, Jacquelin B, Escadafal C, Garneret P, Kwasiborski A, Manuguerra JC, Tabeling P (2017)
Paper-based RNA detection and multiplexed analysis for Ebola virus diagnostics. Sci Rep
7(1):1347
Miller S, Naccache SN, Samayoa E, Messacar K, Arevalo S, Federman S, Ingebrigtsen D (2019)
Laboratory validation of a clinical metagenomic sequencing assay for pathogen detection in
cerebrospinal fluid. Genome Res 29(5):831–842
Mirian M, Khanahmad H, Darzi L, Salehi M, Sadeghi-Aliabadi H (2017) Oligonucleotide aptam-
ers: potential novel molecules against viral hepatitis. Res Pharm Sci 12(2):88
Mohan H, Gill PS, Kumar A (2019) Hemagglutinin gene based biosensor for early detection of
swine flu (H1N1) infection in human. Int J Biol Macromol 130:720–726
Oh S, Kim J, Tran VT, Lee DK, Ahmed SR, Hong JC, Lee J (2018) Magnetic nanozyme-linked
immunosorbent assay for ultrasensitive influenza A virus detection. ACS Appl Mater Interfaces
10(15):12534–12543
Ojha R, Nandani R, Pandey RK, Mishra A, Prajapati VK (2019) Emerging role of circulating
microRNA in the diagnosis of human infectious diseases. J Cell Physiol 234(2):1030–1043
Omar NAS, Fen YW (2018) Recent development of SPR spectroscopy as potential method for
diagnosis of dengue virus E-protein. Sens Rev 38(1):106–116
17 Advanced Immunotechnological Methods for Detection and Diagnosis of Viral… 275
Pisanic N, Rahman A, Saha SK, Labrique AB, Nelson KE, Granger DA, Heaney CD (2017)
Development of an oral fluid immunoassay to assess past and recent hepatitis E virus (HEV)
infection. J Immunol Methods 448:1–8
Qadiri SSN, Kim SJ, Krishnan R, Kim JO, Kim WS, Oh MJ (2019) Development of an in-situ
hybridization assay using riboprobes for detection of viral haemorrhagic septicemia virus
(VHSV) mRNAs in a cell culture model. J Virol Methods 264:1–10
Qin P, Park M, Alfson K, Tamhankar M, Carrion R, Patterson J, Du K (2019a) Rapid and fully
microfluidic Ebola virus detection with CRISPR-Cas13a. ACS Sens 4:1048–1054
Qin S, Volokhov D, Rodionova E, Wirblich C, Schnell MJ, Chizhikov V, Dabrazhynetskaya A
(2019b) A new recombinant rabies virus expressing a green fluorescent protein: a novel and
fast approach to quantify virus neutralizing antibodies. Biologicals 59:56–61
Roingeard P, Raynal PI, Eymieux S, Blanchard E (2019) Virus detection by transmission electron
microscopy: still useful for diagnosis and a plus for biosafety. Rev Med Virol 29(1):e2019
Saito S, Takayama I, Nakauchi M, Nagata S, Oba K, Odagiri T, Kageyama T (2019) Development
and evaluation of a new real-time RT-PCR assay for detecting the latest H9N2 influenza viruses
capable of causing human infection. Microbiol Immunol 63(1):21–31
Santiago GA, Vázquez J, Courtney S, Matías KY, Andersen LE, Colón C, Muñoz-Jordan JL (2018)
Performance of the Trioplex real-time RT-PCR assay for detection of Zika, dengue, and chi-
kungunya viruses. Nat Commun 9(1):1391
Shirato K, Semba S, El-Kafrawy SA, Hassan AM, Tolah AM, Takayama I, Azhar EI (2018)
Development of fluorescent reverse transcription loop-mediated isothermal amplification
(RT-LAMP) using quenching probes for the detection of the Middle East respiratory syndrome
coronavirus. J Virol Methods 258:41–48
Souf S (2016) Recent advances in diagnostic testing for viral infections. Bioscience Horizons: Int
J Stud Res 9
