Tissue Engineering For Tissue And Organ
Regeneration D Eberli download
https://ebookbell.com/product/tissue-engineering-for-tissue-and-
organ-regeneration-d-eberli-4115082
Explore and download more ebooks at ebookbell.com
Here are some recommended products that we believe you will be
interested in. You can click the link to download.
Tissue Cell And Organ Engineering Nanotechnologies For The Life
Sciences Volume 9 1st Edition Challa S S R Kumar
https://ebookbell.com/product/tissue-cell-and-organ-engineering-
nanotechnologies-for-the-life-sciences-volume-9-1st-edition-challa-s-
s-r-kumar-2176726
Silk Biomaterials For Tissue Engineering And Regenerative Medicine
Subhas Kundu
https://ebookbell.com/product/silk-biomaterials-for-tissue-
engineering-and-regenerative-medicine-subhas-kundu-4660634
Electrospun Materials For Tissue Engineering And Biomedical
Applications Research Design And Commercialization 1st Edition Tamer
Uyar And Erich Kny Eds
https://ebookbell.com/product/electrospun-materials-for-tissue-
engineering-and-biomedical-applications-research-design-and-
commercialization-1st-edition-tamer-uyar-and-erich-kny-eds-6614086
Electrospun Materials For Tissue Engineering And Biomedical
Applications Research Design And Commercialization Tamer Uyar
https://ebookbell.com/product/electrospun-materials-for-tissue-
engineering-and-biomedical-applications-research-design-and-
commercialization-tamer-uyar-6670478
Extracellular Matrix For Tissue Engineering And Biomaterials 1st Ed
Anna C Berardi
https://ebookbell.com/product/extracellular-matrix-for-tissue-
engineering-and-biomaterials-1st-ed-anna-c-berardi-7149442
Biomaterials For Tissue Engineering And Regeneration Ressler A
https://ebookbell.com/product/biomaterials-for-tissue-engineering-and-
regeneration-ressler-a-48949304
Biomaterials For Tissue Engineering And Regeneration Antonia Ressler
https://ebookbell.com/product/biomaterials-for-tissue-engineering-and-
regeneration-antonia-ressler-55247884
Vascularization For Tissue Engineering And Regenerative Medicine 1st
Edition Wolfgang Holnthoner Andrea Banfi James Kirkpatrick Heinz Redl
https://ebookbell.com/product/vascularization-for-tissue-engineering-
and-regenerative-medicine-1st-edition-wolfgang-holnthoner-andrea-
banfi-james-kirkpatrick-heinz-redl-32560254
Stem Cell Therapy And Tissue Engineering For Cardiovascular Repair
From Basic Research To Clinical Applications 1st Edition Stefan Klotz
https://ebookbell.com/product/stem-cell-therapy-and-tissue-
engineering-for-cardiovascular-repair-from-basic-research-to-clinical-
applications-1st-edition-stefan-klotz-4191650
TISSUE ENGINEERING
FOR TISSUE AND ORGAN
REGENERATION
Edited by Daniel Eberli
Tissue Engineering for Tissue and Organ Regeneration
Edited by Daniel Eberli
Published by InTech
Janeza Trdine 9, 51000 Rijeka, Croatia
Copyright © 2011 InTech
All chapters are Open Access articles distributed under the Creative Commons
Non Commercial Share Alike Attribution 3.0 license, which permits to copy,
distribute, transmit, and adapt the work in any medium, so long as the original
work is properly cited. After this work has been published by InTech, authors
have the right to republish it, in whole or part, in any publication of which they
are the author, and to make other personal use of the work. Any republication,
referencing or personal use of the work must explicitly identify the original source.
Statements and opinions expressed in the chapters are these of the individual contributors
and not necessarily those of the editors or publisher. No responsibility is accepted
for the accuracy of information contained in the published articles. The publisher
assumes no responsibility for any damage or injury to persons or property arising out
of the use of any materials, instructions, methods or ideas contained in the book.
Publishing Process Manager Romina Krebel
Technical Editor Teodora Smiljanic
Cover Designer Jan Hyrat
Image Copyright Arun K. Sharma, 2011.
First published August, 2011
Printed in Croatia
A free online edition of this book is available at www.intechopen.com
Additional hard copies can be obtained from orders@intechweb.org
Tissue Engineering for Tissue and Organ Regeneration, Edited by Daniel Eberli
p. cm.
ISBN 978-953-307-688-1
Contents
Preface IX
Part 1 Cardiac Muscle 1
Chapter 1 Myocardial Tissue Engineering 3
Tatsuya Shimizu
Chapter 2 Cardiac Muscle Engineering:
Strategies to Deliver Stem Cells to the Damaged Site 19
Giancarlo Forte, Stefania Pagliari, Francesca Pagliari,
Paolo Di Nardo and Takao Aoyagi
Chapter 3 Cardiovascular Tissue Engineering Based on
Fibrin-Gel-Scaffolds 35
Stefan Jockenhoevel and Thomas C. Flanagan
Chapter 4 Rapid Prototyping of Engineered Heart Tissues through
Miniaturization and Phenotype-Automation 49
Tetsuro Wakatsuki
Part 2 Skeletal Muscle 59
Chapter 5 Tissue Engineering of Skeletal Muscle 61
Klumpp Dorothee, Horch Raymund E. and Beier Justus P.
Chapter 6 Skeletal Muscle Tissue Engineering
Using Biological Scaffolds for Repair of
Abdominal Wall Defects in a Rabbit Model 81
Zuki Abu Bakar, Ayele Taddese Tsedeke,
Noorjahan Banu Mohamed Alitheen
and Noordin Mohamed Mustapha
Chapter 7 Skeletal and Adipose Tissue Engineering with
Adipose-Derived Stromal Cells 107
Jeong S Hyun, Emily R Nelson, Daniel Montoro,
Benjamin Levi and Michael T. Longaker
VI Contents
Part 3 Ligaments 129
Chapter 8 Tissue Engineering of Ligaments 131
Sarah Rathbone and Sarah Cartmell
Chapter 9 Potential of Tissue-Engineered Ligament
Substitutes for Ruptured ACL Replacement 163
Goulet F., Chabaud S., Simon F., Napa I.D.,
Moulin V. and Hart D.A.
Part 4 Cartilage 179
Chapter 10 Joint Cartilage Tissue Engineering
and Pre-Clinical Safety
and Efficacy Testing 181
Thomas G. Koch, Lorenzo Moroni,
Younes Leysi-Derilou and Lise C. Berg
Chapter 11 Cartilage Regeneration from Bone Marrow Cells
Using RWV Bioreactor and Its Automation System
for Clinical Application 217
Toshimasa Uemura, Masanori Nishi,
Kunitomo Aoki and Takashi Tsumura
Chapter 12 Cartilage Tissue Engineering: the Application of
Nanomaterials and Stem Cell Technology 233
Adelola O. Oseni, Claire Crowley, Maria Z. Boland,
Peter E. Butler and Alexander M. Seifalian
Part 5 Hollow Organs 267
Chapter 13 Bioengineering of Colo-Rectal Tissue 269
Roman Inglin, Lukas Brügger, Daniel Candinas and Daniel Eberli
Chapter 14 Aspects of Urological Tissue Engineering 285
Arun K. Sharma and Dorota I. Rozkiewicz
Part 6 Craniofacial Tissues 315
Chapter 15 Tooth Organ Engineering:
Biological Constraints Specifying
Experimental Approaches 317
Sabine Kuchler-Bopp, Laetitia Keller, Anne Poliard and Herve Lesot
Chapter 16 Transplantation of Corneal Stroma Reconstructed
with Gelatin and Multipotent Precursor Cells
from Corneal Stroma 347
Tatsuya Mimura, Yasuhiko Tabata and Shiro Amano
Contents VII
Chapter 17 Human Ear Cartilage 363
Lu Zhang, Qiong Li, Yu Liu, Guangdong Zhou,
Wei Liu and Yilin Cao
Part 7 Central Nervous System 377
Chapter 18 Advances in the Combined Use of Adult Cell Therapy and
Scaffolds for Brain Tissue Engineering 379
Elisa Garbayo, Gaëtan J.-R. Delcroix,
Paul C. Schiller and Claudia N. Montero-Menei
Part 8 Endocrine Organs 415
Chapter 19 Regenerative Medicine and Tissue Engineering for the
Treatment of Diabetes 417
Matsumoto S, SoRelle JA and Shimoda M
Chapter 20 Perspectives of Islet Cell Transplantation as a Therapeutic
Approach for Diabetes Mellitus 435
Prabha D. Nair and Neena Aloysious
Preface
Over the last decade Tissue Engineering progressed rapidly and first biological
substitutes were developed for several tissues in the body. Today, Tissue Engineering
is one of the major approaches of Regenerative Medicine and represents a growing
and exciting field of research. With the understanding and application of new
knowledge of structure, biology, physiology and cell culture techniques, Tissue
Engineering may offer new treatment alternatives for organ replacement or repair
deteriorated organs. Among the clinical applications of Tissue Engineering are the
production of artificial skin for burn patients, tissue engineered trachea, cartilage for
knee-replacement procedures, urinary bladder replacement, urethra substitutes and
cellular therapies for the treatment of urinary incontinence.
The classical principle of Tissue Engineering is to dissociate cells from a tissue biopsy,
to expand them in culture, and to seed them onto a scaffold material in vitro in order
to generate a viable tissue construct prior to re-implantation into the recipient's
organism. In the appropriate biochemical and biomechanical environment these
tissues will unfold their full functional potential and serve as native tissue equivalents.
Tissue Engineering products may be fully functional at the time of treatment, or have
potential to integrate and evolve into the expected functional tissue after implantation.
While these steps may seem logical and easy to understand, the underlying biology is
far more complicated and more profound questions have to be answered before the
engineering of tissue and organs becomes a routine practice.
Even so, the Tissue Engineering approach has major advantages over traditional organ
transplantation and circumvents the problem of organ shortage. Tissues reconstructed
from readily available biopsy material implicate only minimal or no immunogenicity
when reimplanted in the patient. This eventually conquers several limitations
encountered in tissue transplantation approaches.
This book is aimed at anyone interested in the application of Tissue Engineering in
different organ systems. With a colorful mix of topics which explain the obstacles and
possible solutions, it offers insights into a wide variety of strategies applying the
principles of Tissue Engineering to tissue and organ regeneration. As more and more
applications move toward clinical application, a reliable preclinical model system to
X Preface
evaluate the developed techniques becomes crucial. Several animal models and Tissue
Engineering approaches for a variety of organ systems are presented in this book.
Finally, I would like to thank all the authors who have supported this book with their
contributions.
Daniel Eberli
University Zürich
Switzerland
Part 1
Cardiac Muscle
1
Myocardial Tissue Engineering
Tatsuya Shimizu
Institute of Advanced Biomedical Engineering & Science
Tokyo Women’s Medical University
Japan
1. Introduction
Many lives are lost due to heart diseases including myocardial infarction and
cardiomyopathy. Recent reports have demonstrated that regenerative medicine has
promising potential for recovering severe heart failure. Regenerative therapies for heart
failure include cytokine, gene and cell therapy. Because many types of cardiovascular stem
cells have been identified and their clinical potentials have been demonstrated for the past
decade, cell injection therapy has most attracted both researchers and clinicians (Wollert
2008). On the other hand, significant cell loss due to washing out and cell death has become
problematic in cell injection technique. So, as next generation of regenerative therapy for
impaired heart, transplantation of myocardial patches fabricated by tissue engineering
technology are emerging and are clinically applied. Furthermore, several challenges for
fabricating functional myocardial tissues/organs, which are electrically communicated,
pulsate synchronously and evoke contraction power, have also started (Zimmermann, Didie
et al. 2006). These ambitious challenges may lead to reconstruction of malformed hearts and
become alternative therapy for heart transplantation.
Heart tissues are composed of high-dense cylindrical cardiomyocytes and fibroblasts with
abundant vascular network and collagen-based extracellular matrix (ECM). Cardiomyocytes
pulsate via sodium and calcium ion transient through cell membrane. They are also
electrically coupled by gap junctions composed of connexion 43 and rapid electrical
propagation realizes simultaneous beating as a whole. Continuous blood flow supplies
oxygen and nutrition, and withdraw the waste for high metabolic demand of heart tissues.
These structure and function produce mechanical contractions as a blood pump. Therefore
the researchers should take into account high density culture of cardiomyocyte and
surrounding cells, sufficient micro blood vessel fabrication, cell/ECM orientation and
proper cell-to-cell coupling for engineering heart tissues/organs.
Here, previous and current status of cell injection therapy, myocardial patch transplantation
and pulsatile myocardial tissue fabrication is described with some future views.
2. Cell injection therapy
Cell injection therapy for damaged heart has been researched since the early 1990’s. Many
researchers have demonstrated the therapeutic potential of isolated cell transplantation into
myocardium using various types of cell sources both in animal models and in some clinical
Tissue Engineering for Tissue and Organ Regeneration
4
trials (Puceat 2008). The mechanism of myocardial tissue regeneration has not been
completely cleared, but most researchers have agreed that transplanted cells secrete several
cytokines which promote neovascularization, prohibit fibrosis, decrease cell death and
recruit stem cells, leading to heart function improvement. It has been also asserted that
some of injected cells differentiate into functional cardiomyocytes and may directly
contribute to heart contraction improvement. Although some differences may exist in
according to cell types, multifactorial mechanisms seem to relate with myocardial tissue
regeneration.
In addition to cell sourcing, different routes are used for cell administration. Systematic
intravenous infusion is performed through a central or peripheral vein. This method is
simple and less invasive, however widespread distribution cause low ratio of cell
engraftment. Most popular approach is intracoronary cell infusion via a balloon-catheter.
Injected cells are reached directly in the target myocardial region, however, cells have to
transmigrate across endothelium wall. Intracardiac injection is performed via pericardium
during open heart surgery and via endocardium by a catheter with a 3-D
electromechanical mapping system (NOGA mapping system). These methods realize
relatively targeted delivery, but myocardial damage and arrhythmia induction are
problematic. Future clarification will be needed to decide which is the best approach for
cell injection.
2.1 Skeletal myoblasts
Skeletal myoblasts were the first cell source to enter the clinical application for heart tissue
repair. They lie in a quiescent state on the basal membrane of myofibers and have the
potential to start to proliferate and differentiate into functional skeletal muscle in response
to muscle damage. They can be isolated autologously and be expanded from a single biopsy.
In addition, skeketal myoblasts are relatively resistant to ischemia. Menasche and colleagues
first applied skeletal myoblast injection via epicardium for patients undergoing coronary
artery bypass grafting (CABG) (Menasche, Hagege et al. 2001). The phase I clinical study
(MAGIC I) have shown the feasibility of skeletal myoblast implantation, however, increased
risk of ventricular arrhythmias after the operation. Then, MAGIC II trial was performed to
clarify the safety and efficacy, in which all patients received preventive medication and an
implantable cardioverter-defibrillator for rescuing critical ventricular arrhythmias. In result,
skeletal myoblast injection failed to significantly improve heart function, leading to sample
size reduction (Menasche, Alfieri et al. 2008). On the other hand, the trial indicated the
possibility that high dose cell injection might recover left ventricular dilatation. In addition,
the other clinical trials of catheter-based myoblast implantation via endocardium have
revealed functional efficacy (Opie and Dib 2006). According to these results, not the
regenerative potential of myoblasts themselves but the amount of injected cells and delivery
system may affect the efficacy. Therefore, it seems that skeletal myoblasts should not be
excluded as a cell source for heart tissue repair. More optimization of cell delivery and
comparison of cell sources can address these critical issues.
2.2 Bone marrow-derived cells
Bone marrow-derived cells are the most used cells in clinical trials for myocardial tissue
repair (Wollert 2008). The discovery of circulating progenitor cells originated from human
bone marrow has stimulated research and clinical use of bone marrow-derived cells
Myocardial Tissue Engineering 5
(Asahara, Murohara et al. 1997). Bone marrow cells contain different stem and progenitor
cells which will differentiate into various types of cells including endothelial cells, smooth
muscle cells and cardiomyocytes. Bone marrow mononuclear cells (BMNCs), which can
be isolated simply by gradient sedimentation after bone marrow aspiration without
culture expansion, have been clinically injected via coronary artery from the first. BMNCs
include heterogeneous cell population of monocytes, hematopoietic stem cells and
endothelial progenitor cells (EPCs). Therefore, some groups have used BMNCs selected
by surface markers (CD34+, CD133+) and demonstrated more efficacy of their injection. As
another cell population, mesenchymal stem cells (MSCs) have been researched and
clinically used. Although, MSCs represent between 0.01 and 0.001% of all nucleated cells in
bone marrow, they can be readily expanded in culture. MSCs have the potential to
differentiate into various types of cells and injected MSCs in heart seem to differentiate
into myocardial composing cells. Recent studies have revealed rare happening of
cardiomyocyte differentiation, therefore MSCs seem to recover heart function via their
cytokine secretion and partial differentiation into vascular cells. As a unique feature,
MSCs have the potential to escape from immune detection due to the direct inflammatory
inhibition and the lack of cell-surface molecules. This property has realized allogenic
mesenchymal stem cell transplantation in clinic and has given high impact on cell therapy
research field.
Recent randomized controlled trials of bone marrow-derived cell injection revealed overall
feasibility and safety. However the data has revealed only marginal increases of ejection
fraction (EF) even in positive studies (0-5%) (Martin-Rendon, Brunskill et al. 2008). For
establishing more effective bone marrow-derived cell therapy, optimization of cell source,
cell dose, delivery method and deliver timing will be needed.
2.3 Adipose-derived stem cells
In addition to bone marrow-derived MSCs, stem cells isolated from the stroma of adipose
tissues have represented regenerative potential for heart tissues (Psaltis, Zannettino et al.
2008). Adipose tissue-derived stem cells (ASCs) display features similar to that of bone
marrow-derived MSCs and their angiogenic potential have been reported. Some studies
have also revealed cardiomyocyte differentiation from ASCs. It has not been clarified which
mesenchymal stem cells are superior to other cell types, however, reatively easy isolation of
adipose tissue may push the clinical application of ASCs.
2.4 Cardiac stem cells
Cardiac stem cells (CSCs) are also possible cell source for myocardial tissue regeneration.
Two groups first reported CSC existence in 2003 (Beltrami, Barlucchi et al. 2003; Oh,
Bradfute et al. 2003). Until then, it was common knowledge that heart was a post mitotic
organ, but those reports accelerated the researches for identifying surface marker of CSCs
and culturing them. Islet-1, Sca-1 and c-kit have been known as CSC markers. Recently, it
has been also confirmed that heart has renewal ability at normal state and the annual rate of
turning over is 1% at the age of 25 (Bergmann, Bhardwaj et al. 2009). Although the ability of
CSCs may increase after heart injury, newly formed cardyomyocytes are not sufficient for
replacing damaged muscle tissues. Therefore isolation and expansion of CSCs have been
extensively examined. Some groups have used a different approach to make cardiospheres
from biopsied myocardium, which lead to efficient CSC expansion (Lee, White et al. 2011).
Tissue Engineering for Tissue and Organ Regeneration
6
Clinical trials for injection therapy of autologous CSCs isolated from biopsy sample are now
on going.
2.5 Embryonic stem cells
Although abundant studies demonstrated that MSCs, ASCs and CSCs have the potential of
cardiomyocyte differentiation regarding gene and protein expression, there are no studies
clearly showing beating cardiomyocytes differentiated from those stem cells. On the other
hand, many researchers have confirmed that embryonic stem cells (ESCs) can differentiate
into beating cardiomyocytes in vitro and implantation of ESC-derived cardiomyocytes
improves damaged heart function. Several signal pathways for cardiac differentiation have
been already clarified and various molecules have been reported as its promoters. For
example, noggin increased cardiac differentiation efficacy via regulation of Bone
morphogenetic protein (BMP) signalling pathway (Yuasa, Itabashi et al. 2005) and insulin-
like growth-factor-binding protein 4 (IGFBP4) promotes cardiogenesis by inhibitor of
canonical Wnt signalling (Zhu, Shiojima et al. 2008). In addition, fibloblast growth factor
(FGF), retinoic acid, ascorbic acid and cyclosporine A have been reported to have the
potential to enhance cardiac differentiation from ESCs. The important issue as well as
cardiac differentiation is purification of cardiomyocytes from heterogeneous cell mixture,
because contamination of immature cells leads to teratoma formation. Although gene-
modified ESCs harboring neomycin resistance gene or green fluorescent protein (GFP) gene
in the cardiac-specific gene locus are very useful in non-clinical experiments, safe and
efficient isolation technologies will be needed for clinical application. Culture media control
focusing on the differences of cell metabolism may be useful for safe cell selection. Moreover
immune response of the host is another critical issue. Nucler transfer or cell banking is
possible approach avoiding immunoreaction.
Electrical communication and simultaneous beating of implanted ESC-derived
cardiomyocytes should be also requested for improving damaged heart function without
arrhythmia. In vivo electrophisiological analyses and the transplantation technology for
synchronization will be essential for clinical application of these cells.
2.6 Induced pluripotent stem cells
Induced pluripotent stem cells (iPSCs) also hold great promise for myocardial tissue
engineering (Vunjak-Novakovic, Tandon et al. 2010). Terminally differentiated cells can be
reprogrammed to have the same potential as ESCs by introducing 3 or 4 transcriptional
factor genes. Furthemore none-gene transfer technologies have been developed in the
world. The superiority of iPSCs to ESCs is autologous cells, which do not cause immune
response. Cardiac differentiation of human iPSCs has been reported in the same manner
with ESCs.
Several critical issues must be clarified for clinical use, but ESCs/iPSCs-derived
cardiomyocytes should contribute to myocardial tissue engineering in the view point of
their pulsatile function and scaling-up.
2.7 Problems of cell injection therapy
Cell injection therapies for heart failure are now world-widely performed. While moderate
success of direct cell injection has been observed, the efficacies seem not to reach the level
that general clinicians think cell therapy a reliable treatment for heart failure. More
Myocardial Tissue Engineering 7
optimization of cell source, cell preparation process, injection route, injection timing and
patient population may increase the effectiveness; however one of the essential issues is
cell delivery methodology. Cell injection therapy has significant difficulties about cell
retention in the target tissue. The shape, size, and position of the grafted cells are often
uncontrollable and large amount of the cells are washed-out. Moreover, once retaining
cells die due to necrosis and apoptosis. Time course quantification with TUNEL assay
demonstrated that a large number of the grafted cells die within a few days after injection
in rat models (Zhang, Methot et al. 2001). In the clinical trial using bone marrow-derived
cells, it has been also demonstrated that only 1-3% of the cells infused via coronary
arteries could be detected by 3D positron emission tomography (PET) imaging of the
patient heart. In this study, a large percentage of cells were found in the liver and spleen
immediately after the procedures (Hofmann, Wollert et al. 2005). To clear the problem of
cell loss, hydrogel-cell mixture injection has been pursued. Fibrin, collagen and alginate
hydrogels are now used. Hydrogels with cells are injected as a liquid phase through
syringe or catheter, then, they are polymerized and fixed in the target tissues (Kofidis, de
Bruin et al. 2004). In hydrogel-cell mixture injection therapy, local tissue damage due to
space occupation of hydrogel itself and inflammatory reaction due to hydrogel
biodegradation are problematic.
Therefore, more advanced cell delivery systems have been requested to spread the
regenerative therapy as one of the reliable treatments for heart failure.
3. Tissue engineering
Recent advance of tissue engineering technologies have realized the transplantation of
tissue-engineered construct “myocardial patch” covering over damaged heart surface
instead of simple cell injection into myocardium. Grafted cells within myocardial patches
can survive more and secrete more cytokines, resulting in more heart function
improvement. Furthermore pulsatile myocardial tissues have been successfully engineered
by using cardiomyocytes as a seeding cell source. These tissues may directly help heart
contraction and total heart wall replacement may be possible in future. There are several
contexts of tissue engineering.
3.1 Scaffold-based tissue engineering
Most popular technology of tissue engineering is to seed cells into 3-D pre-fabricated
biodegradable scaffolds which are made from synthetic polymer and biological material.
Hydrogel formation after mixing cells and scaffold solution is another approach.
Decellularized tissues have been also used as scaffolds. These scaffolds play as alternatives for
extra cellular matrix (ECM), therefore, their cell-adhesiveness and porosity affect survived cell
amount and engineered tissue quality. Scaffold modification can control its biodegradation
and tissue formation. Growth factor linkage leads to accelerating tissue formation. Now these
scaffold-based tissue engineering has been widely applied to cardiovascular tissue
regeneration as well as other tissue repair (Vunjak-Novakovic, Tandon et al. 2010).
3.2 Cell sheet-based tissue engineering
In contrast to scaffold-based tissue engineering, our group have developed unique
technique involving cell sheet stacking to fabricate 3-D tissues (Shimizu, Yamato et al. 2003).
Tissue Engineering for Tissue and Organ Regeneration
8
Cell sheets are 2-D connecting pure cells without any scaffolds, therefore cell-dense 3-D
tissues can be fabricated by stacking cell sheets. Cell sheets are harvested from intelligent
culture surface “temperature-responsive culture surface”, which are covalently grafted with
temperature-responsive polymer, poly (N-isopropylacrylamide) (PIPAAm) (Okano, Yamada
et al. 1993). The surfaces are slightly hydrophobic and cell-adhesive at 37˚C, on the other
hand, the surface changes to hydrophilic and not cell adhesive below 32˚C. Confluently
cultured cells on the surface can detach as a contiguous cell sheet simply by reducing
temperature. Furthermore, biological molecules underneath cell sheets are also preserved
and play a critical role as an adhesive agent during cell sheet stacking. Cell sheet-based
tissue engineering has been applied for a wide range of regenerative medicine including
corneal epithelial replacement, heart tissue repair, pneumothorax repair, liver tissue repair
and so on.
According to the spread of the concept fabricating 3-D tissues from 2-D confluent cells,
several other technologies using this concept have emerged. Cell sheet fabrication
techniques using fibrin coated dishes or nanofibrous polycaprolactone meshes have been
reported (Shin, Ishii et al. 2004; Itabashi, Miyoshi et al. 2005). Cell sheet-like constructs have
been also engineered using magnetite nanoparticles (Ito, Hibino et al. 2005). Magnetically
labelled cells are attached on culture materials by magnetic force and confluent cells are
harvested as a cell sheet by magnetic force release. Thus, cell sheet-based tissue engineering
has now spread in the world as scaffold-free tissue engineering.
4. Myocardial patch transplantation
Both scaffold-based and cell sheet-based tissue engineering have been used for myocardial
patch fabrication. Not only cardiomyocytes but also other types of cells have been used for
creating myocardial patches and some myocardial patches using non-cardiomyocytes have
been already clinically transplanted over damaged hearts. (Fig. 1.)
4.1 Scaffold-based mayocardial patch
In myocardial patch fabrication, synthetic polymer, biological material and decellularized
tissue have been used as prefabricated scaffolds. Li and colleagues, who were one of the
pioneer groups of myocardial tissue engineering, first demonstrated that gelatine sponges
seeded with cardiac cells have therapeutic potentials for cryoinjured rat hearts (Li, Jia et al.
1999). Leor and colleagues reported that bioengineered heart grafts using porous alginate
scaffolds attenuated left ventricular dilatation and heart function deterioration in infarction
model (Leor, Aboulafia-Etzion et al. 2000). Eschenhagen and Zimmermann’s group have
developed innovative myocardial tissue engineering approach (Zimmermann,
Schneiderbanger et al. 2002). They have fabricated 3-D tissues by gelling mixture of cardiac
cells and collagen solution. The constructs induced systolic wall thickening of the left
ventricle infracted area and improved fractional shortening of damaged hearts in rat
myocardial infarction model (Zimmermann, Melnychenko et al. 2006). Small intestinal
submucosa (SIS) has also been used as a scaffold for myocardial patch. MSC-seeded SIS
improved heart contraction in rabbit infarction model (Tan, Zhi et al. 2009). There have been
various types of myocardial patches using different scaffolds and different cell sources.
Although implantable human myocardial patches using beating cardiomyocytes have not
been established now, clinical trials of collagen-based myocardial patch with bone marrow
Myocardial Tissue Engineering 9
cells (MAGNUM trial) (Chachques, Trainini et al. 2007) and vicryl mesh-based myocardial
patches with fibroblasts (Anginera) ((Mirsadraee, Wilcox et al. 2006)) have revealed
feasibility and safety of myocardial patch transplantation.
Diseased heart
Cells
Stacking
3. Cell sheet-based TE
Temperature-responsive
culture surface
Cells & ECM
mixture
Gelling
Scaffold
1. Scaffold-based TE 2. Hydrogel-based TE
Cell sheet
Seeding
Fig. 1. Tissue engineering (TE) strategies for myocardial patch fabrication
4.2 Cell sheet-based myocardial patch
Many types of cell sheets have been reported to improve impaired heart function (Shimizu,
Sekine et al. 2009). Cell sheets are transplanted onto heart surface directly via open heart
surgery and cells can be more effectively delivered as thin, but large-area cell-dense grafts
than isolated cell injection. Scaffold-based myocardial patches are usually transplanted on
myocardium with suture, on the other hand, cell sheets are transplanted with no suture
because biological adhesive proteins underneath cell sheets promote the attachment. When
neonatal rat cardiac cell sheets were transplanted onto infracted rat hearts, grafted
cardiomyocytes communicated with host myocardium via gap junctions and blood vessels
formed within the graft, resulting in significant improvement of heart function (Miyagawa,
Sawa et al. 2005; Sekine, Shimizu et al. 2006).
Sawa and colleagues have started to use skeletal myoblasts for cell sheet fabrication, because
myoblasts can be isolated autologously and are relatively resistant to ischemic condition.
Tissue Engineering for Tissue and Organ Regeneration
10
The recovery of heart function by skeletal myoblast transplantation has been confirmed in
rat ischemic model, in dilated cardiomyopathy hamster model, in pacing-induced canine
heart failure model and in pig infarction model (Memon, Sawa et al. 2005; Hata, Matsumiya
et al. 2006; Kondoh, Sawa et al. 2006; Miyagawa, Saito et al. 2010). Regarding stacking cell
sheet number, 3-5 layers are optimal and more layering cause primary necrosis of the
constructs (Sekiya, Matsumiya et al. 2009). They have demonstrated more hematopoietic
stem cells and less fibrosis in cell sheet transplantation than in isolated cell injection in
accordance with more expression of stromal-derived factor 1 (SDF-1), hepatocyte growth
factor (HGF), and vascular endothelial growth factor (VEGF). Based on these results, clinical
trial of autologous myoblast sheet transplantation for severe heart failure has started and the
detailed results will appear soon.
In the same manner with cell injection therapy, MSCs are used as a candidate cell source for
human implantable cell sheet. Adipose tissue-derived MSCs and menstrual blood-derived
MSCs have improved damaged heart function in rat infarction model (Miyahara, Nagaya et
al. 2006; Hida, Nishiyama et al. 2008). MSCs can gradually grow to form a thick stratum
containing newly formed blood vessels and some cells seem to differentiate into
cardiomyocytes at least by histological analyses. Further studies will be needed to confirm
the differentiation into functional beating cardiomyocytes and possibilities to differentiate
into unexpected cell types.
As emerging cell source, cell sheets of stem cell antigen 1-positive (Sca-1-positive) CSCs
ameliorates cardiac dysfunction in mouse infarction model through cardiomyocyte
differentiation and paracrine mechanisms mediated via soluble vascular cell adhesion
molecule 1 (VCAM-1)/very late antigen-4 (VLA-4) signaling pathway (Matsuura, Honda et
al. 2009). In addition, cardiac cell sheets originated from ESCs/iPSCs have been successfully
fabricated and their transplantation into animal models is now ongoing.
For enhancing the efficacy of cell sheet transplantation, gene-modified cell sheets have been
examined. Bcl-2 expressed myoblast sheets prolonged survival, increased production of
proangiogenic paracrine mediators, and enhanced the therapeutic efficacy (Kitabayashi,
Siltanen et al. 2010). HGF overexpression in myoblast sheets enhances their angiogenic
potential in rat chronic heart failure model (Siltanen, Kitabayashi et al. 2011). As another
concept, cell sheets co-cultured with endothelial cell sources have been transplanted in rat
infarction models. Transplantation of EPC co-cultured fibroblast sheet improved heart
function more than only fibroblast sheet implantation or EPC injection (Kobayashi, Shimizu
et al. 2008). Furthermore, endothelial cell co-culture within cardiomyocyte sheets induced
more neovascularization and more improvement of cardiac function than only
cardiomyocyte sheets (Sekine, Shimizu et al. 2008). These studies indicate advanced
strategies of cell sheet transplantation.
As mentioned previously, it is considered that the main mechanism of heart function
improvement is neovascularization, fibrosis inhibition, apoptosis inhibition and stem cell
recruitment due to various cytokines secreted from grafted cells. In comparison with cell
injection approach, increase of cell survival within myocardial patches leads to more
cytokine secretion, then, resulting in more function improvement. In addition to cytokine
secretion, myocardial patches may have girdling effect and prohibit heart dilatation.
Therefore, myocardial patch transplantation is quite different cell delivery method from cell
injection and has more potential to rescue diseased hearts. In the case of myocardial patches
using beating cardiomyocytes, direct enhancement of contraction power is additionally
Myocardial Tissue Engineering 11
expected, however, electrical synchronization between host hearts and transplanted patches
is a critical issue to be clarified.
5. Engineering plusatile myocardial tisssue
Beyond myocardial patch fabrication, several research groups have challenged to fabricate
pulsatile myocardial tissues by their original tissue engineering strategies. Bioengineered
contractile myocardial tissues may realize new therapeutics for severe heart diseases and be
useful as alternatives for animal models.
5.1 Pre-fabricated scaffold-based myocardial tissue fabrication
The first approach for engineering functional myocardial tissue is seeding cardiomyocytes
into synthetic or biological 3-D scaffolds. Vunjak-Novakovic and colleagues first reported
that seeding primary cultured cardiomyocytes onto disc-shaped polyglycolic acid (PGA)
scaffolds in rotating bioreactor system resulted in spontaneously pulsatile myocardial
tissues (Papadaki, Bursac et al. 2001). Optimization of cell population, serum concentration
and scaffold coating improved electrical conduction velocity of engineered constructs.
Radisc and colleagues seeded rat cardiomyocytes in Matrigel onto collagen sponges and
stimulated the constructs electrically. The stimulation improved the conductive and
contractile properties in accordance with increased expression of myosin heavy chain and
connexion 43. Furthermore, cardiomyocytes in the electrically stimulated constructs were
more aligned and elongated as same as those in native heart tissue (Radisic, Park et al.
2007).
Following these studies, many research groups have started to engineer myocardial tissue in
vitro by using various types of scaffolds. Scaffold porosity is one of the critical factors for
pre-fabricated scaffold-based tissue engineering. High porosity increases seeded cell
number and facilitates mass transport. Surface modyfication is also important for cell
attachment and survival. Laminin coating improved cardiomyocyte adhesiveness. In
addition, scaffold elasticity and degradability affect contraction property of engineered
myocardium. Further studies are ongoing to development appropriate scaffold materials for
myocardial tissue engineering.
5.2 Hydrogel-based myocardial tissue fabrication
The second approach is to form 3-D tissues by gelling of cardiac cell and matrix solution
mixture. Eschenhagen and Zimmermann have continuously developed this strategy using
collagen gel and successfully engineered macroscopically beating cardiac tissues
(Zimmermann and Cesnjevar 2009). First, neonatal rat cardiomyocytes were suspended in
collagen I solution and the mixture was poured into the mold. After gelling, the constructs
were unidirectionally stretched with the mechanical devise. They have also realized
contraction force measurement. Cyclic stretch introduced cell alignment along the stretching
direction and increased mitochondrial density, leading to native heart-like tissue. The
contraction force of engineered myocardium was comparative with native heart tissue and
responded to pharmacological agents properly. Ring-shaped myocardial tissues were also
fabricated and combined 5 constructs were transplanted onto infarcted rat hearts.
Interestingly, the constructs synchronized to each other and improved damaged heart
function. They have also confirmed that co-culture constructs including cardiomyocytes,
Tissue Engineering for Tissue and Organ Regeneration
12
fibroblasts and endothelial cells were superior to cardiomyocyte rich constructs in
morphology and function. Recently, they have also started to utilize human cardiomyocytes
differentiated from ESC/iPSC as cell source and challenged to create human myocardial
tissues (Zimmermann 2011). In contrast to pre-fabricated scaffold usage, relatively
homogeneous myocardial tissues are engineered by hydrogel-based approach. Therefore
collagen gel-based myocardial tissue engineering has now become popular in the world.
5.3 Cell sheet-based myocardial tissue fabrication
The third approach is to engineer 3-D pulsatile myocardial tissues by stacking cardiac cell
sheets. As mentioned previously, 2-D cell sheets can be harvested from temperature-
responsive culture dishes only by lowering temperature and do not include any materials. 3-
D tissues are constructed by layering cell sheets. Because 2-D confluent cells are directly
stacked without any scaffolds, resulting constructs are cell-dense 3-D tissues. It is well-
known that 2-D confluent cardiomyocytes connect to each other electrically via gap
junctions resulting in synchronized beating. Cardiac cell sheets harvested from temperature-
responsive culture dishes maintain this synchronized pulsation (Shimizu, Yamato et al.
2002). For creating 3-D functional heart tissues by layering cardiac cell sheets, morphological
and electrical communications between cell sheets are critical. Multiple-electrode
extracellular recording system revealed that double-layer rat cardiac cell sheets coupled
electrically about one hour after layering and histological analysis showed the existence of
connexin 43 between two cardiac cell sheets. Adhesive proteins deposited on cell sheet
surface are considered to promote these rapid electrical communications (Haraguchi,
Shimizu et al. 2006). Stacked cardiac cell sheets beat synchronously in macroscopic view and
the constructs transplanted into rat subcutaneous tissues also pulsated continuously at least
up to one year and eight months after implantation. Morphological analyses showed
elongated cardiomyocytes, well-differentiated sarcomeres, gap junctions and multiple blood
vessels, which were characteristic structure of native heart tissue (Shimizu, Yamato et al.
2002). Long-term observation revealed that their size, conduction velocity, and contractile
force increased in proportion to the host growth (Shimizu, Sekine et al. 2006). Recently,
fabrication of cardiac cell sheets using ESC-originated cardiomyocytes have just started and
human cardiac cell sheets will appear in near future.
5.4 Fabrication of vascularized myocardial tissue
One of the major obstacles in myocardial tissue engineering is scaling-up of the constructs.
Insufficient supply of oxygen and nutrient, and waste accumulation limit their thickness.
Actually, cells are sparse in the central area, on the other hand, cells are dense in the outer
surface (100-200μm) area in scaffold-based myocardial tissue engineering. In the case of cell
sheet-based myocardial tissue engineering, thickness limit is approximately 80μm (3 layers)
(Shimizu, Sekine et al. 2006). Several approaches have been examined in the point of view
overcoming diffusion limit. Perfusion of culture media through the constructs using porous
scaffolds is one possible approach. Media penetration increased cell migration depth and
improved cell metabolism. However shear stress due to media flow may prohibit tight cell
attachment on the scaffold material. Media perfusion with oxygen carrier, perfluorocarbon
(PFC) has been also examined for improving oxygen transport. PFC usage increased cell
proliferation and improved pulsatile function. Media penetration is useful to some extent,
however, it becomes more difficult as cell density increases.
Myocardial Tissue Engineering 13
To overcome this problem, it has been requested to develop new technologies for
introducing vasculature or vascular-like structure into engineered tissues. Several
researchers have tried to generate microchannel network within porous 3-D scaffolds by
microfabrication techniques including CO2 laser ablation. The technology has not reached to
mimicking native micro capillary network. On the other hand, recent studies have revealed
that co-cultured endothelial cells within cardiac constructs can spontaneously form
vascular-like network in vitro and tubular formation has been found in some parts. It has
been also confirmed that this pre-vascular structure connected to host blood vessels
immediately after transplantation and the newly developed vessels within the constructs
were blood-supplied within a few days (Sekiya, Shimizu et al. 2006). We have already
demonstrated that the tissue thickness of cardiac cell sheets co-cultured with endothelial
cells were just twice as the thickness of cardiac cell sheets without endothelial cells (Sekine,
Shimizu et al. 2008). Although endothelial cell co-culture is helpful for accelerating blood
vessel formation, more scaling-up is still limited due to primary ischemia until sufficient
vascularization.
One possible idea for scaling-up is utilizing in vivo vascularization power. Our group has
reported that triple-layer cardiac cell sheets were repeatedly implanted after waiting enough
vascular formation within previously implanted tissues. In result, synchronously beating
thick myocardial tissues with sufficient micro capillaries were successfully fabricated and
10-times transplantation of triple-layer constructs (totally 30 sheets) formed 1-mm thick,
pulsatile myocardial tissues. Furthermore, when triple-layer grafts were transplanted
repeatedly over a surgically connectable artery and vein in leg, the multilayer constructs
were blood-supplied from the thick artery and vein. The constructs were successfully
resected with the connectable blood vessels and were ectopically transplanted in neck with
direct vessel anastomoses (Shimizu, Sekine et al. 2006). Recently several groups have also
utilized in vivo power for myocardial tissue engineering. Cardiomyocytes, ECM alternatives
and native blood vessels were packed in the special chamber and incubated in vivo.
Vascularized heart-like tissues were created in the body (Morritt, Bortolotto et al. 2007; Birla,
Dhawan et al. 2009).
Furthemore, next challenge is now in vitro fabrication of vascularized myocardial tissues.
Kofidis and colleagues have constructed fibrin gel-based myocardial tissues containing rat
aortas (1-2mm), through which culture media was perfused (Kofidis, Lenz et al. 2003). Cell
survival and metabolism were improved, however formation of functional blood vessels
connecting with central aortas were not clear. We are now trying to promote endothelial cell
tubular formation within in vitro engineered cardiac tissues and to perfuse culture media
through the newly formed vessels using perfusion bioreactors. Further studies will be
needed to break through the obstacles for in vitro scaling up.
5.5 From tissue engineering to organ engineering
For future organ engineering, some groups have challenged to engineer myocardial
constructs with pumping function. Ott and colleagues have used decellularized organ as a
scaffold. They decellularized rat whole hearts and re-seeded cardiac cells into decellularized
hearts. Heart contraction was recovered and pump function was generated (Ott, Matthiesen
et al. 2008). Zimmermann’s group developed pouch-like myocardial tissue by their
technology as previously described and covered heart with pouch-like constructs (Yildirim,
Naito et al. 2007).
Tissue Engineering for Tissue and Organ Regeneration
14
Regarding cell sheet technology, myocardial tubes have been fabricated by wrapping rat
cardiac cell sheets around fibrin tubes and rat resected aortas. The engineered myocardial
tubes revealed spontaneous, synchronized pulsation and small but significant inner
pressure changes (about 0.1 mmHg) in vitro (Kubo, Shimizu et al. 2007). On the other hand,
resected rat aortas wrapped with cardiac cell sheets were micro surgically transplanted in
place of the abdominal aorta. After 1 month, in vivo myocardial tubes demonstrated
spontaneous beating and evoked independent blood pressures (about 6 mm Hg). The value
of in vivo myocardial tubes was much bigger than in vitro myocardial tubes (Sekine,
Shimizu et al. 2006). Comparing in vitro and in vivo, it was considered that pulsation due to
host blood flow has induced cardiomyocyte hypertrophy, leading to improvement of
pumping function. Therefore pulsatile perfusion bioreactors may improve pumping
function of in vitro engineered myocardial tubes.
Thus, small size myocardial constructs evoking pumping function have been realized.
Expansion and selection of cardiomyocytes, and sufficient blood vessel formation for
scaling-up are now critical issues for organ engineering.
6. Conclusions
As the first generation of cardiac regenerative therapy, many clinical trials of cell injection
therapy have been already performed. The controversial arguments about its effectiveness
will be settled in next several years. Tissue engineered myocardial patches have now
emerged as the second generation and previous studies indicate promising potential for
rescuing damaged heart. As the third generation, tissue-engineered pulsatile myocardial
tissues should support heart contraction physically. Furthermore, future development of cell
sourcing and scaling-up technologies may realize “bioengineered hearts”.
7. Acknowledgment
This work is granted by the Japan Society for the Promotion of Science (JSPS) through the
“Funding Program for World-Leading Innovative R&D on Science and Technology (FIRST
Program),” initiated by the Council for Science and Technology Policy (CSTP).
8. References
Asahara, T., T. Murohara, et al. (1997). Isolation of putative progenitor endothelial cells for
angiogenesis. Science 275(5302): 964-967.
Beltrami, A. P., L. Barlucchi, et al. (2003). Adult cardiac stem cells are multipotent and
support myocardial regeneration. Cell 114(6): 763-776.
Bergmann, O., R. D. Bhardwaj, et al. (2009). Evidence for cardiomyocyte renewal in humans.
Science 324(5923): 98-102.
Birla, R. K., V. Dhawan, et al. (2009). Cardiac cells implanted into a cylindrical, vascularized
chamber in vivo: pressure generation and morphology. Biotechnol Lett 31(2): 191-
201.
Chachques, J. C., J. C. Trainini, et al. (2007). Myocardial assistance by grafting a new
bioartificial upgraded myocardium (MAGNUM clinical trial): one year follow-up.
Cell Transplant 16(9): 927-934.
Myocardial Tissue Engineering 15
Haraguchi, Y., T. Shimizu, et al. (2006). Electrical coupling of cardiomyocyte sheets occurs
rapidly via functional gap junction formation. Biomaterials 27(27): 4765-4774.
Hata, H., G. Matsumiya, et al. (2006). Grafted skeletal myoblast sheets attenuate myocardial
remodeling in pacing-induced canine heart failure model. J Thorac Cardiovasc Surg
132(4): 918-924.
Hida, N., N. Nishiyama, et al. (2008). Novel cardiac precursor-like cells from human
menstrual blood-derived mesenchymal cells. Stem Cells 26(7): 1695-1704.
Hofmann, M., K. C. Wollert, et al. (2005). Monitoring of bone marrow cell homing into the
infarcted human myocardium. Circulation 111(17): 2198-2202.
Itabashi, Y., S. Miyoshi, et al. (2005). A new method for manufacturing cardiac cell sheets
using fibrin-coated dishes and its electrophysiological studies by optical mapping.
Artif Organs 29(2): 95-103.
Ito, A., E. Hibino, et al. (2005). Construction and delivery of tissue-engineered human retinal
pigment epithelial cell sheets, using magnetite nanoparticles and magnetic force.
Tissue Eng 11(3-4): 489-496.
Kitabayashi, K., A. Siltanen, et al. (2010). Bcl-2 expression enhances myoblast sheet
transplantation therapy for acute myocardial infarction. Cell Transplant 19(5): 573-
588.
Kobayashi, H., T. Shimizu, et al. (2008). Fibroblast sheets co-cultured with endothelial
progenitor cells improve cardiac function of infarcted hearts. J Artif Organs 11(3):
141-147.
Kofidis, T., J. L. de Bruin, et al. (2004). Injectable bioartificial myocardial tissue for large-scale
intramural cell transfer and functional recovery of injured heart muscle. J Thorac
Cardiovasc Surg 128(4): 571-578.
Kofidis, T., A. Lenz, et al. (2003). Pulsatile perfusion and cardiomyocyte viability in a solid
three-dimensional matrix. Biomaterials 24(27): 5009-5014.
Kondoh, H., Y. Sawa, et al. (2006). Longer preservation of cardiac performance by sheet-
shaped myoblast implantation in dilated cardiomyopathic hamsters. Cardiovasc Res
69(2): 466-475.
Kubo, H., T. Shimizu, et al. (2007). Creation of myocardial tubes using cardiomyocyte sheets
and an in vitro cell sheet-wrapping device. Biomaterials 28(24): 3508-3516.
Lee, S. T., A. J. White, et al. (2011). Intramyocardial injection of autologous cardiospheres or
cardiosphere-derived cells preserves function and minimizes adverse ventricular
remodeling in pigs with heart failure post-myocardial infarction. J Am Coll Cardiol
57(4): 455-465.
Leor, J., S. Aboulafia-Etzion, et al. (2000). Bioengineered cardiac grafts: A new approach to
repair the infarcted myocardium? Circulation 102(19 Suppl 3): III56-61.
Li, R. K., Z. Q. Jia, et al. (1999). Survival and function of bioengineered cardiac grafts.
Circulation 100(19 Suppl): II63-69.
Martin-Rendon, E., S. J. Brunskill, et al. (2008). Autologous bone marrow stem cells to treat
acute myocardial infarction: a systematic review. Eur Heart J 29(15): 1807-1818.
Matsuura, K., A. Honda, et al. (2009). Transplantation of cardiac progenitor cells ameliorates
cardiac dysfunction after myocardial infarction in mice. J Clin Invest 119(8): 2204-
2217.
Tissue Engineering for Tissue and Organ Regeneration
16
Memon, I. A., Y. Sawa, et al. (2005). Repair of impaired myocardium by means of
implantation of engineered autologous myoblast sheets. J Thorac Cardiovasc Surg
130(5): 1333-1341.
Menasche, P., O. Alfieri, et al. (2008). The Myoblast Autologous Grafting in Ischemic
Cardiomyopathy (MAGIC) trial: first randomized placebo-controlled study of
myoblast transplantation. Circulation 117(9): 1189-1200.
Menasche, P., A. A. Hagege, et al. (2001). Myoblast transplantation for heart failure. Lancet
357(9252): 279-280.
Mirsadraee, S., H. E. Wilcox, et al. (2006). Development and characterization of an acellular
human pericardial matrix for tissue engineering. Tissue Eng 12(4): 763-773.
Miyagawa, S., A. Saito, et al. (2010). Impaired myocardium regeneration with skeletal cell
sheets--a preclinical trial for tissue-engineered regeneration therapy. Transplantation
90(4): 364-372.
Miyagawa, S., Y. Sawa, et al. (2005). Tissue cardiomyoplasty using bioengineered contractile
cardiomyocyte sheets to repair damaged myocardium: their integration with
recipient myocardium. Transplantation 80(11): 1586-1595.
Miyahara, Y., N. Nagaya, et al. (2006). Monolayered mesenchymal stem cells repair scarred
myocardium after myocardial infarction. Nat Med 12(4): 459-465.
Morritt, A. N., S. K. Bortolotto, et al. (2007). Cardiac tissue engineering in an in vivo
vascularized chamber. Circulation 115(3): 353-360.
Oh, H., S. B. Bradfute, et al. (2003). Cardiac progenitor cells from adult myocardium:
homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A
100(21): 12313-12318.
Okano, T., N. Yamada, et al. (1993). A novel recovery system for cultured cells using
plasma-treated polystyrene dishes grafted with poly(N-isopropylacrylamide). J
Biomed Mater Res 27(10): 1243-1251.
Opie, S. R. and N. Dib (2006). Surgical and catheter delivery of autologous myoblasts in
patients with congestive heart failure. Nat Clin Pract Cardiovasc Med 3 Suppl 1: S42-
45.
Ott, H. C., T. S. Matthiesen, et al. (2008). Perfusion-decellularized matrix: using nature's
platform to engineer a bioartificial heart. Nat Med 14(2): 213-221.
Papadaki, M., N. Bursac, et al. (2001). Tissue engineering of functional cardiac muscle:
molecular, structural, and electrophysiological studies. Am J Physiol Heart Circ
Physiol 280(1): H168-178.
Psaltis, P. J., A. C. Zannettino, et al. (2008). Concise review: mesenchymal stromal cells:
potential for cardiovascular repair. Stem Cells 26(9): 2201-2210.
Puceat, M. (2008). Pharmacological approaches to regenerative strategies for the treatment of
cardiovascular diseases. Curr Opin Pharmacol 8(2): 189-192.
Radisic, M., H. Park, et al. (2007). Biomimetic approach to cardiac tissue engineering. Philos
Trans R Soc Lond B Biol Sci 362(1484): 1357-1368.
Sekine, H., T. Shimizu, et al. (2008). Endothelial cell coculture within tissue-engineered
cardiomyocyte sheets enhances neovascularization and improves cardiac function
of ischemic hearts. Circulation 118(14 Suppl): S145-152.
Sekine, H., T. Shimizu, et al. (2006). Cardiomyocyte bridging between hearts and
bioengineered myocardial tissues with mesenchymal transition of mesothelial cells.
J Heart Lung Transplant 25(3): 324-332.
Myocardial Tissue Engineering 17
Sekine, H., T. Shimizu, et al. (2006). Pulsatile myocardial tubes fabricated with cell sheet
engineering. Circulation 114(1 Suppl): I87-93.
Sekiya, N., G. Matsumiya, et al. (2009). Layered implantation of myoblast sheets attenuates
adverse cardiac remodeling of the infarcted heart. J Thorac Cardiovasc Surg 138(4):
985-993.
Sekiya, S., T. Shimizu, et al. (2006). Bioengineered cardiac cell sheet grafts have intrinsic
angiogenic potential. Biochem Biophys Res Commun 341(2): 573-582.
Shimizu, T., H. Sekine, et al. (2006). Long-term survival and growth of pulsatile myocardial
tissue grafts engineered by the layering of cardiomyocyte sheets. Tissue Eng 12(3):
499-507.
Shimizu, T., H. Sekine, et al. (2009). Cell sheet-based myocardial tissue engineering: new
hope for damaged heart rescue. Curr Pharm Des 15(24): 2807-2814.
Shimizu, T., H. Sekine, et al. (2006). Polysurgery of cell sheet grafts overcomes diffusion
limits to produce thick, vascularized myocardial tissues. FASEB J 20(6): 708-710.
Shimizu, T., M. Yamato, et al. (2002). Electrically communicating three-dimensional cardiac
tissue mimic fabricated by layered cultured cardiomyocyte sheets. J Biomed Mater
Res 60(1): 110-117.
Shimizu, T., M. Yamato, et al. (2002). Fabrication of pulsatile cardiac tissue grafts using a
novel 3-dimensional cell sheet manipulation technique and temperature-responsive
cell culture surfaces. Circ Res 90(3): e40.
Shimizu, T., M. Yamato, et al. (2003). Cell sheet engineering for myocardial tissue
reconstruction. Biomaterials 24(13): 2309-2316.
Shin, M., O. Ishii, et al. (2004). Contractile cardiac grafts using a novel nanofibrous mesh.
Biomaterials 25(17): 3717-3723.
Siltanen, A., K. Kitabayashi, et al. (2011). hHGF Overexpression in Myoblast Sheets
Enhances Their Angiogenic Potential in Rat Chronic Heart Failure. PLoS One 6(4):
e19161.
Tan, M. Y., W. Zhi, et al. (2009). Repair of infarcted myocardium using mesenchymal stem
cell seeded small intestinal submucosa in rabbits. Biomaterials 30(19): 3234-3240.
Vunjak-Novakovic, G., N. Tandon, et al. (2010). Challenges in cardiac tissue engineering.
Tissue Eng Part B Rev 16(2): 169-187.
Wollert, K. C. (2008). Cell therapy for acute myocardial infarction. Curr Opin Pharmacol 8(2):
202-210.
Yildirim, Y., H. Naito, et al. (2007). Development of a biological ventricular assist device:
preliminary data from a small animal model. Circulation 116(11 Suppl): I16-23.
Yuasa, S., Y. Itabashi, et al. (2005). Transient inhibition of BMP signaling by Noggin induces
cardiomyocyte differentiation of mouse embryonic stem cells. Nat Biotechnol 23(5):
607-611.
Zhang, M., D. Methot, et al. (2001). Cardiomyocyte grafting for cardiac repair: graft cell
death and anti-death strategies. J Mol Cell Cardiol 33(5): 907-921.
Zhu, W., I. Shiojima, et al. (2008). IGFBP-4 is an inhibitor of canonical Wnt signalling
required for cardiogenesis. Nature 454(7202): 345-349.
Zimmermann, W. H. (2011). Embryonic and embryonic-like stem cells in heart muscle
engineering. J Mol Cell Cardiol 50(2): 320-326.
Zimmermann, W. H. and R. Cesnjevar (2009). Cardiac tissue engineering: implications for
pediatric heart surgery. Pediatr Cardiol 30(5): 716-723.
Tissue Engineering for Tissue and Organ Regeneration
18
Zimmermann, W. H., M. Didie, et al. (2006). Heart muscle engineering: an update on cardiac
muscle replacement therapy. Cardiovasc Res 71(3): 419-429.
Zimmermann, W. H., I. Melnychenko, et al. (2006). Engineered heart tissue grafts improve
systolic and diastolic function in infarcted rat hearts. Nat Med 12(4): 452-458.
Zimmermann, W. H., K. Schneiderbanger, et al. (2002). Tissue engineering of a differentiated
cardiac muscle construct. Circ Res 90(2): 223-230.
2
Cardiac Muscle Engineering: Strategies to
Deliver Stem Cells to the Damaged Site
Giancarlo Forte1, Stefania Pagliari2, Francesca Pagliari2,
Paolo Di Nardo2 and Takao Aoyagi1
1Biomaterials Center, International Center for Materials Nanoarchitectonics (MANA),
National Institute for Materials Science (NIMS), Tsukuba,
2Laboratorio di Cardiologia Molecolare e Cellulare, Dipartimento di Medicina Interna,
Università di “Tor Vergata”, Roma,
1Japan
2Italy
1. Introduction
In healthy human hearts, only 10-20% of the total cells are contractile cardiomyocytes and, at
the age of 25 years, no more than 1% of them are annually substituted by progenitor cells,
this percentage reducing to less than 0.5% at the age of 75. In total, less than 50% of
cardiomyocytes are renewed during a normal human life span [1]. For this reason, the topic
of cardiac repair is among the major challenges for the tissue engineers worldwide. In fact,
cardiac diseases are a predominant cause of mortality and morbidity in industrialized
countries, despite the recent advancements achieved in pharmacological treatment and
interventional cardiology procedures. Nonetheless, end-stage heart failure management still
relies on organ transplantation as unique approach, and, notwithstanding the use of massive
immunosuppressive drugs, still a percentage falling within 20%-40% of patients encounters
immune rejection during the first year post-transplant [2]. Among the patients not facing
severe immune rejection, almost 70% is forced to retire or reduce their working activity,
their survival rate falling below 70% during the first five years post organ transplantation
[3]. Last, but not least, the economic impact of cardiovascular diseases and stroke has been
estimated in 2010 at $503.2 billion [4].