Tiwari PM, Vanover D, Lindsay KE, Bawage SS, Kirschman JL, Bhosle S, Santangelo PJ (2018)
Engineered mRNA-expressed antibodies prevent respiratory syncytial virus infection. Nat
Commun 9(1):3999
Torrance L, Jones RAC (1981) Recent developments in serological methods suited for use in rou-
tine testing for plant viruses. Plant Pathol 30(1):1–24
Tsang MK, Ye W, Wang G, Li J, Yang M, Hao J (2016) Ultrasensitive detection of Ebola virus
oligonucleotide based on upconversion nanoprobe/nanoporous membrane system. ACS Nano
10(1):598–605
Van Belkum A, Niesters HG (1995) Nucleic acid amplification and related techniques in microbio-
logical diagnosis and epidemiology. Cell Mol Biol 41:615–623
Wang Y, Zhang C, Zhang P, Guo G, Jiang T, Zhao X, Tian Y (2018) Serum exosomal micro RNA
s combined with alpha-fetoprotein as diagnostic markers of hepatocellular carcinoma. Cancer
Med 7(5):1670–1679
Wang R, Ongagna-Yhombi SY, Lu Z, Centeno-Tablante E, Colt S, Cao XE, Erickson D (2019)
A rapid diagnostic platform for colorimetric differential detection of dengue and chikungunya
viral infections. Anal Chem 8:5415–5423
Wesolowska-Andersen A, Everman JL, Davidson R, Rios C, Herrin R, Eng C, Fingerlin TE (2017)
Dual RNA-seq reveals viral infections in asthmatic children without respiratory illness which
are associated with changes in the airway transcriptome. Genome Biol 18(1):12
Whiteside TL (2006, February) Immune suppression in cancer: effects on immune cells, mecha-
nisms and future therapeutic intervention. In Seminars in cancer biology (Vol. 16, No. 1, pp.
3–15). Academic Press, Cambridge
Wong SJ, Furuya A, Zou J, Xie X, Dupuis AP II, Kramer LD, Shi PY (2017) A multiplex micro-
sphere immunoassay for Zika virus diagnosis. EBioMedicine 16:136–140
Xi Y, Xu CZ, Xie ZZ, Zhu DL, Dong JM, Xiao G (2019) Development of a reverse transcription
recombinase polymerase amplification assay for rapid detection of human respiratory syncytial
virus. Mol Cell Probes 45:8–13
Zhang H, Rao H, Wang Y, Wang J, Kong X, Ji Y, Luo B (2019) Evaluation of an antigen assay
for diagnosing acute and chronic hepatitis E genotype 4 infection. J Gastroenterol Hepatol
34(2):458–465
Current Advances in Multi-Epitope Viral
Vaccines Development and Research 18
A. M. V. N. Prathyusha and Pallaval Veera Bramhachari
Abstract
Viral diseases are major public health concern and cause significant morbidity
and mortality globally. The following dreadful viruses viz. Influenza virus, Ebola
virus (EBOV), and Sudan virus are the most recent viruses to cause a global
health concern. Despite recent progress in reduced deaths by viral infection, need
for new molecular, immunoinformatic tools, together with safe plus effective
vaccines is prerequisite due to the pros and cons of prophylactic and therapeutic
vaccines in failure to provide optimal protection. The challenges for viral vaccine
advancements are not restricted for recognition of suitable antigens or adjuvants
and delivery methods, nonetheless cover technical and manufacturing hurdles in
transforming a vaccine to clinic. Research and process improvement is techno-
logical basis which prompts production of new vaccines, essential for commer-
cialization. In this review, we emphasize the present status and recent advances
in designing and developing viral vaccines.