Currently, post-infarction myocardial revascularization protocols include the administration
of raw bone marrow stem cells, while a number of clinical trials have been performed or are
currently in progress in which different cell subsets are implanted in the damaged tissue by
means of surgical techniques. The results of such trials are still controversial. In fact, when
autologous skeletal myoblasts were injected into the heart of patients suffering from
ischemic cardiomyopathy, the modest functional improvement obtained was impaired by
the arising of arrhythmia events, thus requiring the adoption of a pacemaker [5]. On the
other side, intracoronary administration of bone marrow mesenchymal stem cells resulted
in minimal improvements in cardiac contractile function in patients with dilated
cardiomyopathy [6]. These mild results were mostly ascribed to a paracrine effect exerted on
host tissue, rather than to a direct contribution of stem cells to the contractile activity.
Tissue Engineering for Tissue and Organ Regeneration
20
Thus, among the criticisms to be challenged before efficient cell therapy protocols for
cardiac diseases can be setup, the choice of the appropriate cell subset to generate new
vessels and contractile cardiomyocytes, as well as the route of cell delivery remain key steps.
The solution of such problems requires additional efforts in basic research to clarify the
processes leading to stem cell differentiation as well as technological advancements to setup
efficient protocols to implant the cells.
In principle, adult stem cells could be extracted from patient’s own tissues and expanded in
culture by means of well-known techniques (Figure 1).
Fig. 1. Cardiac Tissue Engineering paradigm. Adult stem cells can be harvested, purified
from the patient and expanded in culture. Such cells can be delivered to the injured heart by
injection (intramural or through bloodstream with or without injectable carriers), or in the
form of solid bio-constructs. Stem cell-derived bio-constructs can be obtained by culturing
the cells on scaffolds or by scaffold-free technology
Nonetheless, a number of issues should be challenged before safe procedures to
manipulate stem cells in vitro for cardiac transplant can be setup. In fact, stem cells should
be amplified in vitro to reach a critical number (Figure 1). During this passage, malignant
transformation is likely to occur in ex vivo cells when standard culture conditions are
adopted to expand stem cells [7, 8]. On the other side, stem cells could encounter
senescence after a short number of passages in vitro [9]. Moreover, the use of animal-
derived supplements during the phase of cell expansion would hinder the use of stem
cells for cardiac cell therapy.
Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 21
The employment of autologous stem cells would avoid the problem of immune rejection
and the need for immune-suppressive drugs, while, in the treatment of pathologies for
which a genetic basis is suspected the use of autologous cells is hampered. As far as the use
of autologous cells is concerned, the possibility that a significant patient-to-patient
variability in stem cell quality exists should be taken into account [10]. Finally, the use of
cellular and tissue-based products in human disease therapy is subjected to regulations
issued by the European Union and Food and Drug Administration (FDA) aimed at
establishing classification criteria for advanced therapy medicinal products (ATMP). In
particular, the European Regulation states that human cells to be used in cell therapy have
to comply with the principles of Good Manufacturing Practice (GMP) protocols [11, 12].
2. Adult stem cells for cardiac repair
A number of stem cells and progenitors have been so far proposed for cardiac repair, due to
the inability of cardiomyocytes to proliferate after birth [1]. Among the cell sources
challenged for the possibility to produce new cardiomyocytes, skeletal myoblasts have
proven to be able to acquire a contractile phenotype in vitro [13]. Moreover, when implanted
in vivo in a canine model of dilated cardiomyopathy (DCM), they attenuated cardiac
remodeling [14]. This result is likely to be due to the fusion of skeletal myoblasts with the
surrounding myocardium rather than to direct cell differentiation, as suggested by in vitro
experiments [15]. As discussed in the following section, clinical trials demonstrated that
skeletal myoblasts are not able to couple electrically with host tissue, leading to arrhythmia
events [5].
The role of hematopoietic stem cells (HSC) in cardiac repair has been investigated by several
research groups and their contribution to cardiac regeneration in vivo has been heavily
debated, being the ability of HSC to transdifferentiate to other lineages still questionable.
Indeed, evidence of the ability of bone marrow-derived c-kit+ HSC to help cardiac tissue
healing has been given using two different approaches: c-kit+ cells were (i) either delivered
to the infarcted site by intramural injection [16] or (ii) mobilized from bone marrow through
growth factor administration [17]. More recently, elegant experiments compellingly clarified
that HSC are not able to acquire contractile phenotype in vivo [18-20]. Nonetheless, a subset
of bone marrow hematopoietic precursors expressing CD34 and CD133 has been proven to
contain endothelial progenitors. Thus, they have been tested for revascularization protocols
in hind limb ischemic animals and could be proposed for cardiac infarction therapy [21]. On
the other hand, the results obtained in preliminary investigations in which another bone
marrow-derived stem cell subset, mesenchymal stem cells (BM-MSC or MSC) were
challenged as a candidate for cellular cardiomyoplasty, raised great enthusiasm for such a
cell subpopulation. Recent studies clarified that the direct contribution of MSC to cardiac
repair in terms of production of new contractile cells is minimal if any, while a paracrine
effect on the diseased tissue of such cells is universally recognized [22]. Such cells are also
appealing for their ability to induce a certain degree of immune tolerance [23].
The presence of a small reservoir of cardiac resident progenitor cells (CPC or CSC) has been
recently demonstrated in human as well as in other mammals’ heart [24]. Such tissue-
resident cells participate in myocardial homeostasis and retain a limited regenerative
capacity throughout organism lifespan [1]. All the subsets so far identified through the
expression of stemness markers (c-kit+, Sca-1+, Islet-1+) demonstrated the ability to give
birth to new contractile cells in vitro, while only c-kit+, Sca-1+ progenitors were shown to be
Tissue Engineering for Tissue and Organ Regeneration
22
involved in post-natal cardiac tissue homeostasis in vivo [25]. In fact, the presence of Islet-1+
cells appears to be limited to fetal life and their contribution to the endogenous program of
cardiovascular repair is still unknown; on the other hand, the very low number of c-kit+ and
Sca-1+ cells in the myocardium is considered the limiting factor of cardiac regeneration [26].
Furthermore, among the adult stem cells, a novel “artificial” subset can be recognized:
induced pluripotent stem cells (iPSC, Figure 2). This cell type can be produced in vitro by
transducing somatic cells with a combination of transcription factors able to induce the
nuclear reprogramming of differentiated cells. These cells, which display the functional
features of pluripotent embryonic stem cells, have been credited of the ability to produce
new cardiomyocytes. They could thus be the source of autologous, although genetically
modified, patient-specific contractile cells [27]. Moreover, the possibility to directly obtain
functional cardiomyocytes by the genetic reprogramming of postnatal cardiac or dermal
fibroblasts has been demonstrated [28]. Such a result was firstly obtained in vitro but also
when the cells were transplanted into mouse hearts one day after transduction of
transcription factors (GATA-4, MEF-2c, Tbx-5) known to be involved in cardiac muscle
development. Nonetheless, the reprogramming and differentiation efficiency of these cells
appears to be really low, thus requiring an efficient purification step before they can be
implanted in vivo. Additionally, safety concerns due to the use of genetically modified cells
and / or viral vectors remain.
Fig. 2. Induced Pluripotent Stem Cell Generation. Induced pluripotent Stem Cells (iPSC) can
be generated by reprogramming somatic cells through their transduction with four
transcription factors. iPSC share functional similarities with Embryonic Stem Cells (ESC)
and can be differentiated towards cardiomyocytes, thus representing an autologous source
of contractile cells
Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 23
3. Stem cell delivery to the injured heart
As previously said, cell route of delivery to damaged heart represents the major topic in
the setup of efficient, minimally invasive techniques to treat cardiac pathologies.
Recently, a number of techniques to deliver stem cells to the injured site have been
proposed but questions remain regarding the optimal approach able to favor high cell
retention, differentiation rate and clinically relevant improvement in cardiac
performance.
a) Direct injection
Stem cell direct intramural injection, including trans-epicardial and trans-endocardial cell
injection, is the elective strategy for patients with severe occlusion of coronary vessels. In
particular, trans-epicardial approach consists in the direct injection of a high number of cells
into the infarcted area or around the border zone. Endocardial stem cell injection is
performed using catheters such as MyoStarTM injection catheter (Biosense Webster)
integrated with imaging systems like NOGA® system (Cordis Corp., Warren, NJ, USA),
which allows real-time three-dimensional reconstruction of left ventricle as well as the
targeting and functional assessment of specific myocardial area [29]. Such procedures are
highly invasive since they require open-heart surgery and gave contrasting results so far.
For example, pre-clinical studies performed on experimental animals demonstrated that,
although a certain extent of cardiac repair was achieved when bone marrow Stro-3+
perivascular cells are implanted in vivo, the cells vanished from the application site within
few days [30]. In other reports, when Sca-1+ cardiac resident stem cells were injected in
infarction border zone, a modest but significant improvement in cardiac function was
reported, with evidence of cell engraftment and differentiation [31]. Finally, in another
pre-clinical study, bone marrow-derived c-kit+ cells were shown to repair entire
ventricular areas while massively engrafting and differentiating in contractile and
vascular figures in vivo [32]. Of interest, independent groups already demonstrated that c-
kit+ bone marrow-derived hematopoietic stem cells fail to acquire contractile phenotype
when implanted in diseased myocardium [19, 20]. Such discrepancies are not surprising
since different stem cell subsets or preparation protocols were probably used in these
studies.
Stem cells can be delivered intravenously to the heart, through coronary arteries or even
through retrograde coronary sinus. The major drawback of stem cells being infused through
peripheral venous system seems to be the low retention of cells into infarcted area. Results
obtained in pre-clinical animal models showed that this minimally invasive approach results
in a significant percentage of injected cells being sequestered in lungs, liver or spleen, due to
blood flow [33]. On the other hand, intracoronary or retrograde coronary sinus infusion of
the cells are mainly performed after acute myocardial infarction using an angioplasty
balloon and high pressure to deliver cells to heart muscle [34]. The coronary route was
proven to be free of stem cell systemic delivery, while a limited number of cells could be
found in the infarcted area [35].
Finally, an interesting attempt with stem cells being injected into the pericardial cavity has
been proposed. By this means, a higher number of cells could be deposited and retained
in the pericardial cavity, while migration across the visceral pericardium is required
(Table 1).
Tissue Engineering for Tissue and Organ Regeneration
24
Table 1. Advantages and disadvantages of injecting stem cells by intravenous,
intracoronary, intramyocardial, retrograde coronary sinus or intra-pericardial route
b) Injectable scaffolds
Injectable scaffolds are defined as materials offering the unique solution of replacing
damaged myocardial ECM and/or delivering cells directly to the infarcted region while
holding the potential for minimally invasive delivery [36]. Such scaffolds can be composed
of biocompatible microspheres or in situ gelling materials having reasonable dimensions as
to surpass capillary barrier. They are considered a promising tool for stem cell delivery to
damaged myocardium. In situ gelling materials are generally made of components of
extracellular matrix (ECM), which are induced to a transition after being implanted in situ.
Complex injectable gelling materials have been prepared by decellularization technique out
of ventricular or epicardial ECM, thus possibly avoiding animal-derived components and
paving the way to the definition of patient-specific treatments.
The use of injectable, synthetic microspheres has already been proven promising in the
treatment of neurological diseases in vivo [37]. Recently the possibility of using injectable
scaffolds in cardiac cell therapy has been explored by interfacing murine mesenchymal
(mMSC) and cardiac stem cell (mCSC) lines with poly-lactic acid (PLA) microspheres
having a diameter of 30 and 100 μm. Preliminary in vitro experiments demonstrated that
such cells can be grown onto PLA microspheres while preserving their phenotype, but the
formation of cell clumps can hamper the application of this technique [38]. The use of
dynamic seeding techniques (i.e. bioreactors) would favor a more homogeneous distribution
of the cells. An interesting approach has been recently proposed to deliver human
mesenchymal stem cells to the injured myocardium: RGD-modified alginate microsphere
(diameter: 200-700 μm) encapsulation of hMSC was setup. In vitro experiments showed that
hMSC could survive, proliferate and migrate through the porous material. When
intramyocardially injected in a rat model of myocardial infarction by left anterior
descendant coronary (LAD) ligation, cell-loaded alginate microspheres promoted
angiogenesis and prevented LV negative remodeling [39]. Nonetheless, few human cells
were found in the injection area after few days, while microbead remains were still present
Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 25
within host myocardium 10 weeks after the injection. The aspect of microbead resorption
should thus be addressed before clinical perspectives could be foreseen.
c) Scaffold-based technology
The possibility of using biocompatible scaffolds to deliver stem cells to the injured heart has
been explored by a number of independent research groups so far. The scaffolds proposed
are natural of synthetic but when designing cardiac-specific constructs, a number of
requirements should be fulfilled. For example, it cannot be neglected that myocardial
contractile function relies on the transmission of electrical and mechanical forces throughout
a functional syncytium. So, the integrity of the tissue has to be preserved. For this reason, a
cardiac-specific scaffold should comply with tissue architecture and thus be deformable
enough to indulge and, if possible sustain cardiac contraction. Moreover, as far as stem cell
engraftment is concerned, scaffolds should be able to start at least cell alignment and
commitment to favor stem cell electromechanical coupling with host tissue. In this respect,
the work of Mandoli and collaborators using Cerium Oxyde nanoparticles to affect poly-
lactic acid film surface and obtain a controlled nanorugosity appears intriguing [40]. In fact,
far from being a noxious compound for stem cells, ceria was able to induce cardiac stem cell
alignment and growth. Nonetheless, cardiac tissue is extremely complex and highly
demanding in terms of blood supply and catabolite removal, so that porous scaffolds that
could allow microvascular branches formation and oxygen perfusion are to be preferred. To
fulfill such requirements, the first attempts were performed by the group of Thomas
Eschenhagen. Neonatal cardiomyocytes were seeded in Collagen I + Matrigel to produce
Engineered Heart Tissue (EHT). Continuous contractile activity up to 1 week in vitro as well
as cell survival and integration in vivo in singenic rat hearts were reported [41]. In another
attempt, anisotropic accordion-like honeycomb scaffolds were prepared by excimer laser
microablation using poly(glycerol sebacate) as an elastomeric tool to mimic anisotropic
cardiac muscle stiffness distribution [42]. Although the authors demonstrated that such
scaffolds promote neonatal rat cardiomyocyte alignment and contraction, in vivo testing has
not been performed so far. The same material has been utilized to produce elastomeric
patches on which human embryonic stem cell-derived cardiomyocytes were grown,
showing that it is indeed possible to observe spontaneous beating activity in vitro up to 3
months [43]. Such patches were shown to be suitable as delivery systems and, when sutured
in the absence of cells onto healthy rat left ventricle, they did not affect cardiac contractile
activity. More basic studies were also conducted to study the ability of stem cells to interface
with different synthetic and natural materials. In this respect, few research groups focused
on the possibility to drive a certain extent of stem cell commitment through tailoring
scaffold physical and chemical properties, independently of biological cues. In this respect, a
common agreement on the ability of stem cells to sense substrate rugosity and elasticity has
been reached [44]. Thus, in order to rule out the occurrence of spontaneous events of
differentiation in implanted cells, the possibility to induce in vitro stem cell commitment on
scaffolds towards a desired phenotype is being investigated. Indeed, Engler and
collaborators compellingly demonstrated that the possibility to affect stem cell fate
determination by simply tuning substrate elasticity as to match tissue-specific stiffness,
exists. Recently, this concept has been corroborated by other research groups, showing that
cardiac resident progenitors (Sca-1+ CPC) can be committed to cardiac phenotype by the
physico-chemical signals arising from matrix, but biological factors are needed to complete
the differentiation process [45, 46].
Tissue Engineering for Tissue and Organ Regeneration
26
d) Preparation of thick cardiac substitutes by Scaffold-free technology
To overcome the problem of poor cell retention reported in cell injection experiments in the
heart [30] and avoid the release of possibly harmful scaffold byproducts, scaffold-free
technology has been developed, in which cells are grown in a monolayer onto thermo-
responsive surfaces and easily detached in the form of cell sheet by lowering the
temperature [47]. Such technology takes advantage of the ability of polymers like poly-N-
isopropylacrylamide (PNIPAAm) to shift between hydrophobic and hydrophilic status
when the temperature ranges from 37ºC to 32ºC. Cell sheets can be serially stacked to obtain
multilayered scaffoldless constructs (Figure 3). Such an approach has already been applied
to obtain cell sheets composed of rodent [48, 49] and human [50] cells. Given the need for
thick cardiac substitutes suited to comply with cardiac muscle continuous contractility,
thermo-responsive technology has been envisaged as a possible answer to the lack of heart
donors. Pre-clinical trials performed onto experimentally infarcted animals demonstrated
that when a murine adipose-derived monolayer sheet is leant onto injured myocardium, it
can be retained and help tissue repair [48]. Similarly, striking results are obtained when a
Sca-1+ cardiac progenitor cell-derived sheet is used [49]. Finally, an interesting approach has
been recently proposed to deliver cardiac stem cells cultured in the form of cardiospheres to
the injured heart: cardiospheres were embedded into a cardiac stromal cell-derived sheet
obtained by using poly-lysine/ collagen IV-coated dishes [51]. The formation of mature
vessels as well as new cardiomyocytes in vivo was reported after 3 weeks.
Fig. 3. Generation of scaffoldless multilayered bio-constructs by means of thermo-
responsive technology: cells grown in a monolayer onto thermo-responsive poly-N-
isopropilacrylamide (PNIPAAm)-coated dishes can be detached by lowering the
temperature below 32°C. At 37°C the surface is highly hydrophobic and allows cell
adhesion. When the temperature is lowered, PNIPAAm becomes hydrophilic, the cell sheet
is detached and extracellular matrix (ECM) preserved. Multilayered cell sheets can be
obtained by serially stacking monolayered sheets
Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 27
4. Clinical trials
In the attempt to transfer bench experience to bedside, a number of clinical trials in which
different stem cell or progenitor subsets are used have been approved (see
http://www.clinicaltrials.gov). Most of them are still in the recruitment phase while some
already gave indications and preliminary results. Since most of the ongoing trials are based
on the injection of raw stem cell preparations (mostly bone marrow-derived cells), the time
and route of cell application remain the key problems to be addressed before proceeding to
routine clinical practice. In this respect, recent animal experiments demonstrated that the
acute phase of myocardial infarction is probably not suitable for stem cell engraftment and
differentiation [52]. Therefore, the right moment in which stem cells should be delivered is
to be studied. An overview on some of the ongoing clinical trials is given below.
1. MAGIC (Myoblast Autologous Grafting in Ischemic Cardiomyopathy). In one of the
first phase II clinical trials setup to study the possibility to use stem cells to treat cardiac
pathologies, ninety-seven (97) patients undergoing coronary artery bypass grafting
(CABG) were enrolled. 400-800 X 106 autologous myoblasts harvested from patient
muscle biopsy were implanted in the akinetic area of ventricular wall 21 days after in
vitro culture. The follow-up after 30 days and 6 months demonstrated the arising of
arrhythmia events, thus requiring the implantation of pacemaker. Moreover, no cardiac
function improvement was reported. Such negative results were ascribed to the
inability of skeletal myoblasts to balance cell death and achieve complete
electromechanical integration with the recipient myocardium. Finally, skeletal myoblast
administration was reported to determine no enhancement in major cardiac adverse
events and mild effects on left ventricular remodeling process [53, 54]. More recently,
final results from SEISMIC [Safety and Effects of Implanted (Autologous) Skeletal
Myoblasts (MyoCell) Using an Injection Catheter] Trial, a phase II-a study
encompassing 40 patients experiencing congestive heart failure and receiving
percutaneous intramyocardial injection of autologous skeletal myoblasts, reported the
feasibility and safety of this procedure without significant arrhythmogenic events
recorded at 6-month follow-up with respect to control groups, although left ventricular
ejection fraction did not result significantly improve. These encouraging results suggest
that myoblast cell therapy could be considered as a potential effective treatment when
associated with standard medical therapy in patients with previously implanted cardiac
defibrillators [55].
2. TOPCARE-CHD, -AMI, -DCM (Transplantation of Progenitor Cells and
Regeneration Enhancement in Acute Myocardial Infarction, Chronic Stable Ischemic
Heart disease or Dilated Cardiomyopathy). In this complex clinical trial, a total of 346
patients were classified to CHD, AMI or DCM pathologies and infused either with bone
marrow cells (BMCs), blood-derived stem cells, or no infusion. In TOPCARE-CHD, 121
patients (mean age: 59) with chronic stable ischemic heart disease (CHD) were treated.
Although complications occurred in 21% of the patients during 3 months follow-up,
BMC intracoronary administration was related with a reduction of both brain and atrial
natriuretic peptide (NTP) serum levels (indicators of LV remodelling process) in the
remaining population (79%), especially in patients with higher NTP levels at baseline
and receiving a greater BMC number with a high functional capacity. Moreover, these
results were also correlated with a left ventricular ejection fraction (LVEF) increase and
better survival during the further follow-up, suggesting that cell therapy could be
Tissue Engineering for Tissue and Organ Regeneration
28
associated with cardiac function enhancements in patients with advanced chronic post-
infarction heart failure [56]. Similarly, two hundred and four (204) patients were treated
using bone-marrow-derived progenitor cells directly into the infarct artery three to
seven days after an acute myocardial infarction (AMI). A statistically significant 2.5%
improvement in left ventricular ejection fraction at four months was reported for
patients randomized to the bone marrow injection [57]. Finally, intracoronary infusion
of bone marrow cells was performed in 33 patients with dilated cardiomyopathy
(DCM) by using an over-the-wire balloon catheter. Three month follow-up
demonstrated an improvement in left ventricular pump function while a modest
improvement in Brain Natriuretic Peptide (BNP) levels was reported after 1 year [6].
Importantly, the conditions chosen in the present clinical trial were representative of
different conditions (acute, chronic phase) encountered in the clinic. Unfortunately, no
clear indication on stem cell characterization or on their actual ability to regenerate
contractile cells is available.
3. TRACIA STUDY (Intracoronary Autologous Stem Cell Transplantation in ST
Elevation Myocardial Infarction). The phase II/ III clinical trial aimed at evaluating the
effects of intracoronary administration of adult stem cells on LV ejection fraction and
major adverse cardiovascular events (MACE) after 6 months follow-up. For this reason,
1-2 million CD34+ cells were injected through the infarct-related artery few days after
post-infarct angioplasty using an "over-the-wire" catheter in 80 patients aging from 20
to 75 years. The results of this study are still to be published.
4. Combined CABG and Stem-Cell Transplantation for Heart Failure. Intramyocardial
delivery of autologous bone marrow cells extracted from iliac crest and purified by
Ficoll centrifugation, during cardiac surgery for CABG intervention in 30 patients, as
compared to 30 patients undergoing CABG without cell infusion. Although information
on the number and characteristics of cells to be injected has not been given, the trial is
currently ongoing and the follow-up is scheduled in 6-12 months
(http://clinicaltrials.gov).
5. POSEIDON-Pilot Study (The Percutaneous Stem Cell Injection Delivery Effects on
Neomyogenesis Pilot Study) Poseidon-pilot Study is a phase I/ II multi-center trial in
which the trans-endocardial injection of autologous Mesenchymal Stem Cells (20-, 100-,
200 X 106) is compared to autologous non-purified bone marrow cells and to allogeneic
human Mesenchymal Stem Cells. The implant is performed during cardiac
catheterization using the Biocardia Helical Infusion Catheter in fifty (50) patients
suffering from chronic ischemic left ventricular dysfunction secondary to myocardial
infarction. The data collection is currently ongoing.
6. SCIPIO (Cardiac Stem Cell Infusion in Patients With Ischemic CardiOmyopathy).
This phase I clinical trial is aimed at assessing the safety and effectiveness of
intracoronary autologous cardiac stem cell therapy. As such, forty (40) patients
suffering from ischemic cardiomyopathy are exposed to intracoronary injection of
cardiac resident stem cells (CSC). Cardiac stem cells are harvested from right atrial
appendages and selected for c-kit expression, cultured and expanded in vitro prior to
injecting them via intracoronary route, three to five months after CABG surgery. The
hypothesis is that CSC infused into nonviable myocardial segments will regenerate
infarcted myocardium by differentiating into cardiomyocytes and vascular cells. The
preliminary results are encouraging: in the nine patients treated at four months after
Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 29
CSC infusion, LVEF increased from 31.3 + 2.5 percent before CSC infusion to 38.8 + 3.2
percent four months after CSC infusion. Moreover, in the five patients in whom data
are available at 12 months after stem cell infusion, the improvement in LVEF observed
at four months was even greater, averaging 15% at 12 months. The follow-up is
scheduled in 1,5 years.
7. ALCADIA (AutoLogous Human CArdiac-Derived Stem Cell to Treat Ischemic
cArdiomyopathy). In this phase I, multicenter clinical trial, a rather different approach
is followed. In fact, patients’ own cardiac stem cells obtained by endo-myocardial
biopsies are delivered by a single intramyocardial injection. The cells injected are 0.5
million cells/kg (patient body weight) and their engraftment should be favored by the
concomitant implantation of gelatin hydrogel sheet releasing human recombinant beta
Fibroblast Growth Factor (bFGF), during CABG surgery. The study has been designed
to treat refractory heart failure, ischemic cardiomyopathy or ventricular dysfunction
cases. Importantly, this is the first clinical trial, to our knowledge, in which a human
recombinant growth factor is used. Unfortunately, the number of enrolled patients is
limited to six (6).
8. REGEN-IHD (Bone Marrow Derived Adult Stem Cells for Chronic Heart Failure). In
this phase II/ III study, granulocyte-colony stimulating factor (G-CSF) is
subcutaneously administered for 5 days to patients with heart failure secondary to
ischemic heart disease to mobilize CD34+ bone marrow stem cells. A concomitant
intracoronary or intramyocardial administration of bone marrow derived stem cells is
performed. The number of enrolled patients is high (165) and the aim of the study is to
compare the effects of G-CSF and autologous bone marrow progenitor cell infusion on
the quality of life and left ventricular function in the patients. The follow-up timepoint
is scheduled in 6-12 months.
A number of papers reporting statistical analyses and comparisons among the clinical trials
in which stem and progenitor cells have been adopted are currently available. [For further
information, please refer to www.clinicaltrials.gov].
5. Conclusions
The possibility to treat cardiac diseases by cell therapy techniques is an extraordinary
promise. While a number of different approaches has been so far proposed to setup
minimally invasive techniques for cardiac repair, few of them being already in the clinical
experimental phase, basic questions still need to be addressed. In fact, the molecular
processes leading to cardiac differentiation still need to be fully clarified, while the impact of
novel, genetically modified cell types obtained from adult differentiated cells on cardiac
microenvironment deserve further investigations. More importantly, the seek to identify
suitable delivery systems (i.e. scaffolds) able to foster stem cell survival, growth and
differentiation, while degrading without negative effects as the formation of new tissue
occurs is still open. A look at the literature reveals that an impressive effort to translate the
information obtained by in vitro and pre-clinical studies to the bedside is being produced. In
particular, a number of stem cell subsets, which have been previously tested in vitro and in
animal models, are currently being tested in phase I, II clinical trials. As expected, the
predominant delivery system used in the ongoing clinical trials is intracoronary or
intramural injection of stem cells. The possibility to adopt tissue engineering techniques to
Tissue Engineering for Tissue and Organ Regeneration
30
design patient-specific cardiac substitutes containing synthetic or natural scaffolds is still far
from being taken into consideration for clinical application, since any single formulation will
have to be approved before clinical testing.
6. References
[1] Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, Walsh S, Zupicich J,
Alkass K, Buchholz BA, Druid H, Jovinge S, Frisén J. Evidence for Cardiomyocyte
Renewal in Humans. Science 2009: 324; 98-102.
[2] Patel JK, Kobashigawa JA. Should we be doing routine biopsy after heart transplantation
in a new era of anti-rejection? Curr Opin Cardiol 2006; 21: 127-131.
[3] Hertz MI, Aurora P, Christie JD, Dobbels F, Edwards LB, Kirk R, Kucheryavaya AY,
Rahmel AO, Rowe AW, Stehlik J, Taylor DO. Scientific Registry of the International
Society for Heart and Lung Transplantation. J Heart Lung Transplant 2009: 28; 989-
1049.