Keywords
Viral vaccines · Multi-epitopes · Inluenza virus · Ebola virus (EBOV) · Hepatitis
E virus
18.1 Introduction
During the past three decades several notable zoonotic virus infections, viz. HIV,
Nipah (NiV) viruses, Hendra (HeV), Ebola, avian influenza, Marburg filoviruses,
Lassa virus (LASV), Crimean–Congo hemorrhagic fever (CCHF) viruses and many
more have emerged suddenly from anonymity and became serious health threats
Subunit vaccines comprise merely antigenic part of virus with potential to promote
immune response while overcoming problems associated with conventional vac-
cines. Exploitation of vaccination strategy in combating virus is a major concern
since synthesis of conventional vaccines often has several obstacles. Hasan et al.
developed novel reverse vaccinology approach that aims to link immunogenetics,
immunogenomics using bioinformatics to identify new vaccine targets. Detection of
T cell and B cell epitopes and HLA (human leukocyte antigen) using in silico
approaches reinforced a possibility in potent vaccine candidate discovery. Vaccine
candidates may be identified as foreign molecules after injection to body. Therefore,
prediction of antigenicity is a crucial part in synthesis of subunit vaccines.
Furthermore, assessment of hydrophilicity is a significant criterion for B cell epit-
ope prediction. Hasan and group reported a design to synthesize nonallergic, immu-
nogenic, and thermostable chimeric ME monovalent subunit vaccine against avian
influenza A (H7N9) virus, and Marburg virus using vaccinomics approach. Viral
proteome was assessed for epitopes with high antigenicity using TMHMM, VaxiJen
v2.0 server (transmembrane topology screening), AllerTOP, AllergenFP, PA3P,
Allermatch servers (allergenicity and toxicity analysis), IEDB’s epitope conser-
vancy analysis tool (population coverage assessment), and MGL Tools (molecular
docking). Two proteins, envelope glycoprotein (GP) and matrix protein (VP40),
generated potent T cell epitopes and are recognized to be most antigenic proteins in
Marburg virus. PEP-FOLD de novo approach was used in prediction of 3D-peptide
structures from amino acid sequences of top ranked epitopes. Finally, three vaccines
were designed most effectively using suitable adjuvants with PADRE sequence,
combined in sequential manner with highly immunogenic (CTL, HTL, and
BCL) respectively. Such constructed vaccines with predicted epitopes were vali-
dated experimentally using animal models for their nonallergic reactions and immu-
nogenic potential (Hasan et al. 2019a, b). Using similar approaches subunit vaccines
may be produced against Chikungunya virus (Narula et al. 2018), Kaposi’s sarcoma-
associated herpesvirus (KSHV) (Chauhan et al. 2019) respectively.
DNA vaccines imply a convenient method to design vaccines with tailored epitopes
being administered into plasmid vectors with desirable memory response.
Furthermore, to evade unfavorable immunodominant epitopes from pathogens, chi-
meric multi-epitope-based DNA vaccine was designed against subgroup J avian
leukosis virus in chickens (Xu et al. 2016). Bounds and his group using in silico
algorithms designed DNA construct entailing HLA class II-restricted T cell epit-
opes obtained from GP and NP (nucleocapsid protein) of EBOV (Ebola virus),
SUDV (Sudan virus), and structural proteins of VEEV (Venezuelan equine enceph-
alitis virus). Identified epitopes with high specificity were examined for binding
ability to soluble HLA molecules. Then vaccinated BALB/c mice with ME-DNA
280 A. M. V. N. Prathyusha and P. V. Bramhachari
vaccine developed and evaluated for immune response using interferon (IFN)-g
ELISpot analyses. These studies provide a concept for designing an ME immuno-
gen along with evaluation focusing on immune response concerning preferred T cell
epitopes (Bounds et al. 2017).