[4] American Heart Association. Heart disease and stroke statistics-2010 update. Dallas,
Texas: American Heart Association; 2010. © 2010, American Heart Association.
[5] Menasché P, Alfieri O, Janssens S, McKenna W, Reichenspurner H, Trinquart L, Vilquin
JT, Marolleau JP, Seymour B, Larghero J, Lake S, Chatellier G, Solomon S, Desnos
M, Hagège AA. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy
(MAGIC) trial: first randomized placebo-controlled study of myoblast
transplantation. Circulation. 2008; 117: 1189-1200.
[6] Fischer-Rasokat U, Assmus B, Assmus B, Seeger FH, Honold J, Leistner D, Fichtlscherer
S, Schächinger V, Tonn T, Martin H, Dimmeler S, Zeiher MA. A pilot trial to assess
potential effects of selective intracoronary bone marrow-derived progenitor cell
infusion in patients with nonischemic dilated cardiomyopathy: final 1-year results
of the transplantation of progenitor cells and functional regeneration enhancement
pilot trial in patients with nonischemic dilated cardiomyopathy. Circ Heart Fail
2009; 2: 417-423.
[7] Foudah D, Redaelli S, Donzelli E, Bentivegna A, Miloso M, Dalprà L, Tredici G.
Monitoring the genomic stability of in vitro cultured rat bone-marrow-derived
mesenchymal stem cells. Chromosome Res. 2009; 17: 1025-1039.
[8] Momin EN, Vela G, Zaidi HA, Quiñones-Hinojosa A. The Oncogenic Potential of
Mesenchymal Stem Cells in the Treatment of Cancer: Directions for Future
Research. Curr Immunol Rev. 2010; 6:137-148
[9] Vacanti V, Kong E, Suzuki G, Sato K, Canty JM, Lee T. Phenotypic changes of adult
porcine mesenchymal stem cells induced by prolonged passaging in culture. J Cell
Physiol 2005: 194-201.
[10] Itzhaki-Alfia A, Leor J, Raanani E, Sternik L, Spiegelstein D, Netser S, Holbova R,
Pevsner-Fischer M, Lavee J, Barbash IM. Patient characteristics and cell source
determine the number of isolated human cardiac progenitor cells. Circulation 2009;
120: 2559-2566.
[11] Regulation (EC) No 1394/2007 of the European Parliament and of the Council of 13
November 2007 on advanced therapy medicinal products and amending Directive
2001/83/EC and Regulation (EC) No 726/2004.
[12] Food and Drug Administration 21 CFR 1271 (2006).
Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 31
[13] Formigli L, Francini F, Tani A, Squecco R, Nosi D, Polidori L, Nistri S, Chiappini L,
Cesati V, Pacini A, Perna AM, Orlandini GE, Zecchi Orlandini S, Bani D.
Morphofunctional integration between skeletal myoblasts and adult
cardiomyocytes in coculture is favoured by direct cell-cell contacts and relaxin
treatment. Am J Physiol Cell Physiol 2005; 288: C795-804.
[14] Hata H, Matsumiya G, Miyagawa S, Kondoh H, Kawaguchi N, Matsuura N, Shimizu T,
Okano T, Matsuda H, Sawa H. Grafted skeletal myoblasts sheets attenuate
myocardial remodelling in pacing-induced canine heart failure model. J Thorac
Cardiovasc Surg 2009; 138: 460-467.
[15] Reinecke H, Minami E, Poppa V, Murry CE. Evidence for fusion between cardiac and
skeletal muscle cells. Circ Res 2004; 94: e56-e60.
[16] Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R,
Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate
infarcted myocardium. Nature 2001; 410: 221-229.
[17] Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, Nadal-Ginard B, Bodine
DM, Leri A, Anversa P. Mobilized bone marrow cells repair the infarcted heart,
improving function and survival. Proc Natl Acad USA 2001; 98: 10344-10349
[18] Wagers AJ, Sherwood RI, Christensen JL, Weissman IL. Little evidence for
developmental plasticity of adult hematopoietic stem cells. Science 2002; 297: 2256-
2259.
[19] Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M,
Pasumarthi KB, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams
DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac
myocytes in myocardial infarcts. Nature. 2004; 428: 664-668.
[20] Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC.
Haematopoietic stem cells adopt mature haematopoietic fates in ishaemic
myocardium. Nature 2004; 428: 668-673.
[21] Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B,
Schatteman G, Isner JM. Isolation of putative progenitor cells for angiogenesis.
Science 1997; 275: 964-967.
[22] Nesselmann C, Ma N, Bieback K, Wagner W, Ho A, Konttinen YT, Zhang H, Hinescu
ME, Steinhoff G. Mesenchymal stem cells and cardiac repair. J Cell Mol Med 2008;
12: 1795-1810.
[23] Amado L, Saliaris A, Schuleri K, St. John M, Xie JS, Cattaneo S, Durand DJ, Fitton T,
Kuang JQ, Stewart G, Lehrke S, Baumgartner WW, Martin BJ, Heldman AW, Hare
JM. Cardiac repair with intramyocardial injection of allogenic mesenchymal stem
cells after myocardial infarction. Proc Natl Acad Sci USA 2005; 102: 11474-11479.
[24] Quaini, F., Urbanek, K., Beltrami, AP, Finato, N, Beltrami, CA, Nadal-Ginard, B,
Kajstura, J, Leri, A, and Anversa, P. Chimerism of the transplanted heart. N Engl J
Med 2002; 346: 5-15.
[25] Bearzi C, Rota M, Hosoda T, Tillmanns J, Nascimbene A, De Angelis A, Yasuzawa-
Amano S, Trofimova I, Siggins RW, Lecapitaine N, Cascapera S, Beltrami AP,
D’Alessandro DA, Zias E, Quaini F, Urbanek K, Michler RE, Bolli R, Kajstura J, Leri
A, Anversa P. Human cardiac stem cells. Proc Natl Acad Sci U S A. 2007; 104: 14068 –
14073.
Tissue Engineering for Tissue and Organ Regeneration
32
[26] Di Nardo P, Forte G, Ahluwalia A, Minieri M. Cardiac progenitor cells: Potency and
control. J. Cell. Physiol. 2010; 224: 590–600.
[27] Yamanaka S, Blau HM. Nuclear reprogramming to a pluripotent state by three
approaches. Nature 2010; 465: 704-712.
[28] Ieda M, Fu JD, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, Srivastava D.
Direct reprogramming of fibroblasts into functional cardiomyocytes by defined
factors. Cell. 2010; 142: 375-386.
[29] Perin EC and Lopez J. Methods in stem cell delivery in cardiac diseases. Nat Clinic Pract
Cardiovasc Med 2006: 3 S1.
[30] Dixon JA, Gorman RC, Stroud RE, Bouges S, Hirotsugu H, Gorman JH 3rd, Martens TP,
Itescu S, Schuster MD, Plappert T, St John-Sutton MG, Spinale FG. Mesenchymal
cell transplantation and myocardial remodeling after myocardial infarction.
Circulation. 2009; 120: S220-S229.
[31] Smits AM, van Vliet P, Metz CH, Korfage T, Sluijter JPG, Doevendans PA, Goumans
MJ. Human cardiomyocyte progenitor cells differentiate into functional mature
cardiomyocytes: an in vitro model for studying human cardiac physiology and
pathophysiology. Nat Protoc 2009; 4: 232-243.
[32] Rota M, Kajstura J, Hosoda T, Bearzi C, Vitale S, Esposito G, Iaffaldano G, Padin-
Iruegas ME, Gonzalez A, Rizzi R, Small N, Muraski J, Alvarez R, Chen X, Urbanek
K, Bolli R, Houser SR, Leri A, Sussman MA, Anversa P: Bone marrow cells adopt
the cardiomyogenic fate in vivo. Proc Natl Acad Sci U S A 2007, 104: 17783-17788.
[33] Gao J, Dennis JE, Muzic RF, Lundberg M, Caplan L. The dynamic in vivo distribution of
bone marrow-derived mesenchymal stem cells after infusion. Cells Tissues Organs
2001; 169: 12-20.
[34] Bui QT, Gertz ZM, Wilensky RL. Intracoronary delivery of bone-marrow-derived stem
cells. Stem Cell Res Ther. 2010; 1:29-35.
[35] Bartunek J, Vanderheyden M, Vandekerckhove B, Mansour S, De Bruyne B, De Bondt P,
Van Haute I, Lootens N, Heyndrickx G, Wijns W. Intracoronary injection of CD133-
positive enriched bone marrow progenitor cells promotes cardiac recovery after
recent myocardial infarction: feasibility and safety. Circulation 2005; 112: 178–183.
[36] Singelyn JM, Christman KL. Injectable materials for the treatment of myocardial
infarction and heart failure: the promise of decellularized matrices. J Cardiovasc
Transl Res. 2010; 3: 478-486.
[37] Menei P, Montero-Menei C, Venier MC, Benoit JP. Drug delivery into the brain using
poly(lactide-co-glycolide) microspheres. Expert Opin Drug Deliv. 2005 2: 363-376.
[38] Forte G, Franzese O, Pagliari S, Pagliari F, Cossa P, Laudisi A, Di Francesco AM,
Fiaccavento R, Carotenuto F, Bonmassar E, Fiaccavento R, Minieri M, Di Nardo P.
Interfacing Sca-1pos Mesenchymal Stem Cells with Biocompatible Scaffolds with
Different Chemical Composition and Geometry. J Biomed Biotechnol 2009; doi:
10.1155/2009/910610.
[39] Yu J, Du KT, Fang Q, Gu Y, Mihardja SS, Sievers RE, Wu JC, Lee RJ. The use of human
mesenchymal stem cells encapsulated in RGD modified alginate microspheres in
the repair of myocardial infarction in the rat. Biomaterials. 2010; 31: 7012-7020
[40] Mandoli C, Pagliari F, Pagliari S, Forte G, Di Nardo P, Licoccia S, Traversa E. Stem cell
aligned growth induced by CeO2 nanoparticles in PLGA scaffolds with improved
bioactivity for regenerative medicine. Adv Funct Mater 2010; 20: 1617-1624.
Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 33
[41] Zimmermann WH, Melnychenko I, Wasmeier G, Didié M, Naito H, Nixdorff U, Hess A,
Budinsky L, Brune K, Michaelis B, Dhein S, Schwoerer A, Ehmke H, Eschenhagen
T. Engineered heart tissue grafts improve systolic and diastolic function in
infarcted rat hearts. Nat Med. 2006; 12: 452-458.
[42] Engelmayr GC Jr, Cheng M, Bettinger CJ, Borenstein JT, Langer R, Freed LE. Accordion-
like honeycombs for tissue engineering of cardiac anisotropy. Nat Mater. 2008; 7:
1003-1010.
[43] Chen QZ, Ishii H, Thouas GA, Lyon AR, Wright JS, Blaker JJ, Chrzanowski W,
Boccaccini AR, Ali NN, Knowles JC, Harding SE. An elastomeric patch derived
from poly(glycerol sebacate) for delivery of embryonic stem cells to the heart.
Biomaterials. 2010; 31: 3885-3893.
[44] Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage
specification. Cell. 2006; 126: 677-689.
[45] Forte G, Carotenuto F, Pagliari F, Pagliari S, Cossa P, Fiaccavento R, Ahluwalia A, Vozzi
G, Vinci B, Serafino A, Rinaldi A, Traversa E, Carosella L, Minieri M, Di Nardo P.
Criticatility of the biological and physical stimuli array inducing resident stem cell
determination. Stem Cells 2008; 26: 2093-2103.
[46] Pagliari S, Vilela-Silva AC, Forte G, Pagliari F, Mandoli C, Vozzi G, Pietronave S, Prat
M, Licoccia S, Ahluwalia A, Traversa E, Minieri M, Di Nardo P. Cooperation of
Biological and Mechanical Signals in Cardiac Progenitor Cell Differentiation. Adv
Mater 2010; 23: 514-518.
[47] Masuda S, Shimizu T, Yamato M, Okano T. Cell sheet engineering for heart tissue
repair. Adv Drug Deliv Rev. 2008; 60: 277-285.
[48] Miyahara Y, Nagaya N, Kataoka M, Yanagawa B, Tanaka K, Hao H, Ishino, Ishida H,
Shimizu T, Kangawa K, Sano S, Okano T, Kitamura S, Mori H. Monolayered
mesenchymal stem cells repair scarred myocardium after myocardial infarction.
Nat Med 2006; 12: 459-465.
[49] Matsuura K, Honda A, Nagai T, Fukushima N, Iwanaga K, Tokunaga M, Shimizu T,
Okano T, Kasanuki H, Hagiwara N, Komuro I. Transplantation of cardiac
progenitor cells ameliorates cardiac dysfunction after myocardial infarction in
mice. J Clin Invest. 2009; 119: 2204–2217.
[50] Arauchi A, Shimizu T, Yamato M, Obara T, Okano T. Tissue-engineered thyroid cell
sheet rescued hypothyroidism in rat models after receiving total thyroidectomy
comparing with nontransplantation models. Tissue Eng Part A. 2009; 15: 3943-3949.
[51] Zakharova L, Mastroeni D, Mutlu N, Molina M, Goldman S, Diethrich E, Gaballa MA.
Transplantation of cardiac progenitor cell sheet onto infarcted heart promotes
cardiogenesis and improves function. Cardiovasc Res. 2010; 87: 40-49.
[52] Chen YR, Li Y, Chen L, Yang XC, Su PX, Cai J. The infarcted myocardium does not
selectively promote embryonic stem cell differentiation into cardiomyocytes.
Cardiovasc. Pathol. 2010; doi:10.1016/j.carpath.2009.12.003.
[53] Menasché P. Stem cell therapy for heart failure: are arrhythmias a real safety concern?
Circulation. 2009; 119:2735-2740.
[54] Zenovich AG, Davis BH, Taylor DA. Comparison of intracardiac cell transplantation:
autologous skeletal myoblasts versus bone marrow cells. Handb Exp Pharmacol.
2007; 180: 117-165.
Tissue Engineering for Tissue and Organ Regeneration
34
[55] Duckers HJ, Houtgraaf J, Hehrlein C, Schofer J, Waltenberger J, Gershlick A, Bartunek J,
Nienaber C, Macaya C, Peters N, Smits P, Siminiak T, van Mieghem W, Legrand V,
Serruys PW. Final results of a phase IIa, randomised, open-label trial to evaluate
the percutaneous intramyocardial transplantation of autologous skeletal myoblasts
in congestive heart failure patients: the SEISMIC trial. EuroIntervention. 2011; 6: 805-
812.
[56] Assmus B, Fischer-Rasokat U, Honold J, Seeger FH, Fichtlscherer S, Tonn T, Seilfried E,
Schaechinger V, Dimmeller S, Zeiher AM; TOPCARE-CHD Registry.
Transcoronary transplantation of functionally competent BMCs is associated with a
decrease in natriuretic peptide serum levels and improved survival of patients with
chronic postinfarction heart failure: results of the TOPCARE-CHD Registry. Circ
Res 2007; 100: 1234-1241.
[57] Schaechinger V, Assmus B, Britten MB, Honold J, Lehmann R, Teupe C, Abolmaali ND,
Vogl TJ, Hofmann WK, Martin H, Dimmeler S, Zeiher AM. Transplantation of
progenitor cells and regeneration enhancement in acute myocardial infarction: final
one-year results of the TOPCARE-AMI trial. J Am Coll Cardiol 2004; 44: 1690-1698.
3
Cardiovascular Tissue Engineering
Based on Fibrin-Gel-Scaffolds
Stefan Jockenhoevel1 and Thomas C. Flanagan2
1Department of Tissue Engineering & Textile Implants,
AME-Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen,
2School of Medicine & Medical Science, Health Sciences Centre,
University College Dublin, Dublin
1Germany
2Ireland
1. Introduction
Cardiovascular disease is a major cause of death in the Western World. Novel drugs and
innovative devices have enhanced the quality of life for patients with cardiovascular
disease, but such treatments are not without limitations and complications. The major
constraint with these current treatments is the inability for growth, repair and remodeling of
the structure. The emergence of tissue engineering as an alternative therapy for
cardiovascular disease has generated an intensity of research into the development of many
components of the cardiovascular system, including heart valves, small-calibre vascular
grafts and biological stent materials. The composition of the biomaterial used as a support
for the developing cardiovascular structure is a key mediator of cell behaviour and function
in the tissue, and the ideal scaffold biomaterial for development of a successful end-product
continues to be a matter of debate. Fibrin, a major structural protein involved in wound
healing, represents an ideal scaffold for the rapid synthesis of autologous tissue-engineered
cardiovascular grafts, as its primary scaffold constituents (fibrinogen and thrombin) can be
isolated directly from a blood sample of the patient requiring the graft. Fibrin gel scaffolds
offer immediate high cell seeding efficiency and homogenous cell distribution by gelation
entrapment, and have a degradation rate that can be controlled by protease inhibitors, e.g.
tranexamic acid or aprotinin. Fibrin is also known to stimulate the secretion of reinforcing
extracellular matrix (ECM) proteins by seeded cells. The potential to control the fibrin
polymerisation process also offers the opportunity to produce complex 3-D structures, like
heart valve prostheses and to embed porous, textile or metal (stent) structures. This book
chapter reviews the properties of fibrin that make it an ideal scaffold candidate for
applications in the area of cardiovascular tissue engineering, and documents the successful
development of fibrin-based heart valves, vascular grafts and biostents for clinical application.
2. Scaffold materials
Scaffolds play a central role in cardiovascular tissue engineering. Essential requirements for
the ideal cardiovascular scaffold are easy handling properties and the ability to mould
Tissue Engineering for Tissue and Organ Regeneration
36
complex 3-D structures from the material, such as aortic roots or vessels with complex side
branches. The scaffold material should neither be toxic, nor elicit any immunological side
effects. The diffusion barrier of the scaffold material should have the lowest possible
resistivity in order to guarantee an optimal nutrition supply in thicker tissues. Furthermore,
both the mechanical and the chemical properties (e.g. the integration of growth factors) of
the scaffold material should be modifiable. Controllable degradation of the material is also
important in order to adapt the structural support of the scaffold with regard to the
developing tissue.
A multitude of scaffolds are currently employed in the field of tissue engineering, e.g.
synthetic polymers (polyurethanes, polyglyolic acid, polylactic acid, polyhydroxybutyrate,
copolymers of lactic and glycolic acids, polyanhydrides, polyorthoesters) and natural
polymers (chitosan, glycosaminoglycans, collagen), or biological scaffolds such as
acellularised porcine aortic conduits (Bader et al., 1998; Chevallay & Herbage, 2000;
Flanagan et al., 2006; Freed et al., 1994; Grande et al., 1997). Scaffold-related problems
including cytotoxic degradation products, fixed degradation times, limited mechanical
properties and the absence of growth modulation, etc. necessitate further extensive
investigations in developing the ideal cardiovascular scaffold.
3. Fibrin as scaffold material?
Based on the assumption that successful tissue engineering should mimic the process of
tissue regeneration, and that regeneration is closely related to haemostatis, fibrin (gel) seems
to be an ideal candidate as a tissue engineering scaffold by virtue of its role as a
“physiological scaffold” in tissue regeneration. Several influences of fibrin gel on tissue
development have been described in the literature: it is known that fibrin gel is one of the
major ligands for ß3 integrins, which leads to cell migration into a wound/tissue-engineered
construct (Ikari et al., 2000; Nomura et al., 1999). Thrombin, fibrinogen, fibrin monomers
and fibrinopepide B all increase DNA synthesis in smooth muscle cells (SMCs) and
consequently the proliferation of the cell (Pakala et al., 2001).
3.1 Physiology of fibrin
Fibrin is the end-product of the coagulation cascade following the conversion of fibrinogen
in the presence of thrombin and calcium (Figure 1). Fibrinogen is a soluble plasma
glycoprotein, which is produced by the liver. Fibrinogen is an acute phase protein with a
normal blood concentration of 1.4 – 3.5 g/l. The fibrinogen molecule has a length of 45 nm, a
molecular weight of 340 KDa and consists of 2 subunits and 3 polypeptides chains - α, β and
γ. During the polymerisation process, the fibrinopeptide A of the α-chain and the
fibrinopeptide B of the ß-chain are cleaved by thrombin. The exposed N-terminal positions
of the fibrinopeptides bind to the γ-chain of the fibrinogen and produce the so-called
proteofibrils. In the subsequent step, the lateral association leads to apposition of the
proteofibrils to form a 3-D fibrin network structure (Meyer, 2004). FXIIIa stabilises fibrin
further by incorporation of the fibrinolysis receptors, alpha-2-antiplasmin and TAFI
(thrombin activatable fibrinolysis inhibitor, procarboxypeptidase B), and binding to several
adhesive proteins of various cells (Muszbek et al., 2008).
The polymerised fibrin gel matrix is a hydro-gel, which contains ~95-98% water. The water
content can easily be exchanged against a buffer solution or a cell culture medium, allowing
an optimal nutrition supply of the cells that are embedded after the gelation process.
Cardiovascular Tissue Engineering Based on Fibrin-Gel-Scaffolds 37
Fig. 1. Coagulation cascade: the conversion of fibrinogen into fibrin is triggered by thrombin
and calcium
3.2 Production of autologous fibrin
The classical approach for production of autologous fibrin is the cryoprecipitation method:
after the production of platelet-poor plasma (PPP), the plasma is frozen at -80°C and thawed
overnight at +4°C. The precipitate formed contains ~60-70% of fibrinogen. After
centrifugation, the supernatant is decanted and the precipitate is subsequently washed twice
in rinsed water. After the precipitate is dissolved in water, overnight dialysis against
calcium-free TRIS buffer solution is necessary to provide optimal conditions for the
embedded cells.
The cryoprecipitation method has two major disadvantages: (1) the efficiency of fibrinogen
isolation is relatively low with only ~20-25% of the total fibrinogen content removed, and (2)
the production process is time-consuming (~2 days). The low isolation efficiency is
particularly problematic regarding the use of autologous fibrin gel scaffolds in paediatric
Tissue Engineering for Tissue and Organ Regeneration
38
patients, as the volume of sampled blood needs to be kept to an absolute minimum.
Therefore, the use of alternative precipitation methods with different chemicals has been
evaluated: (1) ethanol (Kjaergard et al., 1992; Weis-Fogh, 1988), (2) ammonium sulphate
alone, and (3) in combination with the cryoprecipitation method (Wolf, 1983), (4) albumin
plus cryoprecipitation, and (5) polyethylene glycol (PEG) plus cryoprecipitation (Epstein et
al., 1986). Heselhaus investigated each of these different precipitation methods with regard
to their efficiency and their use in the development of fibrin scaffold materials for
cardiovascular applications (Heselhaus, 2011):
Fig. 2. [A] Fibrin gel after polymerisation in a 6-well plate. [B-D] Scanning electron
microscopy (SEM) images demonstrating the nano-fibre network structure of the fibrin,
which enables the gentle embedding of cells, with a vascular smooth muscle cell (SMC)
shown in [D] immediately after the gelation of the fibrin within a web-like network
surrounded by cell culture medium
Figure 3 demonstrates that all of the reported alternative methods are more efficient than
the standard cryoprecipitation method. Here, the technique using ethanol as the
precipitation reagent is observed as the most efficient method, with an isolation efficiency of
~80%, ~4-times higher than the efficiency of the standard method. The technique applying
both albumin and cryoprecipitation indicates a false positive high result due to
contamination of the precipitate with albumin (shown by a significant band in SDS gel
electrophoreses) (Heselhaus, 2011).
Another Random Scribd Document
with Unrelated Content
TARTALOM.
VII. Az a másik 3
VIII. Boszu, a maga módja szerint 56
IX. A két fogoly 76
X. Különböző fogadtatások 131
XI. Az a bizonyos szegény asszony 156
Javítások.
Az eredeti szöveg helyesírásán nem változtattunk.
A nyomdai hibákat javítottuk. Ezek listája:
14 az kérdezné azt kérdezné
34 Maxenphutsch urat Maxenpfutsch urat
37 a bagoly! a bagoly!“
42 „Megitati“ csárdában „Megitató“ csárdában
53 hol gyermekrée hol gyermekére
60 gyanuja a Garamvölgyi gyanuja a Garanvölgyi
65 knyszeritnének, hogy kényszeritnének, hogy
74 hogy kit? hogy kit?“
79 Ezt azt utolsó Ezt az utolsó
80 nagy htrtelen nagy hirtelen
83 Hah! Az égre Ne Hah! Az égre! Ne
110 ön elfo-fogadta ön elfogadta
124 kocsis perse kocsis persze
125 megitató csárdánál Megitató csárdánál
125 szaladt egyesen szaladt egyenesen
125 kisszony meg nem kisasszony meg nem
126 Hermin a fedett Eliz a fedett
127 tudott dükösködni tudott dühösködni
129 ne történjék! ne történjék!“
131 IX. Különböző fogadtatások X. Különböző fogadtatások
131 lehessen vvnni lehessen venni
133 egyib jobbról egyik jobbról
136 Bizonyosan tévedész Bizonyosan tévedés
141 a ha-hazajövetelét a hazajövetelét
146 Önt azt mondta Ön azt mondta
148 jövök jöbbet jövök többet
156 olyan azszonyra olyan asszonyra
159 ki iudott ki tudott
167 már többet. már többet.“
170 arrál álmodik arról álmodik
171 megasszony fog lenni menyasszony fog lenni
173 meg nem gyulnék meg nem gyógyulnék
*** END OF THE PROJECT GUTENBERG EBOOK AZ UJ FÖLDESUR
(2. KÖTET) ***
Updated editions will replace the previous one—the old editions will
be renamed.
Creating the works from print editions not protected by U.S.
copyright law means that no one owns a United States copyright in
these works, so the Foundation (and you!) can copy and distribute it
in the United States without permission and without paying
copyright royalties. Special rules, set forth in the General Terms of
Use part of this license, apply to copying and distributing Project
Gutenberg™ electronic works to protect the PROJECT GUTENBERG™
concept and trademark. Project Gutenberg is a registered trademark,
and may not be used if you charge for an eBook, except by following
the terms of the trademark license, including paying royalties for use
of the Project Gutenberg trademark. If you do not charge anything
for copies of this eBook, complying with the trademark license is
very easy. You may use this eBook for nearly any purpose such as
creation of derivative works, reports, performances and research.
Project Gutenberg eBooks may be modified and printed and given
away—you may do practically ANYTHING in the United States with
eBooks not protected by U.S. copyright law. Redistribution is subject
to the trademark license, especially commercial redistribution.
START: FULL LICENSE
THE FULL PROJECT GUTENBERG LICENSE
PLEASE READ THIS BEFORE YOU DISTRIBUTE OR USE THIS WORK
To protect the Project Gutenberg™ mission of promoting the free
distribution of electronic works, by using or distributing this work (or
any other work associated in any way with the phrase “Project
Gutenberg”), you agree to comply with all the terms of the Full
Project Gutenberg™ License available with this file or online at
www.gutenberg.org/license.
Section 1. General Terms of Use and
Redistributing Project Gutenberg™
electronic works
1.A. By reading or using any part of this Project Gutenberg™
electronic work, you indicate that you have read, understand, agree
to and accept all the terms of this license and intellectual property
(trademark/copyright) agreement. If you do not agree to abide by all
the terms of this agreement, you must cease using and return or
destroy all copies of Project Gutenberg™ electronic works in your
possession. If you paid a fee for obtaining a copy of or access to a
Project Gutenberg™ electronic work and you do not agree to be
bound by the terms of this agreement, you may obtain a refund
from the person or entity to whom you paid the fee as set forth in
paragraph 1.E.8.
1.B. “Project Gutenberg” is a registered trademark. It may only be
used on or associated in any way with an electronic work by people
who agree to be bound by the terms of this agreement. There are a
few things that you can do with most Project Gutenberg™ electronic
works even without complying with the full terms of this agreement.
See paragraph 1.C below. There are a lot of things you can do with
Project Gutenberg™ electronic works if you follow the terms of this
agreement and help preserve free future access to Project
Gutenberg™ electronic works. See paragraph 1.E below.
1.C. The Project Gutenberg Literary Archive Foundation (“the
Foundation” or PGLAF), owns a compilation copyright in the
collection of Project Gutenberg™ electronic works. Nearly all the
individual works in the collection are in the public domain in the
United States. If an individual work is unprotected by copyright law
in the United States and you are located in the United States, we do
not claim a right to prevent you from copying, distributing,
performing, displaying or creating derivative works based on the
work as long as all references to Project Gutenberg are removed. Of
course, we hope that you will support the Project Gutenberg™
mission of promoting free access to electronic works by freely
sharing Project Gutenberg™ works in compliance with the terms of
this agreement for keeping the Project Gutenberg™ name associated
with the work. You can easily comply with the terms of this
agreement by keeping this work in the same format with its attached
full Project Gutenberg™ License when you share it without charge
with others.
1.D. The copyright laws of the place where you are located also
govern what you can do with this work. Copyright laws in most
countries are in a constant state of change. If you are outside the
United States, check the laws of your country in addition to the
terms of this agreement before downloading, copying, displaying,
performing, distributing or creating derivative works based on this
work or any other Project Gutenberg™ work. The Foundation makes
no representations concerning the copyright status of any work in
any country other than the United States.
1.E. Unless you have removed all references to Project Gutenberg:
1.E.1. The following sentence, with active links to, or other
immediate access to, the full Project Gutenberg™ License must
appear prominently whenever any copy of a Project Gutenberg™
work (any work on which the phrase “Project Gutenberg” appears,
or with which the phrase “Project Gutenberg” is associated) is
accessed, displayed, performed, viewed, copied or distributed:
This eBook is for the use of anyone anywhere in the United
States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it away
or re-use it under the terms of the Project Gutenberg License
included with this eBook or online at www.gutenberg.org. If you
are not located in the United States, you will have to check the
laws of the country where you are located before using this
eBook.
1.E.2. If an individual Project Gutenberg™ electronic work is derived
from texts not protected by U.S. copyright law (does not contain a
notice indicating that it is posted with permission of the copyright
holder), the work can be copied and distributed to anyone in the
United States without paying any fees or charges. If you are
redistributing or providing access to a work with the phrase “Project
Gutenberg” associated with or appearing on the work, you must
comply either with the requirements of paragraphs 1.E.1 through
1.E.7 or obtain permission for the use of the work and the Project
Gutenberg™ trademark as set forth in paragraphs 1.E.8 or 1.E.9.
1.E.3. If an individual Project Gutenberg™ electronic work is posted
with the permission of the copyright holder, your use and distribution
must comply with both paragraphs 1.E.1 through 1.E.7 and any
additional terms imposed by the copyright holder. Additional terms
will be linked to the Project Gutenberg™ License for all works posted
with the permission of the copyright holder found at the beginning
of this work.
1.E.4. Do not unlink or detach or remove the full Project
Gutenberg™ License terms from this work, or any files containing a
part of this work or any other work associated with Project
Gutenberg™.
1.E.5. Do not copy, display, perform, distribute or redistribute this
electronic work, or any part of this electronic work, without
prominently displaying the sentence set forth in paragraph 1.E.1
with active links or immediate access to the full terms of the Project
Gutenberg™ License.
1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if you
provide access to or distribute copies of a Project Gutenberg™ work
in a format other than “Plain Vanilla ASCII” or other format used in
the official version posted on the official Project Gutenberg™ website
(www.gutenberg.org), you must, at no additional cost, fee or
expense to the user, provide a copy, a means of exporting a copy, or
a means of obtaining a copy upon request, of the work in its original
“Plain Vanilla ASCII” or other form. Any alternate format must
include the full Project Gutenberg™ License as specified in
paragraph 1.E.1.
1.E.7. Do not charge a fee for access to, viewing, displaying,
performing, copying or distributing any Project Gutenberg™ works
unless you comply with paragraph 1.E.8 or 1.E.9.
1.E.8. You may charge a reasonable fee for copies of or providing
access to or distributing Project Gutenberg™ electronic works
provided that:
• You pay a royalty fee of 20% of the gross profits you derive
from the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information
about donations to the Project Gutenberg Literary Archive
Foundation.”
• You provide a full refund of any money paid by a user who
notifies you in writing (or by e-mail) within 30 days of receipt
that s/he does not agree to the terms of the full Project
Gutenberg™ License. You must require such a user to return or
destroy all copies of the works possessed in a physical medium
and discontinue all use of and all access to other copies of
Project Gutenberg™ works.
• You provide, in accordance with paragraph 1.F.3, a full refund of
any money paid for a work or a replacement copy, if a defect in
the electronic work is discovered and reported to you within 90
days of receipt of the work.
• You comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.
1.E.9. If you wish to charge a fee or distribute a Project Gutenberg™
electronic work or group of works on different terms than are set
forth in this agreement, you must obtain permission in writing from
the Project Gutenberg Literary Archive Foundation, the manager of
the Project Gutenberg™ trademark. Contact the Foundation as set
forth in Section 3 below.
1.F.
1.F.1. Project Gutenberg volunteers and employees expend
considerable effort to identify, do copyright research on, transcribe
and proofread works not protected by U.S. copyright law in creating
the Project Gutenberg™ collection. Despite these efforts, Project
Gutenberg™ electronic works, and the medium on which they may
be stored, may contain “Defects,” such as, but not limited to,
incomplete, inaccurate or corrupt data, transcription errors, a
copyright or other intellectual property infringement, a defective or
damaged disk or other medium, a computer virus, or computer
codes that damage or cannot be read by your equipment.
1.F.2. LIMITED WARRANTY, DISCLAIMER OF DAMAGES - Except for
the “Right of Replacement or Refund” described in paragraph 1.F.3,
the Project Gutenberg Literary Archive Foundation, the owner of the
Project Gutenberg™ trademark, and any other party distributing a
Project Gutenberg™ electronic work under this agreement, disclaim
all liability to you for damages, costs and expenses, including legal
fees. YOU AGREE THAT YOU HAVE NO REMEDIES FOR
NEGLIGENCE, STRICT LIABILITY, BREACH OF WARRANTY OR
BREACH OF CONTRACT EXCEPT THOSE PROVIDED IN PARAGRAPH
1.F.3. YOU AGREE THAT THE FOUNDATION, THE TRADEMARK
OWNER, AND ANY DISTRIBUTOR UNDER THIS AGREEMENT WILL
NOT BE LIABLE TO YOU FOR ACTUAL, DIRECT, INDIRECT,
CONSEQUENTIAL, PUNITIVE OR INCIDENTAL DAMAGES EVEN IF
YOU GIVE NOTICE OF THE POSSIBILITY OF SUCH DAMAGE.
1.F.3. LIMITED RIGHT OF REPLACEMENT OR REFUND - If you
discover a defect in this electronic work within 90 days of receiving
it, you can receive a refund of the money (if any) you paid for it by
sending a written explanation to the person you received the work
from. If you received the work on a physical medium, you must
return the medium with your written explanation. The person or
entity that provided you with the defective work may elect to provide
a replacement copy in lieu of a refund. If you received the work
electronically, the person or entity providing it to you may choose to
give you a second opportunity to receive the work electronically in
lieu of a refund. If the second copy is also defective, you may
demand a refund in writing without further opportunities to fix the
problem.
1.F.4. Except for the limited right of replacement or refund set forth
in paragraph 1.F.3, this work is provided to you ‘AS-IS’, WITH NO
OTHER WARRANTIES OF ANY KIND, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO WARRANTIES OF
MERCHANTABILITY OR FITNESS FOR ANY PURPOSE.
1.F.5. Some states do not allow disclaimers of certain implied
warranties or the exclusion or limitation of certain types of damages.
If any disclaimer or limitation set forth in this agreement violates the
law of the state applicable to this agreement, the agreement shall be
interpreted to make the maximum disclaimer or limitation permitted
by the applicable state law. The invalidity or unenforceability of any
provision of this agreement shall not void the remaining provisions.
1.F.6. INDEMNITY - You agree to indemnify and hold the Foundation,
the trademark owner, any agent or employee of the Foundation,
anyone providing copies of Project Gutenberg™ electronic works in
accordance with this agreement, and any volunteers associated with
the production, promotion and distribution of Project Gutenberg™
electronic works, harmless from all liability, costs and expenses,
including legal fees, that arise directly or indirectly from any of the
following which you do or cause to occur: (a) distribution of this or
any Project Gutenberg™ work, (b) alteration, modification, or
additions or deletions to any Project Gutenberg™ work, and (c) any
Defect you cause.
Section 2. Information about the Mission
of Project Gutenberg™
Project Gutenberg™ is synonymous with the free distribution of
electronic works in formats readable by the widest variety of
computers including obsolete, old, middle-aged and new computers.
It exists because of the efforts of hundreds of volunteers and
donations from people in all walks of life.
Volunteers and financial support to provide volunteers with the
assistance they need are critical to reaching Project Gutenberg™’s
goals and ensuring that the Project Gutenberg™ collection will
remain freely available for generations to come. In 2001, the Project
Gutenberg Literary Archive Foundation was created to provide a
secure and permanent future for Project Gutenberg™ and future
generations. To learn more about the Project Gutenberg Literary
Archive Foundation and how your efforts and donations can help,
see Sections 3 and 4 and the Foundation information page at
www.gutenberg.org.
Section 3. Information about the Project
Gutenberg Literary Archive Foundation
The Project Gutenberg Literary Archive Foundation is a non-profit
501(c)(3) educational corporation organized under the laws of the
state of Mississippi and granted tax exempt status by the Internal
Revenue Service. The Foundation’s EIN or federal tax identification
number is 64-6221541. Contributions to the Project Gutenberg
Literary Archive Foundation are tax deductible to the full extent
permitted by U.S. federal laws and your state’s laws.
The Foundation’s business office is located at 809 North 1500 West,
Salt Lake City, UT 84116, (801) 596-1887. Email contact links and up
to date contact information can be found at the Foundation’s website
and official page at www.gutenberg.org/contact
Section 4. Information about Donations to
the Project Gutenberg Literary Archive
Foundation
Project Gutenberg™ depends upon and cannot survive without
widespread public support and donations to carry out its mission of
increasing the number of public domain and licensed works that can
be freely distributed in machine-readable form accessible by the
widest array of equipment including outdated equipment. Many
small donations ($1 to $5,000) are particularly important to
maintaining tax exempt status with the IRS.
The Foundation is committed to complying with the laws regulating
charities and charitable donations in all 50 states of the United
States. Compliance requirements are not uniform and it takes a
considerable effort, much paperwork and many fees to meet and
keep up with these requirements. We do not solicit donations in
locations where we have not received written confirmation of
compliance. To SEND DONATIONS or determine the status of
compliance for any particular state visit www.gutenberg.org/donate.
While we cannot and do not solicit contributions from states where
we have not met the solicitation requirements, we know of no
prohibition against accepting unsolicited donations from donors in
such states who approach us with offers to donate.
International donations are gratefully accepted, but we cannot make
any statements concerning tax treatment of donations received from
outside the United States. U.S. laws alone swamp our small staff.
Please check the Project Gutenberg web pages for current donation
methods and addresses. Donations are accepted in a number of
other ways including checks, online payments and credit card
donations. To donate, please visit: www.gutenberg.org/donate.
Section 5. General Information About
Project Gutenberg™ electronic works
Professor Michael S. Hart was the originator of the Project
Gutenberg™ concept of a library of electronic works that could be
freely shared with anyone. For forty years, he produced and
distributed Project Gutenberg™ eBooks with only a loose network of
volunteer support.
Project Gutenberg™ eBooks are often created from several printed
editions, all of which are confirmed as not protected by copyright in
the U.S. unless a copyright notice is included. Thus, we do not
necessarily keep eBooks in compliance with any particular paper
edition.
Most people start at our website which has the main PG search
facility: www.gutenberg.org.
This website includes information about Project Gutenberg™,
including how to make donations to the Project Gutenberg Literary
Archive Foundation, how to help produce our new eBooks, and how
to subscribe to our email newsletter to hear about new eBooks.
Welcome to our website – the perfect destination for book lovers and
knowledge seekers. We believe that every book holds a new world,
offering opportunities for learning, discovery, and personal growth.
That’s why we are dedicated to bringing you a diverse collection of
books, ranging from classic literature and specialized publications to
self-development guides and children's books.
More than just a book-buying platform, we strive to be a bridge
connecting you with timeless cultural and intellectual values. With an
elegant, user-friendly interface and a smart search system, you can
quickly find the books that best suit your interests. Additionally,
our special promotions and home delivery services help you save time
and fully enjoy the joy of reading.
Join us on a journey of knowledge exploration, passion nurturing, and
personal growth every day!
ebookbell.com

Tissue Engineering For Tissue And Organ Regeneration D Eberli

  • 1.
    Tissue Engineering ForTissue And Organ Regeneration D Eberli download https://ebookbell.com/product/tissue-engineering-for-tissue-and- organ-regeneration-d-eberli-4115082 Explore and download more ebooks at ebookbell.com
  • 2.
    Here are somerecommended products that we believe you will be interested in. You can click the link to download. Tissue Cell And Organ Engineering Nanotechnologies For The Life Sciences Volume 9 1st Edition Challa S S R Kumar https://ebookbell.com/product/tissue-cell-and-organ-engineering- nanotechnologies-for-the-life-sciences-volume-9-1st-edition-challa-s- s-r-kumar-2176726 Silk Biomaterials For Tissue Engineering And Regenerative Medicine Subhas Kundu https://ebookbell.com/product/silk-biomaterials-for-tissue- engineering-and-regenerative-medicine-subhas-kundu-4660634 Electrospun Materials For Tissue Engineering And Biomedical Applications Research Design And Commercialization 1st Edition Tamer Uyar And Erich Kny Eds https://ebookbell.com/product/electrospun-materials-for-tissue- engineering-and-biomedical-applications-research-design-and- commercialization-1st-edition-tamer-uyar-and-erich-kny-eds-6614086 Electrospun Materials For Tissue Engineering And Biomedical Applications Research Design And Commercialization Tamer Uyar https://ebookbell.com/product/electrospun-materials-for-tissue- engineering-and-biomedical-applications-research-design-and- commercialization-tamer-uyar-6670478
  • 3.
    Extracellular Matrix ForTissue Engineering And Biomaterials 1st Ed Anna C Berardi https://ebookbell.com/product/extracellular-matrix-for-tissue- engineering-and-biomaterials-1st-ed-anna-c-berardi-7149442 Biomaterials For Tissue Engineering And Regeneration Ressler A https://ebookbell.com/product/biomaterials-for-tissue-engineering-and- regeneration-ressler-a-48949304 Biomaterials For Tissue Engineering And Regeneration Antonia Ressler https://ebookbell.com/product/biomaterials-for-tissue-engineering-and- regeneration-antonia-ressler-55247884 Vascularization For Tissue Engineering And Regenerative Medicine 1st Edition Wolfgang Holnthoner Andrea Banfi James Kirkpatrick Heinz Redl https://ebookbell.com/product/vascularization-for-tissue-engineering- and-regenerative-medicine-1st-edition-wolfgang-holnthoner-andrea- banfi-james-kirkpatrick-heinz-redl-32560254 Stem Cell Therapy And Tissue Engineering For Cardiovascular Repair From Basic Research To Clinical Applications 1st Edition Stefan Klotz https://ebookbell.com/product/stem-cell-therapy-and-tissue- engineering-for-cardiovascular-repair-from-basic-research-to-clinical- applications-1st-edition-stefan-klotz-4191650
  • 5.
    TISSUE ENGINEERING FOR TISSUEAND ORGAN REGENERATION Edited by Daniel Eberli
  • 6.
    Tissue Engineering forTissue and Organ Regeneration Edited by Daniel Eberli Published by InTech Janeza Trdine 9, 51000 Rijeka, Croatia Copyright © 2011 InTech All chapters are Open Access articles distributed under the Creative Commons Non Commercial Share Alike Attribution 3.0 license, which permits to copy, distribute, transmit, and adapt the work in any medium, so long as the original work is properly cited. After this work has been published by InTech, authors have the right to republish it, in whole or part, in any publication of which they are the author, and to make other personal use of the work. Any republication, referencing or personal use of the work must explicitly identify the original source. Statements and opinions expressed in the chapters are these of the individual contributors and not necessarily those of the editors or publisher. No responsibility is accepted for the accuracy of information contained in the published articles. The publisher assumes no responsibility for any damage or injury to persons or property arising out of the use of any materials, instructions, methods or ideas contained in the book. Publishing Process Manager Romina Krebel Technical Editor Teodora Smiljanic Cover Designer Jan Hyrat Image Copyright Arun K. Sharma, 2011. First published August, 2011 Printed in Croatia A free online edition of this book is available at www.intechopen.com Additional hard copies can be obtained from [email protected] Tissue Engineering for Tissue and Organ Regeneration, Edited by Daniel Eberli p. cm. ISBN 978-953-307-688-1
  • 9.
    Contents Preface IX Part 1Cardiac Muscle 1 Chapter 1 Myocardial Tissue Engineering 3 Tatsuya Shimizu Chapter 2 Cardiac Muscle Engineering: Strategies to Deliver Stem Cells to the Damaged Site 19 Giancarlo Forte, Stefania Pagliari, Francesca Pagliari, Paolo Di Nardo and Takao Aoyagi Chapter 3 Cardiovascular Tissue Engineering Based on Fibrin-Gel-Scaffolds 35 Stefan Jockenhoevel and Thomas C. Flanagan Chapter 4 Rapid Prototyping of Engineered Heart Tissues through Miniaturization and Phenotype-Automation 49 Tetsuro Wakatsuki Part 2 Skeletal Muscle 59 Chapter 5 Tissue Engineering of Skeletal Muscle 61 Klumpp Dorothee, Horch Raymund E. and Beier Justus P. Chapter 6 Skeletal Muscle Tissue Engineering Using Biological Scaffolds for Repair of Abdominal Wall Defects in a Rabbit Model 81 Zuki Abu Bakar, Ayele Taddese Tsedeke, Noorjahan Banu Mohamed Alitheen and Noordin Mohamed Mustapha Chapter 7 Skeletal and Adipose Tissue Engineering with Adipose-Derived Stromal Cells 107 Jeong S Hyun, Emily R Nelson, Daniel Montoro, Benjamin Levi and Michael T. Longaker
  • 10.
    VI Contents Part 3Ligaments 129 Chapter 8 Tissue Engineering of Ligaments 131 Sarah Rathbone and Sarah Cartmell Chapter 9 Potential of Tissue-Engineered Ligament Substitutes for Ruptured ACL Replacement 163 Goulet F., Chabaud S., Simon F., Napa I.D., Moulin V. and Hart D.A. Part 4 Cartilage 179 Chapter 10 Joint Cartilage Tissue Engineering and Pre-Clinical Safety and Efficacy Testing 181 Thomas G. Koch, Lorenzo Moroni, Younes Leysi-Derilou and Lise C. Berg Chapter 11 Cartilage Regeneration from Bone Marrow Cells Using RWV Bioreactor and Its Automation System for Clinical Application 217 Toshimasa Uemura, Masanori Nishi, Kunitomo Aoki and Takashi Tsumura Chapter 12 Cartilage Tissue Engineering: the Application of Nanomaterials and Stem Cell Technology 233 Adelola O. Oseni, Claire Crowley, Maria Z. Boland, Peter E. Butler and Alexander M. Seifalian Part 5 Hollow Organs 267 Chapter 13 Bioengineering of Colo-Rectal Tissue 269 Roman Inglin, Lukas Brügger, Daniel Candinas and Daniel Eberli Chapter 14 Aspects of Urological Tissue Engineering 285 Arun K. Sharma and Dorota I. Rozkiewicz Part 6 Craniofacial Tissues 315 Chapter 15 Tooth Organ Engineering: Biological Constraints Specifying Experimental Approaches 317 Sabine Kuchler-Bopp, Laetitia Keller, Anne Poliard and Herve Lesot Chapter 16 Transplantation of Corneal Stroma Reconstructed with Gelatin and Multipotent Precursor Cells from Corneal Stroma 347 Tatsuya Mimura, Yasuhiko Tabata and Shiro Amano
  • 11.
    Contents VII Chapter 17Human Ear Cartilage 363 Lu Zhang, Qiong Li, Yu Liu, Guangdong Zhou, Wei Liu and Yilin Cao Part 7 Central Nervous System 377 Chapter 18 Advances in the Combined Use of Adult Cell Therapy and Scaffolds for Brain Tissue Engineering 379 Elisa Garbayo, Gaëtan J.-R. Delcroix, Paul C. Schiller and Claudia N. Montero-Menei Part 8 Endocrine Organs 415 Chapter 19 Regenerative Medicine and Tissue Engineering for the Treatment of Diabetes 417 Matsumoto S, SoRelle JA and Shimoda M Chapter 20 Perspectives of Islet Cell Transplantation as a Therapeutic Approach for Diabetes Mellitus 435 Prabha D. Nair and Neena Aloysious
  • 13.
    Preface Over the lastdecade Tissue Engineering progressed rapidly and first biological substitutes were developed for several tissues in the body. Today, Tissue Engineering is one of the major approaches of Regenerative Medicine and represents a growing and exciting field of research. With the understanding and application of new knowledge of structure, biology, physiology and cell culture techniques, Tissue Engineering may offer new treatment alternatives for organ replacement or repair deteriorated organs. Among the clinical applications of Tissue Engineering are the production of artificial skin for burn patients, tissue engineered trachea, cartilage for knee-replacement procedures, urinary bladder replacement, urethra substitutes and cellular therapies for the treatment of urinary incontinence. The classical principle of Tissue Engineering is to dissociate cells from a tissue biopsy, to expand them in culture, and to seed them onto a scaffold material in vitro in order to generate a viable tissue construct prior to re-implantation into the recipient's organism. In the appropriate biochemical and biomechanical environment these tissues will unfold their full functional potential and serve as native tissue equivalents. Tissue Engineering products may be fully functional at the time of treatment, or have potential to integrate and evolve into the expected functional tissue after implantation. While these steps may seem logical and easy to understand, the underlying biology is far more complicated and more profound questions have to be answered before the engineering of tissue and organs becomes a routine practice. Even so, the Tissue Engineering approach has major advantages over traditional organ transplantation and circumvents the problem of organ shortage. Tissues reconstructed from readily available biopsy material implicate only minimal or no immunogenicity when reimplanted in the patient. This eventually conquers several limitations encountered in tissue transplantation approaches. This book is aimed at anyone interested in the application of Tissue Engineering in different organ systems. With a colorful mix of topics which explain the obstacles and possible solutions, it offers insights into a wide variety of strategies applying the principles of Tissue Engineering to tissue and organ regeneration. As more and more applications move toward clinical application, a reliable preclinical model system to
  • 14.
    X Preface evaluate thedeveloped techniques becomes crucial. Several animal models and Tissue Engineering approaches for a variety of organ systems are presented in this book. Finally, I would like to thank all the authors who have supported this book with their contributions. Daniel Eberli University Zürich Switzerland
  • 17.
  • 19.
    1 Myocardial Tissue Engineering TatsuyaShimizu Institute of Advanced Biomedical Engineering & Science Tokyo Women’s Medical University Japan 1. Introduction Many lives are lost due to heart diseases including myocardial infarction and cardiomyopathy. Recent reports have demonstrated that regenerative medicine has promising potential for recovering severe heart failure. Regenerative therapies for heart failure include cytokine, gene and cell therapy. Because many types of cardiovascular stem cells have been identified and their clinical potentials have been demonstrated for the past decade, cell injection therapy has most attracted both researchers and clinicians (Wollert 2008). On the other hand, significant cell loss due to washing out and cell death has become problematic in cell injection technique. So, as next generation of regenerative therapy for impaired heart, transplantation of myocardial patches fabricated by tissue engineering technology are emerging and are clinically applied. Furthermore, several challenges for fabricating functional myocardial tissues/organs, which are electrically communicated, pulsate synchronously and evoke contraction power, have also started (Zimmermann, Didie et al. 2006). These ambitious challenges may lead to reconstruction of malformed hearts and become alternative therapy for heart transplantation. Heart tissues are composed of high-dense cylindrical cardiomyocytes and fibroblasts with abundant vascular network and collagen-based extracellular matrix (ECM). Cardiomyocytes pulsate via sodium and calcium ion transient through cell membrane. They are also electrically coupled by gap junctions composed of connexion 43 and rapid electrical propagation realizes simultaneous beating as a whole. Continuous blood flow supplies oxygen and nutrition, and withdraw the waste for high metabolic demand of heart tissues. These structure and function produce mechanical contractions as a blood pump. Therefore the researchers should take into account high density culture of cardiomyocyte and surrounding cells, sufficient micro blood vessel fabrication, cell/ECM orientation and proper cell-to-cell coupling for engineering heart tissues/organs. Here, previous and current status of cell injection therapy, myocardial patch transplantation and pulsatile myocardial tissue fabrication is described with some future views. 2. Cell injection therapy Cell injection therapy for damaged heart has been researched since the early 1990’s. Many researchers have demonstrated the therapeutic potential of isolated cell transplantation into myocardium using various types of cell sources both in animal models and in some clinical
  • 20.
    Tissue Engineering forTissue and Organ Regeneration 4 trials (Puceat 2008). The mechanism of myocardial tissue regeneration has not been completely cleared, but most researchers have agreed that transplanted cells secrete several cytokines which promote neovascularization, prohibit fibrosis, decrease cell death and recruit stem cells, leading to heart function improvement. It has been also asserted that some of injected cells differentiate into functional cardiomyocytes and may directly contribute to heart contraction improvement. Although some differences may exist in according to cell types, multifactorial mechanisms seem to relate with myocardial tissue regeneration. In addition to cell sourcing, different routes are used for cell administration. Systematic intravenous infusion is performed through a central or peripheral vein. This method is simple and less invasive, however widespread distribution cause low ratio of cell engraftment. Most popular approach is intracoronary cell infusion via a balloon-catheter. Injected cells are reached directly in the target myocardial region, however, cells have to transmigrate across endothelium wall. Intracardiac injection is performed via pericardium during open heart surgery and via endocardium by a catheter with a 3-D electromechanical mapping system (NOGA mapping system). These methods realize relatively targeted delivery, but myocardial damage and arrhythmia induction are problematic. Future clarification will be needed to decide which is the best approach for cell injection. 2.1 Skeletal myoblasts Skeletal myoblasts were the first cell source to enter the clinical application for heart tissue repair. They lie in a quiescent state on the basal membrane of myofibers and have the potential to start to proliferate and differentiate into functional skeletal muscle in response to muscle damage. They can be isolated autologously and be expanded from a single biopsy. In addition, skeketal myoblasts are relatively resistant to ischemia. Menasche and colleagues first applied skeletal myoblast injection via epicardium for patients undergoing coronary artery bypass grafting (CABG) (Menasche, Hagege et al. 2001). The phase I clinical study (MAGIC I) have shown the feasibility of skeletal myoblast implantation, however, increased risk of ventricular arrhythmias after the operation. Then, MAGIC II trial was performed to clarify the safety and efficacy, in which all patients received preventive medication and an implantable cardioverter-defibrillator for rescuing critical ventricular arrhythmias. In result, skeletal myoblast injection failed to significantly improve heart function, leading to sample size reduction (Menasche, Alfieri et al. 2008). On the other hand, the trial indicated the possibility that high dose cell injection might recover left ventricular dilatation. In addition, the other clinical trials of catheter-based myoblast implantation via endocardium have revealed functional efficacy (Opie and Dib 2006). According to these results, not the regenerative potential of myoblasts themselves but the amount of injected cells and delivery system may affect the efficacy. Therefore, it seems that skeletal myoblasts should not be excluded as a cell source for heart tissue repair. More optimization of cell delivery and comparison of cell sources can address these critical issues. 2.2 Bone marrow-derived cells Bone marrow-derived cells are the most used cells in clinical trials for myocardial tissue repair (Wollert 2008). The discovery of circulating progenitor cells originated from human bone marrow has stimulated research and clinical use of bone marrow-derived cells
  • 21.
    Myocardial Tissue Engineering5 (Asahara, Murohara et al. 1997). Bone marrow cells contain different stem and progenitor cells which will differentiate into various types of cells including endothelial cells, smooth muscle cells and cardiomyocytes. Bone marrow mononuclear cells (BMNCs), which can be isolated simply by gradient sedimentation after bone marrow aspiration without culture expansion, have been clinically injected via coronary artery from the first. BMNCs include heterogeneous cell population of monocytes, hematopoietic stem cells and endothelial progenitor cells (EPCs). Therefore, some groups have used BMNCs selected by surface markers (CD34+, CD133+) and demonstrated more efficacy of their injection. As another cell population, mesenchymal stem cells (MSCs) have been researched and clinically used. Although, MSCs represent between 0.01 and 0.001% of all nucleated cells in bone marrow, they can be readily expanded in culture. MSCs have the potential to differentiate into various types of cells and injected MSCs in heart seem to differentiate into myocardial composing cells. Recent studies have revealed rare happening of cardiomyocyte differentiation, therefore MSCs seem to recover heart function via their cytokine secretion and partial differentiation into vascular cells. As a unique feature, MSCs have the potential to escape from immune detection due to the direct inflammatory inhibition and the lack of cell-surface molecules. This property has realized allogenic mesenchymal stem cell transplantation in clinic and has given high impact on cell therapy research field. Recent randomized controlled trials of bone marrow-derived cell injection revealed overall feasibility and safety. However the data has revealed only marginal increases of ejection fraction (EF) even in positive studies (0-5%) (Martin-Rendon, Brunskill et al. 2008). For establishing more effective bone marrow-derived cell therapy, optimization of cell source, cell dose, delivery method and deliver timing will be needed. 2.3 Adipose-derived stem cells In addition to bone marrow-derived MSCs, stem cells isolated from the stroma of adipose tissues have represented regenerative potential for heart tissues (Psaltis, Zannettino et al. 2008). Adipose tissue-derived stem cells (ASCs) display features similar to that of bone marrow-derived MSCs and their angiogenic potential have been reported. Some studies have also revealed cardiomyocyte differentiation from ASCs. It has not been clarified which mesenchymal stem cells are superior to other cell types, however, reatively easy isolation of adipose tissue may push the clinical application of ASCs. 2.4 Cardiac stem cells Cardiac stem cells (CSCs) are also possible cell source for myocardial tissue regeneration. Two groups first reported CSC existence in 2003 (Beltrami, Barlucchi et al. 2003; Oh, Bradfute et al. 2003). Until then, it was common knowledge that heart was a post mitotic organ, but those reports accelerated the researches for identifying surface marker of CSCs and culturing them. Islet-1, Sca-1 and c-kit have been known as CSC markers. Recently, it has been also confirmed that heart has renewal ability at normal state and the annual rate of turning over is 1% at the age of 25 (Bergmann, Bhardwaj et al. 2009). Although the ability of CSCs may increase after heart injury, newly formed cardyomyocytes are not sufficient for replacing damaged muscle tissues. Therefore isolation and expansion of CSCs have been extensively examined. Some groups have used a different approach to make cardiospheres from biopsied myocardium, which lead to efficient CSC expansion (Lee, White et al. 2011).