Peptide vaccination can induce both humoral- and cell-mediated immune response
by stimulating T cell immunity in both humans and animals with minimal side
effects. Peptide vaccines are short immunogenic peptide fragments which can elicit
targeted immune response avoiding the chance of allergenic responses. Several pep-
tide vaccines were under progress, viz. vaccine for hepatitis C virus (HCV), human
immunodeficiency virus (HIV), foot and mouth disease, malaria, influenza, swine
fever, human papilloma virus (HPV), and anthrax (Verma et al. 2018). With advance-
ments in computational biology and immunoinformatics approaches, designing
effective strategies for prediction of antigenic epitopes became easier. A peptide-
based vaccine is majorly presented via class II MHC molecules and gets processed
by endocytic pathway. Immune response can also be induced by cytotoxic T cells
(CTL) by cross-presentation where exogenous antigens are processed and presented
onto class I MHC. To improve antigenic presentation, cell penetrating peptides
(CPPs) was one of the promising approach to penetrate peptides efficiently into
cells using cationic peptides (TAT). Commonly used methods for CPP designing
and tagging to antigen were: (1) chemical linkage through covalent bonds, and (2)
recombinant fusion constructs by bacterial expression vectors. Gross et al. tagged a
C-terminal viral protein R (Vpr55-91 and Vpr55-82) fragment of human papilloma-
virus to CPP which paved way for practical ME immunization for neoantigen vac-
cination in cancer patients and is effective now clinically (Gross et al. 2019).
approaches. Such predicted B and T cell epitopes can confer long-lasting immunity
against EBOV with better ability of protection (Yasmin and Nabi 2016).
Strategies for growth of triumphant vaccines are due to design of an antigen delivery
system that optimizes antigen presentation and stimulates broad protective immune
responses. Recent advances in vector delivery technologies, immunology, basic
virology, genetics, and molecular cell biology have led to in-depth understanding of
cellular mechanisms by which vaccines should stimulate the adaptive immune
response, thus presenting novel strategies of vaccination. Classic approach to vac-
cine development is still acquiescent to emerging viruses, the application of molec-
ular techniques in virology has overwhelmingly predisposed our perception of virus
biology, and vaccination schemes based on replication strategies, attenuated and
nonreplicating virus vector approaches that have turned out to be valuable vaccine
platforms. Several virus-like particle (VLP)-based vaccines are currently commer-
cializes in global market, including vaccines against hepatitis B virus and human
papillomavirus (Roldao et al. 2010). Furthermore, through genetic fusion or chemi-
cal conjugation, VLPs are attractive carrier proteins of foreign antigens, since they
can efficiently display them within a host immune system (Tissot et al. 2010;
Plummer and Manchester 2011, Brune et al. 2016). An important advantage of
VLP-based vaccine platforms is that VLPs can present antigens in a dense, repeti-
tive manner, thus effectively enabling the cross-linking of B cell receptors (BCRs)
(Zabel et al. 2014).
Uncertainty of the public health priority and demand for some targets may be
unclear, which increases uncertainty of potential return on investment (ROI).
Furthermore, new vaccine targets are scientifically more complex and challenging.
The challenges presented may require substantial investment in new tools, stan-
dards, analysis methods, other novel approaches to demonstrate safety and effec-
tiveness of vaccine and also limited knowledge in science to progress optimal
vaccines. However, there is a limited understanding of viral pathogenesis and
immune responses against some targeted infectious viral agents, lack of optimal
immune response to potential vaccine candidates, and a limited understanding for
protection mechanism against some vaccine candidates. Conducting clinical trials
to evaluate safety and efficacy of certain preventive vaccines may be particularly
challenging for several reasons, including: relatively low disease incidence (con-
genital cytomegalovirus disease; neonatal group B streptococcal disease); limited
infrastructure in affected geographic areas (EBV vaccine); ethical considerations
(assessing novel approaches to pertussis vaccination of pregnant women to prevent
neonatal pertussis which is recommended); or new settings (hospital acquired
infections).
282 A. M. V. N. Prathyusha and P. V. Bramhachari
Conflict of Interest The authors declare that they have no competing interests.