  • 22.
    Tissue Engineering forTissue and Organ Regeneration 6 Clinical trials for injection therapy of autologous CSCs isolated from biopsy sample are now on going. 2.5 Embryonic stem cells Although abundant studies demonstrated that MSCs, ASCs and CSCs have the potential of cardiomyocyte differentiation regarding gene and protein expression, there are no studies clearly showing beating cardiomyocytes differentiated from those stem cells. On the other hand, many researchers have confirmed that embryonic stem cells (ESCs) can differentiate into beating cardiomyocytes in vitro and implantation of ESC-derived cardiomyocytes improves damaged heart function. Several signal pathways for cardiac differentiation have been already clarified and various molecules have been reported as its promoters. For example, noggin increased cardiac differentiation efficacy via regulation of Bone morphogenetic protein (BMP) signalling pathway (Yuasa, Itabashi et al. 2005) and insulin- like growth-factor-binding protein 4 (IGFBP4) promotes cardiogenesis by inhibitor of canonical Wnt signalling (Zhu, Shiojima et al. 2008). In addition, fibloblast growth factor (FGF), retinoic acid, ascorbic acid and cyclosporine A have been reported to have the potential to enhance cardiac differentiation from ESCs. The important issue as well as cardiac differentiation is purification of cardiomyocytes from heterogeneous cell mixture, because contamination of immature cells leads to teratoma formation. Although gene- modified ESCs harboring neomycin resistance gene or green fluorescent protein (GFP) gene in the cardiac-specific gene locus are very useful in non-clinical experiments, safe and efficient isolation technologies will be needed for clinical application. Culture media control focusing on the differences of cell metabolism may be useful for safe cell selection. Moreover immune response of the host is another critical issue. Nucler transfer or cell banking is possible approach avoiding immunoreaction. Electrical communication and simultaneous beating of implanted ESC-derived cardiomyocytes should be also requested for improving damaged heart function without arrhythmia. In vivo electrophisiological analyses and the transplantation technology for synchronization will be essential for clinical application of these cells. 2.6 Induced pluripotent stem cells Induced pluripotent stem cells (iPSCs) also hold great promise for myocardial tissue engineering (Vunjak-Novakovic, Tandon et al. 2010). Terminally differentiated cells can be reprogrammed to have the same potential as ESCs by introducing 3 or 4 transcriptional factor genes. Furthemore none-gene transfer technologies have been developed in the world. The superiority of iPSCs to ESCs is autologous cells, which do not cause immune response. Cardiac differentiation of human iPSCs has been reported in the same manner with ESCs. Several critical issues must be clarified for clinical use, but ESCs/iPSCs-derived cardiomyocytes should contribute to myocardial tissue engineering in the view point of their pulsatile function and scaling-up. 2.7 Problems of cell injection therapy Cell injection therapies for heart failure are now world-widely performed. While moderate success of direct cell injection has been observed, the efficacies seem not to reach the level that general clinicians think cell therapy a reliable treatment for heart failure. More
  • 23.
    Myocardial Tissue Engineering7 optimization of cell source, cell preparation process, injection route, injection timing and patient population may increase the effectiveness; however one of the essential issues is cell delivery methodology. Cell injection therapy has significant difficulties about cell retention in the target tissue. The shape, size, and position of the grafted cells are often uncontrollable and large amount of the cells are washed-out. Moreover, once retaining cells die due to necrosis and apoptosis. Time course quantification with TUNEL assay demonstrated that a large number of the grafted cells die within a few days after injection in rat models (Zhang, Methot et al. 2001). In the clinical trial using bone marrow-derived cells, it has been also demonstrated that only 1-3% of the cells infused via coronary arteries could be detected by 3D positron emission tomography (PET) imaging of the patient heart. In this study, a large percentage of cells were found in the liver and spleen immediately after the procedures (Hofmann, Wollert et al. 2005). To clear the problem of cell loss, hydrogel-cell mixture injection has been pursued. Fibrin, collagen and alginate hydrogels are now used. Hydrogels with cells are injected as a liquid phase through syringe or catheter, then, they are polymerized and fixed in the target tissues (Kofidis, de Bruin et al. 2004). In hydrogel-cell mixture injection therapy, local tissue damage due to space occupation of hydrogel itself and inflammatory reaction due to hydrogel biodegradation are problematic. Therefore, more advanced cell delivery systems have been requested to spread the regenerative therapy as one of the reliable treatments for heart failure. 3. Tissue engineering Recent advance of tissue engineering technologies have realized the transplantation of tissue-engineered construct “myocardial patch” covering over damaged heart surface instead of simple cell injection into myocardium. Grafted cells within myocardial patches can survive more and secrete more cytokines, resulting in more heart function improvement. Furthermore pulsatile myocardial tissues have been successfully engineered by using cardiomyocytes as a seeding cell source. These tissues may directly help heart contraction and total heart wall replacement may be possible in future. There are several contexts of tissue engineering. 3.1 Scaffold-based tissue engineering Most popular technology of tissue engineering is to seed cells into 3-D pre-fabricated biodegradable scaffolds which are made from synthetic polymer and biological material. Hydrogel formation after mixing cells and scaffold solution is another approach. Decellularized tissues have been also used as scaffolds. These scaffolds play as alternatives for extra cellular matrix (ECM), therefore, their cell-adhesiveness and porosity affect survived cell amount and engineered tissue quality. Scaffold modification can control its biodegradation and tissue formation. Growth factor linkage leads to accelerating tissue formation. Now these scaffold-based tissue engineering has been widely applied to cardiovascular tissue regeneration as well as other tissue repair (Vunjak-Novakovic, Tandon et al. 2010). 3.2 Cell sheet-based tissue engineering In contrast to scaffold-based tissue engineering, our group have developed unique technique involving cell sheet stacking to fabricate 3-D tissues (Shimizu, Yamato et al. 2003).
  • 24.
    Tissue Engineering forTissue and Organ Regeneration 8 Cell sheets are 2-D connecting pure cells without any scaffolds, therefore cell-dense 3-D tissues can be fabricated by stacking cell sheets. Cell sheets are harvested from intelligent culture surface “temperature-responsive culture surface”, which are covalently grafted with temperature-responsive polymer, poly (N-isopropylacrylamide) (PIPAAm) (Okano, Yamada et al. 1993). The surfaces are slightly hydrophobic and cell-adhesive at 37˚C, on the other hand, the surface changes to hydrophilic and not cell adhesive below 32˚C. Confluently cultured cells on the surface can detach as a contiguous cell sheet simply by reducing temperature. Furthermore, biological molecules underneath cell sheets are also preserved and play a critical role as an adhesive agent during cell sheet stacking. Cell sheet-based tissue engineering has been applied for a wide range of regenerative medicine including corneal epithelial replacement, heart tissue repair, pneumothorax repair, liver tissue repair and so on. According to the spread of the concept fabricating 3-D tissues from 2-D confluent cells, several other technologies using this concept have emerged. Cell sheet fabrication techniques using fibrin coated dishes or nanofibrous polycaprolactone meshes have been reported (Shin, Ishii et al. 2004; Itabashi, Miyoshi et al. 2005). Cell sheet-like constructs have been also engineered using magnetite nanoparticles (Ito, Hibino et al. 2005). Magnetically labelled cells are attached on culture materials by magnetic force and confluent cells are harvested as a cell sheet by magnetic force release. Thus, cell sheet-based tissue engineering has now spread in the world as scaffold-free tissue engineering. 4. Myocardial patch transplantation Both scaffold-based and cell sheet-based tissue engineering have been used for myocardial patch fabrication. Not only cardiomyocytes but also other types of cells have been used for creating myocardial patches and some myocardial patches using non-cardiomyocytes have been already clinically transplanted over damaged hearts. (Fig. 1.) 4.1 Scaffold-based mayocardial patch In myocardial patch fabrication, synthetic polymer, biological material and decellularized tissue have been used as prefabricated scaffolds. Li and colleagues, who were one of the pioneer groups of myocardial tissue engineering, first demonstrated that gelatine sponges seeded with cardiac cells have therapeutic potentials for cryoinjured rat hearts (Li, Jia et al. 1999). Leor and colleagues reported that bioengineered heart grafts using porous alginate scaffolds attenuated left ventricular dilatation and heart function deterioration in infarction model (Leor, Aboulafia-Etzion et al. 2000). Eschenhagen and Zimmermann’s group have developed innovative myocardial tissue engineering approach (Zimmermann, Schneiderbanger et al. 2002). They have fabricated 3-D tissues by gelling mixture of cardiac cells and collagen solution. The constructs induced systolic wall thickening of the left ventricle infracted area and improved fractional shortening of damaged hearts in rat myocardial infarction model (Zimmermann, Melnychenko et al. 2006). Small intestinal submucosa (SIS) has also been used as a scaffold for myocardial patch. MSC-seeded SIS improved heart contraction in rabbit infarction model (Tan, Zhi et al. 2009). There have been various types of myocardial patches using different scaffolds and different cell sources. Although implantable human myocardial patches using beating cardiomyocytes have not been established now, clinical trials of collagen-based myocardial patch with bone marrow
  • 25.
    Myocardial Tissue Engineering9 cells (MAGNUM trial) (Chachques, Trainini et al. 2007) and vicryl mesh-based myocardial patches with fibroblasts (Anginera) ((Mirsadraee, Wilcox et al. 2006)) have revealed feasibility and safety of myocardial patch transplantation. Diseased heart Cells Stacking 3. Cell sheet-based TE Temperature-responsive culture surface Cells & ECM mixture Gelling Scaffold 1. Scaffold-based TE 2. Hydrogel-based TE Cell sheet Seeding Fig. 1. Tissue engineering (TE) strategies for myocardial patch fabrication 4.2 Cell sheet-based myocardial patch Many types of cell sheets have been reported to improve impaired heart function (Shimizu, Sekine et al. 2009). Cell sheets are transplanted onto heart surface directly via open heart surgery and cells can be more effectively delivered as thin, but large-area cell-dense grafts than isolated cell injection. Scaffold-based myocardial patches are usually transplanted on myocardium with suture, on the other hand, cell sheets are transplanted with no suture because biological adhesive proteins underneath cell sheets promote the attachment. When neonatal rat cardiac cell sheets were transplanted onto infracted rat hearts, grafted cardiomyocytes communicated with host myocardium via gap junctions and blood vessels formed within the graft, resulting in significant improvement of heart function (Miyagawa, Sawa et al. 2005; Sekine, Shimizu et al. 2006). Sawa and colleagues have started to use skeletal myoblasts for cell sheet fabrication, because myoblasts can be isolated autologously and are relatively resistant to ischemic condition.
  • 26.
    Tissue Engineering forTissue and Organ Regeneration 10 The recovery of heart function by skeletal myoblast transplantation has been confirmed in rat ischemic model, in dilated cardiomyopathy hamster model, in pacing-induced canine heart failure model and in pig infarction model (Memon, Sawa et al. 2005; Hata, Matsumiya et al. 2006; Kondoh, Sawa et al. 2006; Miyagawa, Saito et al. 2010). Regarding stacking cell sheet number, 3-5 layers are optimal and more layering cause primary necrosis of the constructs (Sekiya, Matsumiya et al. 2009). They have demonstrated more hematopoietic stem cells and less fibrosis in cell sheet transplantation than in isolated cell injection in accordance with more expression of stromal-derived factor 1 (SDF-1), hepatocyte growth factor (HGF), and vascular endothelial growth factor (VEGF). Based on these results, clinical trial of autologous myoblast sheet transplantation for severe heart failure has started and the detailed results will appear soon. In the same manner with cell injection therapy, MSCs are used as a candidate cell source for human implantable cell sheet. Adipose tissue-derived MSCs and menstrual blood-derived MSCs have improved damaged heart function in rat infarction model (Miyahara, Nagaya et al. 2006; Hida, Nishiyama et al. 2008). MSCs can gradually grow to form a thick stratum containing newly formed blood vessels and some cells seem to differentiate into cardiomyocytes at least by histological analyses. Further studies will be needed to confirm the differentiation into functional beating cardiomyocytes and possibilities to differentiate into unexpected cell types. As emerging cell source, cell sheets of stem cell antigen 1-positive (Sca-1-positive) CSCs ameliorates cardiac dysfunction in mouse infarction model through cardiomyocyte differentiation and paracrine mechanisms mediated via soluble vascular cell adhesion molecule 1 (VCAM-1)/very late antigen-4 (VLA-4) signaling pathway (Matsuura, Honda et al. 2009). In addition, cardiac cell sheets originated from ESCs/iPSCs have been successfully fabricated and their transplantation into animal models is now ongoing. For enhancing the efficacy of cell sheet transplantation, gene-modified cell sheets have been examined. Bcl-2 expressed myoblast sheets prolonged survival, increased production of proangiogenic paracrine mediators, and enhanced the therapeutic efficacy (Kitabayashi, Siltanen et al. 2010). HGF overexpression in myoblast sheets enhances their angiogenic potential in rat chronic heart failure model (Siltanen, Kitabayashi et al. 2011). As another concept, cell sheets co-cultured with endothelial cell sources have been transplanted in rat infarction models. Transplantation of EPC co-cultured fibroblast sheet improved heart function more than only fibroblast sheet implantation or EPC injection (Kobayashi, Shimizu et al. 2008). Furthermore, endothelial cell co-culture within cardiomyocyte sheets induced more neovascularization and more improvement of cardiac function than only cardiomyocyte sheets (Sekine, Shimizu et al. 2008). These studies indicate advanced strategies of cell sheet transplantation. As mentioned previously, it is considered that the main mechanism of heart function improvement is neovascularization, fibrosis inhibition, apoptosis inhibition and stem cell recruitment due to various cytokines secreted from grafted cells. In comparison with cell injection approach, increase of cell survival within myocardial patches leads to more cytokine secretion, then, resulting in more function improvement. In addition to cytokine secretion, myocardial patches may have girdling effect and prohibit heart dilatation. Therefore, myocardial patch transplantation is quite different cell delivery method from cell injection and has more potential to rescue diseased hearts. In the case of myocardial patches using beating cardiomyocytes, direct enhancement of contraction power is additionally
  • 27.
    Myocardial Tissue Engineering11 expected, however, electrical synchronization between host hearts and transplanted patches is a critical issue to be clarified. 5. Engineering plusatile myocardial tisssue Beyond myocardial patch fabrication, several research groups have challenged to fabricate pulsatile myocardial tissues by their original tissue engineering strategies. Bioengineered contractile myocardial tissues may realize new therapeutics for severe heart diseases and be useful as alternatives for animal models. 5.1 Pre-fabricated scaffold-based myocardial tissue fabrication The first approach for engineering functional myocardial tissue is seeding cardiomyocytes into synthetic or biological 3-D scaffolds. Vunjak-Novakovic and colleagues first reported that seeding primary cultured cardiomyocytes onto disc-shaped polyglycolic acid (PGA) scaffolds in rotating bioreactor system resulted in spontaneously pulsatile myocardial tissues (Papadaki, Bursac et al. 2001). Optimization of cell population, serum concentration and scaffold coating improved electrical conduction velocity of engineered constructs. Radisc and colleagues seeded rat cardiomyocytes in Matrigel onto collagen sponges and stimulated the constructs electrically. The stimulation improved the conductive and contractile properties in accordance with increased expression of myosin heavy chain and connexion 43. Furthermore, cardiomyocytes in the electrically stimulated constructs were more aligned and elongated as same as those in native heart tissue (Radisic, Park et al. 2007). Following these studies, many research groups have started to engineer myocardial tissue in vitro by using various types of scaffolds. Scaffold porosity is one of the critical factors for pre-fabricated scaffold-based tissue engineering. High porosity increases seeded cell number and facilitates mass transport. Surface modyfication is also important for cell attachment and survival. Laminin coating improved cardiomyocyte adhesiveness. In addition, scaffold elasticity and degradability affect contraction property of engineered myocardium. Further studies are ongoing to development appropriate scaffold materials for myocardial tissue engineering. 5.2 Hydrogel-based myocardial tissue fabrication The second approach is to form 3-D tissues by gelling of cardiac cell and matrix solution mixture. Eschenhagen and Zimmermann have continuously developed this strategy using collagen gel and successfully engineered macroscopically beating cardiac tissues (Zimmermann and Cesnjevar 2009). First, neonatal rat cardiomyocytes were suspended in collagen I solution and the mixture was poured into the mold. After gelling, the constructs were unidirectionally stretched with the mechanical devise. They have also realized contraction force measurement. Cyclic stretch introduced cell alignment along the stretching direction and increased mitochondrial density, leading to native heart-like tissue. The contraction force of engineered myocardium was comparative with native heart tissue and responded to pharmacological agents properly. Ring-shaped myocardial tissues were also fabricated and combined 5 constructs were transplanted onto infarcted rat hearts. Interestingly, the constructs synchronized to each other and improved damaged heart function. They have also confirmed that co-culture constructs including cardiomyocytes,
  • 28.
    Tissue Engineering forTissue and Organ Regeneration 12 fibroblasts and endothelial cells were superior to cardiomyocyte rich constructs in morphology and function. Recently, they have also started to utilize human cardiomyocytes differentiated from ESC/iPSC as cell source and challenged to create human myocardial tissues (Zimmermann 2011). In contrast to pre-fabricated scaffold usage, relatively homogeneous myocardial tissues are engineered by hydrogel-based approach. Therefore collagen gel-based myocardial tissue engineering has now become popular in the world. 5.3 Cell sheet-based myocardial tissue fabrication The third approach is to engineer 3-D pulsatile myocardial tissues by stacking cardiac cell sheets. As mentioned previously, 2-D cell sheets can be harvested from temperature- responsive culture dishes only by lowering temperature and do not include any materials. 3- D tissues are constructed by layering cell sheets. Because 2-D confluent cells are directly stacked without any scaffolds, resulting constructs are cell-dense 3-D tissues. It is well- known that 2-D confluent cardiomyocytes connect to each other electrically via gap junctions resulting in synchronized beating. Cardiac cell sheets harvested from temperature- responsive culture dishes maintain this synchronized pulsation (Shimizu, Yamato et al. 2002). For creating 3-D functional heart tissues by layering cardiac cell sheets, morphological and electrical communications between cell sheets are critical. Multiple-electrode extracellular recording system revealed that double-layer rat cardiac cell sheets coupled electrically about one hour after layering and histological analysis showed the existence of connexin 43 between two cardiac cell sheets. Adhesive proteins deposited on cell sheet surface are considered to promote these rapid electrical communications (Haraguchi, Shimizu et al. 2006). Stacked cardiac cell sheets beat synchronously in macroscopic view and the constructs transplanted into rat subcutaneous tissues also pulsated continuously at least up to one year and eight months after implantation. Morphological analyses showed elongated cardiomyocytes, well-differentiated sarcomeres, gap junctions and multiple blood vessels, which were characteristic structure of native heart tissue (Shimizu, Yamato et al. 2002). Long-term observation revealed that their size, conduction velocity, and contractile force increased in proportion to the host growth (Shimizu, Sekine et al. 2006). Recently, fabrication of cardiac cell sheets using ESC-originated cardiomyocytes have just started and human cardiac cell sheets will appear in near future. 5.4 Fabrication of vascularized myocardial tissue One of the major obstacles in myocardial tissue engineering is scaling-up of the constructs. Insufficient supply of oxygen and nutrient, and waste accumulation limit their thickness. Actually, cells are sparse in the central area, on the other hand, cells are dense in the outer surface (100-200μm) area in scaffold-based myocardial tissue engineering. In the case of cell sheet-based myocardial tissue engineering, thickness limit is approximately 80μm (3 layers) (Shimizu, Sekine et al. 2006). Several approaches have been examined in the point of view overcoming diffusion limit. Perfusion of culture media through the constructs using porous scaffolds is one possible approach. Media penetration increased cell migration depth and improved cell metabolism. However shear stress due to media flow may prohibit tight cell attachment on the scaffold material. Media perfusion with oxygen carrier, perfluorocarbon (PFC) has been also examined for improving oxygen transport. PFC usage increased cell proliferation and improved pulsatile function. Media penetration is useful to some extent, however, it becomes more difficult as cell density increases.
  • 29.
    Myocardial Tissue Engineering13 To overcome this problem, it has been requested to develop new technologies for introducing vasculature or vascular-like structure into engineered tissues. Several researchers have tried to generate microchannel network within porous 3-D scaffolds by microfabrication techniques including CO2 laser ablation. The technology has not reached to mimicking native micro capillary network. On the other hand, recent studies have revealed that co-cultured endothelial cells within cardiac constructs can spontaneously form vascular-like network in vitro and tubular formation has been found in some parts. It has been also confirmed that this pre-vascular structure connected to host blood vessels immediately after transplantation and the newly developed vessels within the constructs were blood-supplied within a few days (Sekiya, Shimizu et al. 2006). We have already demonstrated that the tissue thickness of cardiac cell sheets co-cultured with endothelial cells were just twice as the thickness of cardiac cell sheets without endothelial cells (Sekine, Shimizu et al. 2008). Although endothelial cell co-culture is helpful for accelerating blood vessel formation, more scaling-up is still limited due to primary ischemia until sufficient vascularization. One possible idea for scaling-up is utilizing in vivo vascularization power. Our group has reported that triple-layer cardiac cell sheets were repeatedly implanted after waiting enough vascular formation within previously implanted tissues. In result, synchronously beating thick myocardial tissues with sufficient micro capillaries were successfully fabricated and 10-times transplantation of triple-layer constructs (totally 30 sheets) formed 1-mm thick, pulsatile myocardial tissues. Furthermore, when triple-layer grafts were transplanted repeatedly over a surgically connectable artery and vein in leg, the multilayer constructs were blood-supplied from the thick artery and vein. The constructs were successfully resected with the connectable blood vessels and were ectopically transplanted in neck with direct vessel anastomoses (Shimizu, Sekine et al. 2006). Recently several groups have also utilized in vivo power for myocardial tissue engineering. Cardiomyocytes, ECM alternatives and native blood vessels were packed in the special chamber and incubated in vivo. Vascularized heart-like tissues were created in the body (Morritt, Bortolotto et al. 2007; Birla, Dhawan et al. 2009). Furthemore, next challenge is now in vitro fabrication of vascularized myocardial tissues. Kofidis and colleagues have constructed fibrin gel-based myocardial tissues containing rat aortas (1-2mm), through which culture media was perfused (Kofidis, Lenz et al. 2003). Cell survival and metabolism were improved, however formation of functional blood vessels connecting with central aortas were not clear. We are now trying to promote endothelial cell tubular formation within in vitro engineered cardiac tissues and to perfuse culture media through the newly formed vessels using perfusion bioreactors. Further studies will be needed to break through the obstacles for in vitro scaling up. 5.5 From tissue engineering to organ engineering For future organ engineering, some groups have challenged to engineer myocardial constructs with pumping function. Ott and colleagues have used decellularized organ as a scaffold. They decellularized rat whole hearts and re-seeded cardiac cells into decellularized hearts. Heart contraction was recovered and pump function was generated (Ott, Matthiesen et al. 2008). Zimmermann’s group developed pouch-like myocardial tissue by their technology as previously described and covered heart with pouch-like constructs (Yildirim, Naito et al. 2007).
  • 30.
    Tissue Engineering forTissue and Organ Regeneration 14 Regarding cell sheet technology, myocardial tubes have been fabricated by wrapping rat cardiac cell sheets around fibrin tubes and rat resected aortas. The engineered myocardial tubes revealed spontaneous, synchronized pulsation and small but significant inner pressure changes (about 0.1 mmHg) in vitro (Kubo, Shimizu et al. 2007). On the other hand, resected rat aortas wrapped with cardiac cell sheets were micro surgically transplanted in place of the abdominal aorta. After 1 month, in vivo myocardial tubes demonstrated spontaneous beating and evoked independent blood pressures (about 6 mm Hg). The value of in vivo myocardial tubes was much bigger than in vitro myocardial tubes (Sekine, Shimizu et al. 2006). Comparing in vitro and in vivo, it was considered that pulsation due to host blood flow has induced cardiomyocyte hypertrophy, leading to improvement of pumping function. Therefore pulsatile perfusion bioreactors may improve pumping function of in vitro engineered myocardial tubes. Thus, small size myocardial constructs evoking pumping function have been realized. Expansion and selection of cardiomyocytes, and sufficient blood vessel formation for scaling-up are now critical issues for organ engineering. 6. Conclusions As the first generation of cardiac regenerative therapy, many clinical trials of cell injection therapy have been already performed. The controversial arguments about its effectiveness will be settled in next several years. Tissue engineered myocardial patches have now emerged as the second generation and previous studies indicate promising potential for rescuing damaged heart. As the third generation, tissue-engineered pulsatile myocardial tissues should support heart contraction physically. Furthermore, future development of cell sourcing and scaling-up technologies may realize “bioengineered hearts”. 7. Acknowledgment This work is granted by the Japan Society for the Promotion of Science (JSPS) through the “Funding Program for World-Leading Innovative R&D on Science and Technology (FIRST Program),” initiated by the Council for Science and Technology Policy (CSTP). 8. References Asahara, T., T. Murohara, et al. (1997). Isolation of putative progenitor endothelial cells for angiogenesis. Science 275(5302): 964-967. Beltrami, A. P., L. Barlucchi, et al. (2003). Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 114(6): 763-776. Bergmann, O., R. D. Bhardwaj, et al. (2009). Evidence for cardiomyocyte renewal in humans. Science 324(5923): 98-102. Birla, R. K., V. Dhawan, et al. (2009). Cardiac cells implanted into a cylindrical, vascularized chamber in vivo: pressure generation and morphology. Biotechnol Lett 31(2): 191- 201. Chachques, J. C., J. C. Trainini, et al. (2007). Myocardial assistance by grafting a new bioartificial upgraded myocardium (MAGNUM clinical trial): one year follow-up. Cell Transplant 16(9): 927-934.
  • 31.
    Myocardial Tissue Engineering15 Haraguchi, Y., T. Shimizu, et al. (2006). Electrical coupling of cardiomyocyte sheets occurs rapidly via functional gap junction formation. Biomaterials 27(27): 4765-4774. Hata, H., G. Matsumiya, et al. (2006). Grafted skeletal myoblast sheets attenuate myocardial remodeling in pacing-induced canine heart failure model. J Thorac Cardiovasc Surg 132(4): 918-924. Hida, N., N. Nishiyama, et al. (2008). Novel cardiac precursor-like cells from human menstrual blood-derived mesenchymal cells. Stem Cells 26(7): 1695-1704. Hofmann, M., K. C. Wollert, et al. (2005). Monitoring of bone marrow cell homing into the infarcted human myocardium. Circulation 111(17): 2198-2202. Itabashi, Y., S. Miyoshi, et al. (2005). A new method for manufacturing cardiac cell sheets using fibrin-coated dishes and its electrophysiological studies by optical mapping. Artif Organs 29(2): 95-103. Ito, A., E. Hibino, et al. (2005). Construction and delivery of tissue-engineered human retinal pigment epithelial cell sheets, using magnetite nanoparticles and magnetic force. Tissue Eng 11(3-4): 489-496. Kitabayashi, K., A. Siltanen, et al. (2010). Bcl-2 expression enhances myoblast sheet transplantation therapy for acute myocardial infarction. Cell Transplant 19(5): 573- 588. Kobayashi, H., T. Shimizu, et al. (2008). Fibroblast sheets co-cultured with endothelial progenitor cells improve cardiac function of infarcted hearts. J Artif Organs 11(3): 141-147. Kofidis, T., J. L. de Bruin, et al. (2004). Injectable bioartificial myocardial tissue for large-scale intramural cell transfer and functional recovery of injured heart muscle. J Thorac Cardiovasc Surg 128(4): 571-578. Kofidis, T., A. Lenz, et al. (2003). Pulsatile perfusion and cardiomyocyte viability in a solid three-dimensional matrix. Biomaterials 24(27): 5009-5014. Kondoh, H., Y. Sawa, et al. (2006). Longer preservation of cardiac performance by sheet- shaped myoblast implantation in dilated cardiomyopathic hamsters. Cardiovasc Res 69(2): 466-475. Kubo, H., T. Shimizu, et al. (2007). Creation of myocardial tubes using cardiomyocyte sheets and an in vitro cell sheet-wrapping device. Biomaterials 28(24): 3508-3516. Lee, S. T., A. J. White, et al. (2011). Intramyocardial injection of autologous cardiospheres or cardiosphere-derived cells preserves function and minimizes adverse ventricular remodeling in pigs with heart failure post-myocardial infarction. J Am Coll Cardiol 57(4): 455-465. Leor, J., S. Aboulafia-Etzion, et al. (2000). Bioengineered cardiac grafts: A new approach to repair the infarcted myocardium? Circulation 102(19 Suppl 3): III56-61. Li, R. K., Z. Q. Jia, et al. (1999). Survival and function of bioengineered cardiac grafts. Circulation 100(19 Suppl): II63-69. Martin-Rendon, E., S. J. Brunskill, et al. (2008). Autologous bone marrow stem cells to treat acute myocardial infarction: a systematic review. Eur Heart J 29(15): 1807-1818. Matsuura, K., A. Honda, et al. (2009). Transplantation of cardiac progenitor cells ameliorates cardiac dysfunction after myocardial infarction in mice. J Clin Invest 119(8): 2204- 2217.