References
Bounds CE, Terry FE, Moise L, Hannaman D, Martin WD, De Groot AS, Schmaljohn CS (2017)
An immunoinformatics-derived DNA vaccine encoding human class II T cell epitopes of Ebola
virus, Sudan virus, and Venezuelan equine encephalitis virus is immunogenic in HLA trans-
genic mice. Hum Vaccin Immunother 13(12):2824–2836
Brune KD, Leneghan DB, Brian IJ, Ishizuka AS, Bachmann MF, Draper SJ, Biswas S, Howarth
M (2016) Plug-and-display: decoration of virus-like particles via isopeptide bonds for modular
immunization. Sci Rep 6:19234
Chauhan V, Rungta T, Goyal K, Singh MP (2019) Designing a multi-epitope based vaccine to
combat Kaposi Sarcoma utilizing immunoinformatics approach. Sci Rep 9(1):2517
Gross DA, Leborgne C, Chappert P, Masurier C, Leboeuf M, Monteilhet V, Kichler A (2019)
Induction of tumor-specific CTL responses using the C-terminal fragment of Viral protein R as
cell penetrating peptide. Sci Rep 9(1):3937
Hasan M, Azim KF, Begum A, Khan NA, Shammi TS, Imran AS, Urme SRA (2019a) Vaccinomics
strategy for developing a unique multi-epitope monovalent vaccine against Marburg marburg-
virus. Infect Genet Evol 70:140–157
Hasan M, Ghosh PP, Azim KF, Mukta S, Abir RA, Nahar J, Khan MMH (2019b) Reverse vaccinol-
ogy approach to design a novel multi-epitope subunit vaccine against avian influenza A (H7N9)
virus. Microb Pathog 130:19–37
Hassan AO, Amen O, Sayedahmed EE, Vemula SV, Amoah S, York I, Mittal SK (2017) Adenovirus
vector-based multi-epitope vaccine provides partial protection against H5, H7, and H9 avian
influenza viruses. PLoS One 12(10):e0186244
Kanekiyo M, Ellis D, King NP (2019) New vaccine design and delivery technologies. J Infect Dis
219(Supplement_1):S88–S96
Narula A, Pandey RK, Khatoon N, Mishra A, Prajapati VK (2018) Excavating chikungunya
genome to design B and T cell multi-epitope subunit vaccine using comprehensive immunoin-
formatics approach to control chikungunya infection. Infect Genet Evol 61:4–15
18 Current Advances in Multi-Epitope Viral Vaccines Development and Research 283
Plummer EM, Manchester M (2011) Viral nanoparticles and virus-like particles: platforms for
contemporary vaccine design. Wiley Interdiscip Rev Nanomed Nanobiotechnol 3:174–196
Roldao A, Mellado MCM, Castilho LR, Carrondo MJ, Alves PM (2010) Virus-like particles in
vaccine development. Expert Rev Vaccines 9(10):1149–1176
Tissot AC, Renhofa R, Schmitz N, Cielens I, Meijerink E, Ose V, Jennings GT, Saudan P, Pumpens
P, Bachmann MF (2010) Versatile virus-like particle carrier for epitope based vaccines. PLoS
One 5:e9809
Verma S, Sugadev R, Kumar A, Chandna S, Ganju L, Bansal A (2018) Multi-epitope DnaK peptide
vaccine against S. Typhi: an in silico approach. Vaccine 36(28):4014–4022
Xu Q, Cui N, Ma X, Wang F, Li H, Shen Z, Zhao X (2016) Evaluation of a chimeric multi-
epitope-based DNA vaccine against subgroup J avian leukosis virus in chickens. Vaccine
34(33):3751–3756
Yasmin T, Nabi AN (2016) B and T cell epitope-based peptides predicted from evolutionarily
conserved and whole protein sequences of Ebola virus as vaccine targets. Scand J Immunol
83(5):321–337
Zabel F, Mohanan D, Bessa J, Link A, Fettelschoss A, Saudan P, Kundig TM, Bachmann MF
(2014) Viral particles drive rapid differentiation of memory B cells into secondary plasma cells
producing increased levels of antibodies. J Immunol 192:5499–5508