  • 32.
    Tissue Engineering forTissue and Organ Regeneration 16 Memon, I. A., Y. Sawa, et al. (2005). Repair of impaired myocardium by means of implantation of engineered autologous myoblast sheets. J Thorac Cardiovasc Surg 130(5): 1333-1341. Menasche, P., O. Alfieri, et al. (2008). The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial: first randomized placebo-controlled study of myoblast transplantation. Circulation 117(9): 1189-1200. Menasche, P., A. A. Hagege, et al. (2001). Myoblast transplantation for heart failure. Lancet 357(9252): 279-280. Mirsadraee, S., H. E. Wilcox, et al. (2006). Development and characterization of an acellular human pericardial matrix for tissue engineering. Tissue Eng 12(4): 763-773. Miyagawa, S., A. Saito, et al. (2010). Impaired myocardium regeneration with skeletal cell sheets--a preclinical trial for tissue-engineered regeneration therapy. Transplantation 90(4): 364-372. Miyagawa, S., Y. Sawa, et al. (2005). Tissue cardiomyoplasty using bioengineered contractile cardiomyocyte sheets to repair damaged myocardium: their integration with recipient myocardium. Transplantation 80(11): 1586-1595. Miyahara, Y., N. Nagaya, et al. (2006). Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat Med 12(4): 459-465. Morritt, A. N., S. K. Bortolotto, et al. (2007). Cardiac tissue engineering in an in vivo vascularized chamber. Circulation 115(3): 353-360. Oh, H., S. B. Bradfute, et al. (2003). Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A 100(21): 12313-12318. Okano, T., N. Yamada, et al. (1993). A novel recovery system for cultured cells using plasma-treated polystyrene dishes grafted with poly(N-isopropylacrylamide). J Biomed Mater Res 27(10): 1243-1251. Opie, S. R. and N. Dib (2006). Surgical and catheter delivery of autologous myoblasts in patients with congestive heart failure. Nat Clin Pract Cardiovasc Med 3 Suppl 1: S42- 45. Ott, H. C., T. S. Matthiesen, et al. (2008). Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial heart. Nat Med 14(2): 213-221. Papadaki, M., N. Bursac, et al. (2001). Tissue engineering of functional cardiac muscle: molecular, structural, and electrophysiological studies. Am J Physiol Heart Circ Physiol 280(1): H168-178. Psaltis, P. J., A. C. Zannettino, et al. (2008). Concise review: mesenchymal stromal cells: potential for cardiovascular repair. Stem Cells 26(9): 2201-2210. Puceat, M. (2008). Pharmacological approaches to regenerative strategies for the treatment of cardiovascular diseases. Curr Opin Pharmacol 8(2): 189-192. Radisic, M., H. Park, et al. (2007). Biomimetic approach to cardiac tissue engineering. Philos Trans R Soc Lond B Biol Sci 362(1484): 1357-1368. Sekine, H., T. Shimizu, et al. (2008). Endothelial cell coculture within tissue-engineered cardiomyocyte sheets enhances neovascularization and improves cardiac function of ischemic hearts. Circulation 118(14 Suppl): S145-152. Sekine, H., T. Shimizu, et al. (2006). Cardiomyocyte bridging between hearts and bioengineered myocardial tissues with mesenchymal transition of mesothelial cells. J Heart Lung Transplant 25(3): 324-332.
  • 33.
    Myocardial Tissue Engineering17 Sekine, H., T. Shimizu, et al. (2006). Pulsatile myocardial tubes fabricated with cell sheet engineering. Circulation 114(1 Suppl): I87-93. Sekiya, N., G. Matsumiya, et al. (2009). Layered implantation of myoblast sheets attenuates adverse cardiac remodeling of the infarcted heart. J Thorac Cardiovasc Surg 138(4): 985-993. Sekiya, S., T. Shimizu, et al. (2006). Bioengineered cardiac cell sheet grafts have intrinsic angiogenic potential. Biochem Biophys Res Commun 341(2): 573-582. Shimizu, T., H. Sekine, et al. (2006). Long-term survival and growth of pulsatile myocardial tissue grafts engineered by the layering of cardiomyocyte sheets. Tissue Eng 12(3): 499-507. Shimizu, T., H. Sekine, et al. (2009). Cell sheet-based myocardial tissue engineering: new hope for damaged heart rescue. Curr Pharm Des 15(24): 2807-2814. Shimizu, T., H. Sekine, et al. (2006). Polysurgery of cell sheet grafts overcomes diffusion limits to produce thick, vascularized myocardial tissues. FASEB J 20(6): 708-710. Shimizu, T., M. Yamato, et al. (2002). Electrically communicating three-dimensional cardiac tissue mimic fabricated by layered cultured cardiomyocyte sheets. J Biomed Mater Res 60(1): 110-117. Shimizu, T., M. Yamato, et al. (2002). Fabrication of pulsatile cardiac tissue grafts using a novel 3-dimensional cell sheet manipulation technique and temperature-responsive cell culture surfaces. Circ Res 90(3): e40. Shimizu, T., M. Yamato, et al. (2003). Cell sheet engineering for myocardial tissue reconstruction. Biomaterials 24(13): 2309-2316. Shin, M., O. Ishii, et al. (2004). Contractile cardiac grafts using a novel nanofibrous mesh. Biomaterials 25(17): 3717-3723. Siltanen, A., K. Kitabayashi, et al. (2011). hHGF Overexpression in Myoblast Sheets Enhances Their Angiogenic Potential in Rat Chronic Heart Failure. PLoS One 6(4): e19161. Tan, M. Y., W. Zhi, et al. (2009). Repair of infarcted myocardium using mesenchymal stem cell seeded small intestinal submucosa in rabbits. Biomaterials 30(19): 3234-3240. Vunjak-Novakovic, G., N. Tandon, et al. (2010). Challenges in cardiac tissue engineering. Tissue Eng Part B Rev 16(2): 169-187. Wollert, K. C. (2008). Cell therapy for acute myocardial infarction. Curr Opin Pharmacol 8(2): 202-210. Yildirim, Y., H. Naito, et al. (2007). Development of a biological ventricular assist device: preliminary data from a small animal model. Circulation 116(11 Suppl): I16-23. Yuasa, S., Y. Itabashi, et al. (2005). Transient inhibition of BMP signaling by Noggin induces cardiomyocyte differentiation of mouse embryonic stem cells. Nat Biotechnol 23(5): 607-611. Zhang, M., D. Methot, et al. (2001). Cardiomyocyte grafting for cardiac repair: graft cell death and anti-death strategies. J Mol Cell Cardiol 33(5): 907-921. Zhu, W., I. Shiojima, et al. (2008). IGFBP-4 is an inhibitor of canonical Wnt signalling required for cardiogenesis. Nature 454(7202): 345-349. Zimmermann, W. H. (2011). Embryonic and embryonic-like stem cells in heart muscle engineering. J Mol Cell Cardiol 50(2): 320-326. Zimmermann, W. H. and R. Cesnjevar (2009). Cardiac tissue engineering: implications for pediatric heart surgery. Pediatr Cardiol 30(5): 716-723.
  • 34.
    Tissue Engineering forTissue and Organ Regeneration 18 Zimmermann, W. H., M. Didie, et al. (2006). Heart muscle engineering: an update on cardiac muscle replacement therapy. Cardiovasc Res 71(3): 419-429. Zimmermann, W. H., I. Melnychenko, et al. (2006). Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nat Med 12(4): 452-458. Zimmermann, W. H., K. Schneiderbanger, et al. (2002). Tissue engineering of a differentiated cardiac muscle construct. Circ Res 90(2): 223-230.
  • 35.
    2 Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site Giancarlo Forte1, Stefania Pagliari2, Francesca Pagliari2, Paolo Di Nardo2 and Takao Aoyagi1 1Biomaterials Center, International Center for Materials Nanoarchitectonics (MANA), National Institute for Materials Science (NIMS), Tsukuba, 2Laboratorio di Cardiologia Molecolare e Cellulare, Dipartimento di Medicina Interna, Università di “Tor Vergata”, Roma, 1Japan 2Italy 1. Introduction In healthy human hearts, only 10-20% of the total cells are contractile cardiomyocytes and, at the age of 25 years, no more than 1% of them are annually substituted by progenitor cells, this percentage reducing to less than 0.5% at the age of 75. In total, less than 50% of cardiomyocytes are renewed during a normal human life span [1]. For this reason, the topic of cardiac repair is among the major challenges for the tissue engineers worldwide. In fact, cardiac diseases are a predominant cause of mortality and morbidity in industrialized countries, despite the recent advancements achieved in pharmacological treatment and interventional cardiology procedures. Nonetheless, end-stage heart failure management still relies on organ transplantation as unique approach, and, notwithstanding the use of massive immunosuppressive drugs, still a percentage falling within 20%-40% of patients encounters immune rejection during the first year post-transplant [2]. Among the patients not facing severe immune rejection, almost 70% is forced to retire or reduce their working activity, their survival rate falling below 70% during the first five years post organ transplantation [3]. Last, but not least, the economic impact of cardiovascular diseases and stroke has been estimated in 2010 at $503.2 billion [4]. Currently, post-infarction myocardial revascularization protocols include the administration of raw bone marrow stem cells, while a number of clinical trials have been performed or are currently in progress in which different cell subsets are implanted in the damaged tissue by means of surgical techniques. The results of such trials are still controversial. In fact, when autologous skeletal myoblasts were injected into the heart of patients suffering from ischemic cardiomyopathy, the modest functional improvement obtained was impaired by the arising of arrhythmia events, thus requiring the adoption of a pacemaker [5]. On the other side, intracoronary administration of bone marrow mesenchymal stem cells resulted in minimal improvements in cardiac contractile function in patients with dilated cardiomyopathy [6]. These mild results were mostly ascribed to a paracrine effect exerted on host tissue, rather than to a direct contribution of stem cells to the contractile activity.
  • 36.
    Tissue Engineering forTissue and Organ Regeneration 20 Thus, among the criticisms to be challenged before efficient cell therapy protocols for cardiac diseases can be setup, the choice of the appropriate cell subset to generate new vessels and contractile cardiomyocytes, as well as the route of cell delivery remain key steps. The solution of such problems requires additional efforts in basic research to clarify the processes leading to stem cell differentiation as well as technological advancements to setup efficient protocols to implant the cells. In principle, adult stem cells could be extracted from patient’s own tissues and expanded in culture by means of well-known techniques (Figure 1). Fig. 1. Cardiac Tissue Engineering paradigm. Adult stem cells can be harvested, purified from the patient and expanded in culture. Such cells can be delivered to the injured heart by injection (intramural or through bloodstream with or without injectable carriers), or in the form of solid bio-constructs. Stem cell-derived bio-constructs can be obtained by culturing the cells on scaffolds or by scaffold-free technology Nonetheless, a number of issues should be challenged before safe procedures to manipulate stem cells in vitro for cardiac transplant can be setup. In fact, stem cells should be amplified in vitro to reach a critical number (Figure 1). During this passage, malignant transformation is likely to occur in ex vivo cells when standard culture conditions are adopted to expand stem cells [7, 8]. On the other side, stem cells could encounter senescence after a short number of passages in vitro [9]. Moreover, the use of animal- derived supplements during the phase of cell expansion would hinder the use of stem cells for cardiac cell therapy.
  • 37.
    Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site 21 The employment of autologous stem cells would avoid the problem of immune rejection and the need for immune-suppressive drugs, while, in the treatment of pathologies for which a genetic basis is suspected the use of autologous cells is hampered. As far as the use of autologous cells is concerned, the possibility that a significant patient-to-patient variability in stem cell quality exists should be taken into account [10]. Finally, the use of cellular and tissue-based products in human disease therapy is subjected to regulations issued by the European Union and Food and Drug Administration (FDA) aimed at establishing classification criteria for advanced therapy medicinal products (ATMP). In particular, the European Regulation states that human cells to be used in cell therapy have to comply with the principles of Good Manufacturing Practice (GMP) protocols [11, 12]. 2. Adult stem cells for cardiac repair A number of stem cells and progenitors have been so far proposed for cardiac repair, due to the inability of cardiomyocytes to proliferate after birth [1]. Among the cell sources challenged for the possibility to produce new cardiomyocytes, skeletal myoblasts have proven to be able to acquire a contractile phenotype in vitro [13]. Moreover, when implanted in vivo in a canine model of dilated cardiomyopathy (DCM), they attenuated cardiac remodeling [14]. This result is likely to be due to the fusion of skeletal myoblasts with the surrounding myocardium rather than to direct cell differentiation, as suggested by in vitro experiments [15]. As discussed in the following section, clinical trials demonstrated that skeletal myoblasts are not able to couple electrically with host tissue, leading to arrhythmia events [5]. The role of hematopoietic stem cells (HSC) in cardiac repair has been investigated by several research groups and their contribution to cardiac regeneration in vivo has been heavily debated, being the ability of HSC to transdifferentiate to other lineages still questionable. Indeed, evidence of the ability of bone marrow-derived c-kit+ HSC to help cardiac tissue healing has been given using two different approaches: c-kit+ cells were (i) either delivered to the infarcted site by intramural injection [16] or (ii) mobilized from bone marrow through growth factor administration [17]. More recently, elegant experiments compellingly clarified that HSC are not able to acquire contractile phenotype in vivo [18-20]. Nonetheless, a subset of bone marrow hematopoietic precursors expressing CD34 and CD133 has been proven to contain endothelial progenitors. Thus, they have been tested for revascularization protocols in hind limb ischemic animals and could be proposed for cardiac infarction therapy [21]. On the other hand, the results obtained in preliminary investigations in which another bone marrow-derived stem cell subset, mesenchymal stem cells (BM-MSC or MSC) were challenged as a candidate for cellular cardiomyoplasty, raised great enthusiasm for such a cell subpopulation. Recent studies clarified that the direct contribution of MSC to cardiac repair in terms of production of new contractile cells is minimal if any, while a paracrine effect on the diseased tissue of such cells is universally recognized [22]. Such cells are also appealing for their ability to induce a certain degree of immune tolerance [23]. The presence of a small reservoir of cardiac resident progenitor cells (CPC or CSC) has been recently demonstrated in human as well as in other mammals’ heart [24]. Such tissue- resident cells participate in myocardial homeostasis and retain a limited regenerative capacity throughout organism lifespan [1]. All the subsets so far identified through the expression of stemness markers (c-kit+, Sca-1+, Islet-1+) demonstrated the ability to give birth to new contractile cells in vitro, while only c-kit+, Sca-1+ progenitors were shown to be
  • 38.
    Tissue Engineering forTissue and Organ Regeneration 22 involved in post-natal cardiac tissue homeostasis in vivo [25]. In fact, the presence of Islet-1+ cells appears to be limited to fetal life and their contribution to the endogenous program of cardiovascular repair is still unknown; on the other hand, the very low number of c-kit+ and Sca-1+ cells in the myocardium is considered the limiting factor of cardiac regeneration [26]. Furthermore, among the adult stem cells, a novel “artificial” subset can be recognized: induced pluripotent stem cells (iPSC, Figure 2). This cell type can be produced in vitro by transducing somatic cells with a combination of transcription factors able to induce the nuclear reprogramming of differentiated cells. These cells, which display the functional features of pluripotent embryonic stem cells, have been credited of the ability to produce new cardiomyocytes. They could thus be the source of autologous, although genetically modified, patient-specific contractile cells [27]. Moreover, the possibility to directly obtain functional cardiomyocytes by the genetic reprogramming of postnatal cardiac or dermal fibroblasts has been demonstrated [28]. Such a result was firstly obtained in vitro but also when the cells were transplanted into mouse hearts one day after transduction of transcription factors (GATA-4, MEF-2c, Tbx-5) known to be involved in cardiac muscle development. Nonetheless, the reprogramming and differentiation efficiency of these cells appears to be really low, thus requiring an efficient purification step before they can be implanted in vivo. Additionally, safety concerns due to the use of genetically modified cells and / or viral vectors remain. Fig. 2. Induced Pluripotent Stem Cell Generation. Induced pluripotent Stem Cells (iPSC) can be generated by reprogramming somatic cells through their transduction with four transcription factors. iPSC share functional similarities with Embryonic Stem Cells (ESC) and can be differentiated towards cardiomyocytes, thus representing an autologous source of contractile cells
  • 39.
    Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site 23 3. Stem cell delivery to the injured heart As previously said, cell route of delivery to damaged heart represents the major topic in the setup of efficient, minimally invasive techniques to treat cardiac pathologies. Recently, a number of techniques to deliver stem cells to the injured site have been proposed but questions remain regarding the optimal approach able to favor high cell retention, differentiation rate and clinically relevant improvement in cardiac performance. a) Direct injection Stem cell direct intramural injection, including trans-epicardial and trans-endocardial cell injection, is the elective strategy for patients with severe occlusion of coronary vessels. In particular, trans-epicardial approach consists in the direct injection of a high number of cells into the infarcted area or around the border zone. Endocardial stem cell injection is performed using catheters such as MyoStarTM injection catheter (Biosense Webster) integrated with imaging systems like NOGA® system (Cordis Corp., Warren, NJ, USA), which allows real-time three-dimensional reconstruction of left ventricle as well as the targeting and functional assessment of specific myocardial area [29]. Such procedures are highly invasive since they require open-heart surgery and gave contrasting results so far. For example, pre-clinical studies performed on experimental animals demonstrated that, although a certain extent of cardiac repair was achieved when bone marrow Stro-3+ perivascular cells are implanted in vivo, the cells vanished from the application site within few days [30]. In other reports, when Sca-1+ cardiac resident stem cells were injected in infarction border zone, a modest but significant improvement in cardiac function was reported, with evidence of cell engraftment and differentiation [31]. Finally, in another pre-clinical study, bone marrow-derived c-kit+ cells were shown to repair entire ventricular areas while massively engrafting and differentiating in contractile and vascular figures in vivo [32]. Of interest, independent groups already demonstrated that c- kit+ bone marrow-derived hematopoietic stem cells fail to acquire contractile phenotype when implanted in diseased myocardium [19, 20]. Such discrepancies are not surprising since different stem cell subsets or preparation protocols were probably used in these studies. Stem cells can be delivered intravenously to the heart, through coronary arteries or even through retrograde coronary sinus. The major drawback of stem cells being infused through peripheral venous system seems to be the low retention of cells into infarcted area. Results obtained in pre-clinical animal models showed that this minimally invasive approach results in a significant percentage of injected cells being sequestered in lungs, liver or spleen, due to blood flow [33]. On the other hand, intracoronary or retrograde coronary sinus infusion of the cells are mainly performed after acute myocardial infarction using an angioplasty balloon and high pressure to deliver cells to heart muscle [34]. The coronary route was proven to be free of stem cell systemic delivery, while a limited number of cells could be found in the infarcted area [35]. Finally, an interesting attempt with stem cells being injected into the pericardial cavity has been proposed. By this means, a higher number of cells could be deposited and retained in the pericardial cavity, while migration across the visceral pericardium is required (Table 1).
  • 40.
    Tissue Engineering forTissue and Organ Regeneration 24 Table 1. Advantages and disadvantages of injecting stem cells by intravenous, intracoronary, intramyocardial, retrograde coronary sinus or intra-pericardial route b) Injectable scaffolds Injectable scaffolds are defined as materials offering the unique solution of replacing damaged myocardial ECM and/or delivering cells directly to the infarcted region while holding the potential for minimally invasive delivery [36]. Such scaffolds can be composed of biocompatible microspheres or in situ gelling materials having reasonable dimensions as to surpass capillary barrier. They are considered a promising tool for stem cell delivery to damaged myocardium. In situ gelling materials are generally made of components of extracellular matrix (ECM), which are induced to a transition after being implanted in situ. Complex injectable gelling materials have been prepared by decellularization technique out of ventricular or epicardial ECM, thus possibly avoiding animal-derived components and paving the way to the definition of patient-specific treatments. The use of injectable, synthetic microspheres has already been proven promising in the treatment of neurological diseases in vivo [37]. Recently the possibility of using injectable scaffolds in cardiac cell therapy has been explored by interfacing murine mesenchymal (mMSC) and cardiac stem cell (mCSC) lines with poly-lactic acid (PLA) microspheres having a diameter of 30 and 100 μm. Preliminary in vitro experiments demonstrated that such cells can be grown onto PLA microspheres while preserving their phenotype, but the formation of cell clumps can hamper the application of this technique [38]. The use of dynamic seeding techniques (i.e. bioreactors) would favor a more homogeneous distribution of the cells. An interesting approach has been recently proposed to deliver human mesenchymal stem cells to the injured myocardium: RGD-modified alginate microsphere (diameter: 200-700 μm) encapsulation of hMSC was setup. In vitro experiments showed that hMSC could survive, proliferate and migrate through the porous material. When intramyocardially injected in a rat model of myocardial infarction by left anterior descendant coronary (LAD) ligation, cell-loaded alginate microspheres promoted angiogenesis and prevented LV negative remodeling [39]. Nonetheless, few human cells were found in the injection area after few days, while microbead remains were still present
  • 41.
    Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site 25 within host myocardium 10 weeks after the injection. The aspect of microbead resorption should thus be addressed before clinical perspectives could be foreseen. c) Scaffold-based technology The possibility of using biocompatible scaffolds to deliver stem cells to the injured heart has been explored by a number of independent research groups so far. The scaffolds proposed are natural of synthetic but when designing cardiac-specific constructs, a number of requirements should be fulfilled. For example, it cannot be neglected that myocardial contractile function relies on the transmission of electrical and mechanical forces throughout a functional syncytium. So, the integrity of the tissue has to be preserved. For this reason, a cardiac-specific scaffold should comply with tissue architecture and thus be deformable enough to indulge and, if possible sustain cardiac contraction. Moreover, as far as stem cell engraftment is concerned, scaffolds should be able to start at least cell alignment and commitment to favor stem cell electromechanical coupling with host tissue. In this respect, the work of Mandoli and collaborators using Cerium Oxyde nanoparticles to affect poly- lactic acid film surface and obtain a controlled nanorugosity appears intriguing [40]. In fact, far from being a noxious compound for stem cells, ceria was able to induce cardiac stem cell alignment and growth. Nonetheless, cardiac tissue is extremely complex and highly demanding in terms of blood supply and catabolite removal, so that porous scaffolds that could allow microvascular branches formation and oxygen perfusion are to be preferred. To fulfill such requirements, the first attempts were performed by the group of Thomas Eschenhagen. Neonatal cardiomyocytes were seeded in Collagen I + Matrigel to produce Engineered Heart Tissue (EHT). Continuous contractile activity up to 1 week in vitro as well as cell survival and integration in vivo in singenic rat hearts were reported [41]. In another attempt, anisotropic accordion-like honeycomb scaffolds were prepared by excimer laser microablation using poly(glycerol sebacate) as an elastomeric tool to mimic anisotropic cardiac muscle stiffness distribution [42]. Although the authors demonstrated that such scaffolds promote neonatal rat cardiomyocyte alignment and contraction, in vivo testing has not been performed so far. The same material has been utilized to produce elastomeric patches on which human embryonic stem cell-derived cardiomyocytes were grown, showing that it is indeed possible to observe spontaneous beating activity in vitro up to 3 months [43]. Such patches were shown to be suitable as delivery systems and, when sutured in the absence of cells onto healthy rat left ventricle, they did not affect cardiac contractile activity. More basic studies were also conducted to study the ability of stem cells to interface with different synthetic and natural materials. In this respect, few research groups focused on the possibility to drive a certain extent of stem cell commitment through tailoring scaffold physical and chemical properties, independently of biological cues. In this respect, a common agreement on the ability of stem cells to sense substrate rugosity and elasticity has been reached [44]. Thus, in order to rule out the occurrence of spontaneous events of differentiation in implanted cells, the possibility to induce in vitro stem cell commitment on scaffolds towards a desired phenotype is being investigated. Indeed, Engler and collaborators compellingly demonstrated that the possibility to affect stem cell fate determination by simply tuning substrate elasticity as to match tissue-specific stiffness, exists. Recently, this concept has been corroborated by other research groups, showing that cardiac resident progenitors (Sca-1+ CPC) can be committed to cardiac phenotype by the physico-chemical signals arising from matrix, but biological factors are needed to complete the differentiation process [45, 46].
  • 42.
    Tissue Engineering forTissue and Organ Regeneration 26 d) Preparation of thick cardiac substitutes by Scaffold-free technology To overcome the problem of poor cell retention reported in cell injection experiments in the heart [30] and avoid the release of possibly harmful scaffold byproducts, scaffold-free technology has been developed, in which cells are grown in a monolayer onto thermo- responsive surfaces and easily detached in the form of cell sheet by lowering the temperature [47]. Such technology takes advantage of the ability of polymers like poly-N- isopropylacrylamide (PNIPAAm) to shift between hydrophobic and hydrophilic status when the temperature ranges from 37ºC to 32ºC. Cell sheets can be serially stacked to obtain multilayered scaffoldless constructs (Figure 3). Such an approach has already been applied to obtain cell sheets composed of rodent [48, 49] and human [50] cells. Given the need for thick cardiac substitutes suited to comply with cardiac muscle continuous contractility, thermo-responsive technology has been envisaged as a possible answer to the lack of heart donors. Pre-clinical trials performed onto experimentally infarcted animals demonstrated that when a murine adipose-derived monolayer sheet is leant onto injured myocardium, it can be retained and help tissue repair [48]. Similarly, striking results are obtained when a Sca-1+ cardiac progenitor cell-derived sheet is used [49]. Finally, an interesting approach has been recently proposed to deliver cardiac stem cells cultured in the form of cardiospheres to the injured heart: cardiospheres were embedded into a cardiac stromal cell-derived sheet obtained by using poly-lysine/ collagen IV-coated dishes [51]. The formation of mature vessels as well as new cardiomyocytes in vivo was reported after 3 weeks. Fig. 3. Generation of scaffoldless multilayered bio-constructs by means of thermo- responsive technology: cells grown in a monolayer onto thermo-responsive poly-N- isopropilacrylamide (PNIPAAm)-coated dishes can be detached by lowering the temperature below 32°C. At 37°C the surface is highly hydrophobic and allows cell adhesion. When the temperature is lowered, PNIPAAm becomes hydrophilic, the cell sheet is detached and extracellular matrix (ECM) preserved. Multilayered cell sheets can be obtained by serially stacking monolayered sheets
  • 43.
    Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site 27 4. Clinical trials In the attempt to transfer bench experience to bedside, a number of clinical trials in which different stem cell or progenitor subsets are used have been approved (see http://www.clinicaltrials.gov). Most of them are still in the recruitment phase while some already gave indications and preliminary results. Since most of the ongoing trials are based on the injection of raw stem cell preparations (mostly bone marrow-derived cells), the time and route of cell application remain the key problems to be addressed before proceeding to routine clinical practice. In this respect, recent animal experiments demonstrated that the acute phase of myocardial infarction is probably not suitable for stem cell engraftment and differentiation [52]. Therefore, the right moment in which stem cells should be delivered is to be studied. An overview on some of the ongoing clinical trials is given below. 1. MAGIC (Myoblast Autologous Grafting in Ischemic Cardiomyopathy). In one of the first phase II clinical trials setup to study the possibility to use stem cells to treat cardiac pathologies, ninety-seven (97) patients undergoing coronary artery bypass grafting (CABG) were enrolled. 400-800 X 106 autologous myoblasts harvested from patient muscle biopsy were implanted in the akinetic area of ventricular wall 21 days after in vitro culture. The follow-up after 30 days and 6 months demonstrated the arising of arrhythmia events, thus requiring the implantation of pacemaker. Moreover, no cardiac function improvement was reported. Such negative results were ascribed to the inability of skeletal myoblasts to balance cell death and achieve complete electromechanical integration with the recipient myocardium. Finally, skeletal myoblast administration was reported to determine no enhancement in major cardiac adverse events and mild effects on left ventricular remodeling process [53, 54]. More recently, final results from SEISMIC [Safety and Effects of Implanted (Autologous) Skeletal Myoblasts (MyoCell) Using an Injection Catheter] Trial, a phase II-a study encompassing 40 patients experiencing congestive heart failure and receiving percutaneous intramyocardial injection of autologous skeletal myoblasts, reported the feasibility and safety of this procedure without significant arrhythmogenic events recorded at 6-month follow-up with respect to control groups, although left ventricular ejection fraction did not result significantly improve. These encouraging results suggest that myoblast cell therapy could be considered as a potential effective treatment when associated with standard medical therapy in patients with previously implanted cardiac defibrillators [55]. 2. TOPCARE-CHD, -AMI, -DCM (Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction, Chronic Stable Ischemic Heart disease or Dilated Cardiomyopathy). In this complex clinical trial, a total of 346 patients were classified to CHD, AMI or DCM pathologies and infused either with bone marrow cells (BMCs), blood-derived stem cells, or no infusion. In TOPCARE-CHD, 121 patients (mean age: 59) with chronic stable ischemic heart disease (CHD) were treated. Although complications occurred in 21% of the patients during 3 months follow-up, BMC intracoronary administration was related with a reduction of both brain and atrial natriuretic peptide (NTP) serum levels (indicators of LV remodelling process) in the remaining population (79%), especially in patients with higher NTP levels at baseline and receiving a greater BMC number with a high functional capacity. Moreover, these results were also correlated with a left ventricular ejection fraction (LVEF) increase and better survival during the further follow-up, suggesting that cell therapy could be
  • 44.
    Tissue Engineering forTissue and Organ Regeneration 28 associated with cardiac function enhancements in patients with advanced chronic post- infarction heart failure [56]. Similarly, two hundred and four (204) patients were treated using bone-marrow-derived progenitor cells directly into the infarct artery three to seven days after an acute myocardial infarction (AMI). A statistically significant 2.5% improvement in left ventricular ejection fraction at four months was reported for patients randomized to the bone marrow injection [57]. Finally, intracoronary infusion of bone marrow cells was performed in 33 patients with dilated cardiomyopathy (DCM) by using an over-the-wire balloon catheter. Three month follow-up demonstrated an improvement in left ventricular pump function while a modest improvement in Brain Natriuretic Peptide (BNP) levels was reported after 1 year [6]. Importantly, the conditions chosen in the present clinical trial were representative of different conditions (acute, chronic phase) encountered in the clinic. Unfortunately, no clear indication on stem cell characterization or on their actual ability to regenerate contractile cells is available. 3. TRACIA STUDY (Intracoronary Autologous Stem Cell Transplantation in ST Elevation Myocardial Infarction). The phase II/ III clinical trial aimed at evaluating the effects of intracoronary administration of adult stem cells on LV ejection fraction and major adverse cardiovascular events (MACE) after 6 months follow-up. For this reason, 1-2 million CD34+ cells were injected through the infarct-related artery few days after post-infarct angioplasty using an "over-the-wire" catheter in 80 patients aging from 20 to 75 years. The results of this study are still to be published. 4. Combined CABG and Stem-Cell Transplantation for Heart Failure. Intramyocardial delivery of autologous bone marrow cells extracted from iliac crest and purified by Ficoll centrifugation, during cardiac surgery for CABG intervention in 30 patients, as compared to 30 patients undergoing CABG without cell infusion. Although information on the number and characteristics of cells to be injected has not been given, the trial is currently ongoing and the follow-up is scheduled in 6-12 months (http://clinicaltrials.gov). 5. POSEIDON-Pilot Study (The Percutaneous Stem Cell Injection Delivery Effects on Neomyogenesis Pilot Study) Poseidon-pilot Study is a phase I/ II multi-center trial in which the trans-endocardial injection of autologous Mesenchymal Stem Cells (20-, 100-, 200 X 106) is compared to autologous non-purified bone marrow cells and to allogeneic human Mesenchymal Stem Cells. The implant is performed during cardiac catheterization using the Biocardia Helical Infusion Catheter in fifty (50) patients suffering from chronic ischemic left ventricular dysfunction secondary to myocardial infarction. The data collection is currently ongoing. 6. SCIPIO (Cardiac Stem Cell Infusion in Patients With Ischemic CardiOmyopathy). This phase I clinical trial is aimed at assessing the safety and effectiveness of intracoronary autologous cardiac stem cell therapy. As such, forty (40) patients suffering from ischemic cardiomyopathy are exposed to intracoronary injection of cardiac resident stem cells (CSC). Cardiac stem cells are harvested from right atrial appendages and selected for c-kit expression, cultured and expanded in vitro prior to injecting them via intracoronary route, three to five months after CABG surgery. The hypothesis is that CSC infused into nonviable myocardial segments will regenerate infarcted myocardium by differentiating into cardiomyocytes and vascular cells. The preliminary results are encouraging: in the nine patients treated at four months after
  • 45.
    Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site 29 CSC infusion, LVEF increased from 31.3 + 2.5 percent before CSC infusion to 38.8 + 3.2 percent four months after CSC infusion. Moreover, in the five patients in whom data are available at 12 months after stem cell infusion, the improvement in LVEF observed at four months was even greater, averaging 15% at 12 months. The follow-up is scheduled in 1,5 years. 7. ALCADIA (AutoLogous Human CArdiac-Derived Stem Cell to Treat Ischemic cArdiomyopathy). In this phase I, multicenter clinical trial, a rather different approach is followed. In fact, patients’ own cardiac stem cells obtained by endo-myocardial biopsies are delivered by a single intramyocardial injection. The cells injected are 0.5 million cells/kg (patient body weight) and their engraftment should be favored by the concomitant implantation of gelatin hydrogel sheet releasing human recombinant beta Fibroblast Growth Factor (bFGF), during CABG surgery. The study has been designed to treat refractory heart failure, ischemic cardiomyopathy or ventricular dysfunction cases. Importantly, this is the first clinical trial, to our knowledge, in which a human recombinant growth factor is used. Unfortunately, the number of enrolled patients is limited to six (6). 8. REGEN-IHD (Bone Marrow Derived Adult Stem Cells for Chronic Heart Failure). In this phase II/ III study, granulocyte-colony stimulating factor (G-CSF) is subcutaneously administered for 5 days to patients with heart failure secondary to ischemic heart disease to mobilize CD34+ bone marrow stem cells. A concomitant intracoronary or intramyocardial administration of bone marrow derived stem cells is performed. The number of enrolled patients is high (165) and the aim of the study is to compare the effects of G-CSF and autologous bone marrow progenitor cell infusion on the quality of life and left ventricular function in the patients. The follow-up timepoint is scheduled in 6-12 months. A number of papers reporting statistical analyses and comparisons among the clinical trials in which stem and progenitor cells have been adopted are currently available. [For further information, please refer to www.clinicaltrials.gov]. 5. Conclusions The possibility to treat cardiac diseases by cell therapy techniques is an extraordinary promise. While a number of different approaches has been so far proposed to setup minimally invasive techniques for cardiac repair, few of them being already in the clinical experimental phase, basic questions still need to be addressed. In fact, the molecular processes leading to cardiac differentiation still need to be fully clarified, while the impact of novel, genetically modified cell types obtained from adult differentiated cells on cardiac microenvironment deserve further investigations. More importantly, the seek to identify suitable delivery systems (i.e. scaffolds) able to foster stem cell survival, growth and differentiation, while degrading without negative effects as the formation of new tissue occurs is still open. A look at the literature reveals that an impressive effort to translate the information obtained by in vitro and pre-clinical studies to the bedside is being produced. In particular, a number of stem cell subsets, which have been previously tested in vitro and in animal models, are currently being tested in phase I, II clinical trials. As expected, the predominant delivery system used in the ongoing clinical trials is intracoronary or intramural injection of stem cells. The possibility to adopt tissue engineering techniques to
  • 46.
    Tissue Engineering forTissue and Organ Regeneration 30 design patient-specific cardiac substitutes containing synthetic or natural scaffolds is still far from being taken into consideration for clinical application, since any single formulation will have to be approved before clinical testing. 6. References [1] Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, Frisén J. Evidence for Cardiomyocyte Renewal in Humans. Science 2009: 324; 98-102. [2] Patel JK, Kobashigawa JA. Should we be doing routine biopsy after heart transplantation in a new era of anti-rejection? Curr Opin Cardiol 2006; 21: 127-131. [3] Hertz MI, Aurora P, Christie JD, Dobbels F, Edwards LB, Kirk R, Kucheryavaya AY, Rahmel AO, Rowe AW, Stehlik J, Taylor DO. Scientific Registry of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 2009: 28; 989- 1049. [4] American Heart Association. Heart disease and stroke statistics-2010 update. Dallas, Texas: American Heart Association; 2010. © 2010, American Heart Association. [5] Menasché P, Alfieri O, Janssens S, McKenna W, Reichenspurner H, Trinquart L, Vilquin JT, Marolleau JP, Seymour B, Larghero J, Lake S, Chatellier G, Solomon S, Desnos M, Hagège AA. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial: first randomized placebo-controlled study of myoblast transplantation. Circulation. 2008; 117: 1189-1200. [6] Fischer-Rasokat U, Assmus B, Assmus B, Seeger FH, Honold J, Leistner D, Fichtlscherer S, Schächinger V, Tonn T, Martin H, Dimmeler S, Zeiher MA. A pilot trial to assess potential effects of selective intracoronary bone marrow-derived progenitor cell infusion in patients with nonischemic dilated cardiomyopathy: final 1-year results of the transplantation of progenitor cells and functional regeneration enhancement pilot trial in patients with nonischemic dilated cardiomyopathy. Circ Heart Fail 2009; 2: 417-423. [7] Foudah D, Redaelli S, Donzelli E, Bentivegna A, Miloso M, Dalprà L, Tredici G. Monitoring the genomic stability of in vitro cultured rat bone-marrow-derived mesenchymal stem cells. Chromosome Res. 2009; 17: 1025-1039. [8] Momin EN, Vela G, Zaidi HA, Quiñones-Hinojosa A. The Oncogenic Potential of Mesenchymal Stem Cells in the Treatment of Cancer: Directions for Future Research. Curr Immunol Rev. 2010; 6:137-148 [9] Vacanti V, Kong E, Suzuki G, Sato K, Canty JM, Lee T. Phenotypic changes of adult porcine mesenchymal stem cells induced by prolonged passaging in culture. J Cell Physiol 2005: 194-201. [10] Itzhaki-Alfia A, Leor J, Raanani E, Sternik L, Spiegelstein D, Netser S, Holbova R, Pevsner-Fischer M, Lavee J, Barbash IM. Patient characteristics and cell source determine the number of isolated human cardiac progenitor cells. Circulation 2009; 120: 2559-2566. [11] Regulation (EC) No 1394/2007 of the European Parliament and of the Council of 13 November 2007 on advanced therapy medicinal products and amending Directive 2001/83/EC and Regulation (EC) No 726/2004. [12] Food and Drug Administration 21 CFR 1271 (2006).
  • 47.
    Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site 31 [13] Formigli L, Francini F, Tani A, Squecco R, Nosi D, Polidori L, Nistri S, Chiappini L, Cesati V, Pacini A, Perna AM, Orlandini GE, Zecchi Orlandini S, Bani D. Morphofunctional integration between skeletal myoblasts and adult cardiomyocytes in coculture is favoured by direct cell-cell contacts and relaxin treatment. Am J Physiol Cell Physiol 2005; 288: C795-804. [14] Hata H, Matsumiya G, Miyagawa S, Kondoh H, Kawaguchi N, Matsuura N, Shimizu T, Okano T, Matsuda H, Sawa H. Grafted skeletal myoblasts sheets attenuate myocardial remodelling in pacing-induced canine heart failure model. J Thorac Cardiovasc Surg 2009; 138: 460-467. [15] Reinecke H, Minami E, Poppa V, Murry CE. Evidence for fusion between cardiac and skeletal muscle cells. Circ Res 2004; 94: e56-e60. [16] Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature 2001; 410: 221-229. [17] Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad USA 2001; 98: 10344-10349 [18] Wagers AJ, Sherwood RI, Christensen JL, Weissman IL. Little evidence for developmental plasticity of adult hematopoietic stem cells. Science 2002; 297: 2256- 2259. [19] Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KB, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature. 2004; 428: 664-668. [20] Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ishaemic myocardium. Nature 2004; 428: 668-673. [21] Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor cells for angiogenesis. Science 1997; 275: 964-967. [22] Nesselmann C, Ma N, Bieback K, Wagner W, Ho A, Konttinen YT, Zhang H, Hinescu ME, Steinhoff G. Mesenchymal stem cells and cardiac repair. J Cell Mol Med 2008; 12: 1795-1810. [23] Amado L, Saliaris A, Schuleri K, St. John M, Xie JS, Cattaneo S, Durand DJ, Fitton T, Kuang JQ, Stewart G, Lehrke S, Baumgartner WW, Martin BJ, Heldman AW, Hare JM. Cardiac repair with intramyocardial injection of allogenic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci USA 2005; 102: 11474-11479. [24] Quaini, F., Urbanek, K., Beltrami, AP, Finato, N, Beltrami, CA, Nadal-Ginard, B, Kajstura, J, Leri, A, and Anversa, P. Chimerism of the transplanted heart. N Engl J Med 2002; 346: 5-15. [25] Bearzi C, Rota M, Hosoda T, Tillmanns J, Nascimbene A, De Angelis A, Yasuzawa- Amano S, Trofimova I, Siggins RW, Lecapitaine N, Cascapera S, Beltrami AP, D’Alessandro DA, Zias E, Quaini F, Urbanek K, Michler RE, Bolli R, Kajstura J, Leri A, Anversa P. Human cardiac stem cells. Proc Natl Acad Sci U S A. 2007; 104: 14068 – 14073.
  • 48.
    Tissue Engineering forTissue and Organ Regeneration 32 [26] Di Nardo P, Forte G, Ahluwalia A, Minieri M. Cardiac progenitor cells: Potency and control. J. Cell. Physiol. 2010; 224: 590–600. [27] Yamanaka S, Blau HM. Nuclear reprogramming to a pluripotent state by three approaches. Nature 2010; 465: 704-712. [28] Ieda M, Fu JD, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, Srivastava D. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010; 142: 375-386. [29] Perin EC and Lopez J. Methods in stem cell delivery in cardiac diseases. Nat Clinic Pract Cardiovasc Med 2006: 3 S1. [30] Dixon JA, Gorman RC, Stroud RE, Bouges S, Hirotsugu H, Gorman JH 3rd, Martens TP, Itescu S, Schuster MD, Plappert T, St John-Sutton MG, Spinale FG. Mesenchymal cell transplantation and myocardial remodeling after myocardial infarction. Circulation. 2009; 120: S220-S229. [31] Smits AM, van Vliet P, Metz CH, Korfage T, Sluijter JPG, Doevendans PA, Goumans MJ. Human cardiomyocyte progenitor cells differentiate into functional mature cardiomyocytes: an in vitro model for studying human cardiac physiology and pathophysiology. Nat Protoc 2009; 4: 232-243. [32] Rota M, Kajstura J, Hosoda T, Bearzi C, Vitale S, Esposito G, Iaffaldano G, Padin- Iruegas ME, Gonzalez A, Rizzi R, Small N, Muraski J, Alvarez R, Chen X, Urbanek K, Bolli R, Houser SR, Leri A, Sussman MA, Anversa P: Bone marrow cells adopt the cardiomyogenic fate in vivo. Proc Natl Acad Sci U S A 2007, 104: 17783-17788. [33] Gao J, Dennis JE, Muzic RF, Lundberg M, Caplan L. The dynamic in vivo distribution of bone marrow-derived mesenchymal stem cells after infusion. Cells Tissues Organs 2001; 169: 12-20. [34] Bui QT, Gertz ZM, Wilensky RL. Intracoronary delivery of bone-marrow-derived stem cells. Stem Cell Res Ther. 2010; 1:29-35. [35] Bartunek J, Vanderheyden M, Vandekerckhove B, Mansour S, De Bruyne B, De Bondt P, Van Haute I, Lootens N, Heyndrickx G, Wijns W. Intracoronary injection of CD133- positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction: feasibility and safety. Circulation 2005; 112: 178–183. [36] Singelyn JM, Christman KL. Injectable materials for the treatment of myocardial infarction and heart failure: the promise of decellularized matrices. J Cardiovasc Transl Res. 2010; 3: 478-486. [37] Menei P, Montero-Menei C, Venier MC, Benoit JP. Drug delivery into the brain using poly(lactide-co-glycolide) microspheres. Expert Opin Drug Deliv. 2005 2: 363-376. [38] Forte G, Franzese O, Pagliari S, Pagliari F, Cossa P, Laudisi A, Di Francesco AM, Fiaccavento R, Carotenuto F, Bonmassar E, Fiaccavento R, Minieri M, Di Nardo P. Interfacing Sca-1pos Mesenchymal Stem Cells with Biocompatible Scaffolds with Different Chemical Composition and Geometry. J Biomed Biotechnol 2009; doi: 10.1155/2009/910610. [39] Yu J, Du KT, Fang Q, Gu Y, Mihardja SS, Sievers RE, Wu JC, Lee RJ. The use of human mesenchymal stem cells encapsulated in RGD modified alginate microspheres in the repair of myocardial infarction in the rat. Biomaterials. 2010; 31: 7012-7020 [40] Mandoli C, Pagliari F, Pagliari S, Forte G, Di Nardo P, Licoccia S, Traversa E. Stem cell aligned growth induced by CeO2 nanoparticles in PLGA scaffolds with improved bioactivity for regenerative medicine. Adv Funct Mater 2010; 20: 1617-1624.
  • 49.
    Cardiac Muscle Engineering:Strategies to Deliver Stem Cells to the Damaged Site 33 [41] Zimmermann WH, Melnychenko I, Wasmeier G, Didié M, Naito H, Nixdorff U, Hess A, Budinsky L, Brune K, Michaelis B, Dhein S, Schwoerer A, Ehmke H, Eschenhagen T. Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nat Med. 2006; 12: 452-458. [42] Engelmayr GC Jr, Cheng M, Bettinger CJ, Borenstein JT, Langer R, Freed LE. Accordion- like honeycombs for tissue engineering of cardiac anisotropy. Nat Mater. 2008; 7: 1003-1010. [43] Chen QZ, Ishii H, Thouas GA, Lyon AR, Wright JS, Blaker JJ, Chrzanowski W, Boccaccini AR, Ali NN, Knowles JC, Harding SE. An elastomeric patch derived from poly(glycerol sebacate) for delivery of embryonic stem cells to the heart. Biomaterials. 2010; 31: 3885-3893. [44] Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006; 126: 677-689. [45] Forte G, Carotenuto F, Pagliari F, Pagliari S, Cossa P, Fiaccavento R, Ahluwalia A, Vozzi G, Vinci B, Serafino A, Rinaldi A, Traversa E, Carosella L, Minieri M, Di Nardo P. Criticatility of the biological and physical stimuli array inducing resident stem cell determination. Stem Cells 2008; 26: 2093-2103. [46] Pagliari S, Vilela-Silva AC, Forte G, Pagliari F, Mandoli C, Vozzi G, Pietronave S, Prat M, Licoccia S, Ahluwalia A, Traversa E, Minieri M, Di Nardo P. Cooperation of Biological and Mechanical Signals in Cardiac Progenitor Cell Differentiation. Adv Mater 2010; 23: 514-518. [47] Masuda S, Shimizu T, Yamato M, Okano T. Cell sheet engineering for heart tissue repair. Adv Drug Deliv Rev. 2008; 60: 277-285. [48] Miyahara Y, Nagaya N, Kataoka M, Yanagawa B, Tanaka K, Hao H, Ishino, Ishida H, Shimizu T, Kangawa K, Sano S, Okano T, Kitamura S, Mori H. Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat Med 2006; 12: 459-465. [49] Matsuura K, Honda A, Nagai T, Fukushima N, Iwanaga K, Tokunaga M, Shimizu T, Okano T, Kasanuki H, Hagiwara N, Komuro I. Transplantation of cardiac progenitor cells ameliorates cardiac dysfunction after myocardial infarction in mice. J Clin Invest. 2009; 119: 2204–2217. [50] Arauchi A, Shimizu T, Yamato M, Obara T, Okano T. Tissue-engineered thyroid cell sheet rescued hypothyroidism in rat models after receiving total thyroidectomy comparing with nontransplantation models. Tissue Eng Part A. 2009; 15: 3943-3949. [51] Zakharova L, Mastroeni D, Mutlu N, Molina M, Goldman S, Diethrich E, Gaballa MA. Transplantation of cardiac progenitor cell sheet onto infarcted heart promotes cardiogenesis and improves function. Cardiovasc Res. 2010; 87: 40-49. [52] Chen YR, Li Y, Chen L, Yang XC, Su PX, Cai J. The infarcted myocardium does not selectively promote embryonic stem cell differentiation into cardiomyocytes. Cardiovasc. Pathol. 2010; doi:10.1016/j.carpath.2009.12.003. [53] Menasché P. Stem cell therapy for heart failure: are arrhythmias a real safety concern? Circulation. 2009; 119:2735-2740. [54] Zenovich AG, Davis BH, Taylor DA. Comparison of intracardiac cell transplantation: autologous skeletal myoblasts versus bone marrow cells. Handb Exp Pharmacol. 2007; 180: 117-165.
  • 50.
    Tissue Engineering forTissue and Organ Regeneration 34 [55] Duckers HJ, Houtgraaf J, Hehrlein C, Schofer J, Waltenberger J, Gershlick A, Bartunek J, Nienaber C, Macaya C, Peters N, Smits P, Siminiak T, van Mieghem W, Legrand V, Serruys PW. Final results of a phase IIa, randomised, open-label trial to evaluate the percutaneous intramyocardial transplantation of autologous skeletal myoblasts in congestive heart failure patients: the SEISMIC trial. EuroIntervention. 2011; 6: 805- 812. [56] Assmus B, Fischer-Rasokat U, Honold J, Seeger FH, Fichtlscherer S, Tonn T, Seilfried E, Schaechinger V, Dimmeller S, Zeiher AM; TOPCARE-CHD Registry. Transcoronary transplantation of functionally competent BMCs is associated with a decrease in natriuretic peptide serum levels and improved survival of patients with chronic postinfarction heart failure: results of the TOPCARE-CHD Registry. Circ Res 2007; 100: 1234-1241. [57] Schaechinger V, Assmus B, Britten MB, Honold J, Lehmann R, Teupe C, Abolmaali ND, Vogl TJ, Hofmann WK, Martin H, Dimmeler S, Zeiher AM. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI trial. J Am Coll Cardiol 2004; 44: 1690-1698.
  • 51.
    3 Cardiovascular Tissue Engineering Basedon Fibrin-Gel-Scaffolds Stefan Jockenhoevel1 and Thomas C. Flanagan2 1Department of Tissue Engineering & Textile Implants, AME-Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, 2School of Medicine & Medical Science, Health Sciences Centre, University College Dublin, Dublin 1Germany 2Ireland 1. Introduction Cardiovascular disease is a major cause of death in the Western World. Novel drugs and innovative devices have enhanced the quality of life for patients with cardiovascular disease, but such treatments are not without limitations and complications. The major constraint with these current treatments is the inability for growth, repair and remodeling of the structure. The emergence of tissue engineering as an alternative therapy for cardiovascular disease has generated an intensity of research into the development of many components of the cardiovascular system, including heart valves, small-calibre vascular grafts and biological stent materials. The composition of the biomaterial used as a support for the developing cardiovascular structure is a key mediator of cell behaviour and function in the tissue, and the ideal scaffold biomaterial for development of a successful end-product continues to be a matter of debate. Fibrin, a major structural protein involved in wound healing, represents an ideal scaffold for the rapid synthesis of autologous tissue-engineered cardiovascular grafts, as its primary scaffold constituents (fibrinogen and thrombin) can be isolated directly from a blood sample of the patient requiring the graft. Fibrin gel scaffolds offer immediate high cell seeding efficiency and homogenous cell distribution by gelation entrapment, and have a degradation rate that can be controlled by protease inhibitors, e.g. tranexamic acid or aprotinin. Fibrin is also known to stimulate the secretion of reinforcing extracellular matrix (ECM) proteins by seeded cells. The potential to control the fibrin polymerisation process also offers the opportunity to produce complex 3-D structures, like heart valve prostheses and to embed porous, textile or metal (stent) structures. This book chapter reviews the properties of fibrin that make it an ideal scaffold candidate for applications in the area of cardiovascular tissue engineering, and documents the successful development of fibrin-based heart valves, vascular grafts and biostents for clinical application. 2. Scaffold materials Scaffolds play a central role in cardiovascular tissue engineering. Essential requirements for the ideal cardiovascular scaffold are easy handling properties and the ability to mould
  • 52.
    Tissue Engineering forTissue and Organ Regeneration 36 complex 3-D structures from the material, such as aortic roots or vessels with complex side branches. The scaffold material should neither be toxic, nor elicit any immunological side effects. The diffusion barrier of the scaffold material should have the lowest possible resistivity in order to guarantee an optimal nutrition supply in thicker tissues. Furthermore, both the mechanical and the chemical properties (e.g. the integration of growth factors) of the scaffold material should be modifiable. Controllable degradation of the material is also important in order to adapt the structural support of the scaffold with regard to the developing tissue. A multitude of scaffolds are currently employed in the field of tissue engineering, e.g. synthetic polymers (polyurethanes, polyglyolic acid, polylactic acid, polyhydroxybutyrate, copolymers of lactic and glycolic acids, polyanhydrides, polyorthoesters) and natural polymers (chitosan, glycosaminoglycans, collagen), or biological scaffolds such as acellularised porcine aortic conduits (Bader et al., 1998; Chevallay & Herbage, 2000; Flanagan et al., 2006; Freed et al., 1994; Grande et al., 1997). Scaffold-related problems including cytotoxic degradation products, fixed degradation times, limited mechanical properties and the absence of growth modulation, etc. necessitate further extensive investigations in developing the ideal cardiovascular scaffold. 3. Fibrin as scaffold material? Based on the assumption that successful tissue engineering should mimic the process of tissue regeneration, and that regeneration is closely related to haemostatis, fibrin (gel) seems to be an ideal candidate as a tissue engineering scaffold by virtue of its role as a “physiological scaffold” in tissue regeneration. Several influences of fibrin gel on tissue development have been described in the literature: it is known that fibrin gel is one of the major ligands for ß3 integrins, which leads to cell migration into a wound/tissue-engineered construct (Ikari et al., 2000; Nomura et al., 1999). Thrombin, fibrinogen, fibrin monomers and fibrinopepide B all increase DNA synthesis in smooth muscle cells (SMCs) and consequently the proliferation of the cell (Pakala et al., 2001). 3.1 Physiology of fibrin Fibrin is the end-product of the coagulation cascade following the conversion of fibrinogen in the presence of thrombin and calcium (Figure 1). Fibrinogen is a soluble plasma glycoprotein, which is produced by the liver. Fibrinogen is an acute phase protein with a normal blood concentration of 1.4 – 3.5 g/l. The fibrinogen molecule has a length of 45 nm, a molecular weight of 340 KDa and consists of 2 subunits and 3 polypeptides chains - α, β and γ. During the polymerisation process, the fibrinopeptide A of the α-chain and the fibrinopeptide B of the ß-chain are cleaved by thrombin. The exposed N-terminal positions of the fibrinopeptides bind to the γ-chain of the fibrinogen and produce the so-called proteofibrils. In the subsequent step, the lateral association leads to apposition of the proteofibrils to form a 3-D fibrin network structure (Meyer, 2004). FXIIIa stabilises fibrin further by incorporation of the fibrinolysis receptors, alpha-2-antiplasmin and TAFI (thrombin activatable fibrinolysis inhibitor, procarboxypeptidase B), and binding to several adhesive proteins of various cells (Muszbek et al., 2008). The polymerised fibrin gel matrix is a hydro-gel, which contains ~95-98% water. The water content can easily be exchanged against a buffer solution or a cell culture medium, allowing an optimal nutrition supply of the cells that are embedded after the gelation process.
  • 53.
    Cardiovascular Tissue EngineeringBased on Fibrin-Gel-Scaffolds 37 Fig. 1. Coagulation cascade: the conversion of fibrinogen into fibrin is triggered by thrombin and calcium 3.2 Production of autologous fibrin The classical approach for production of autologous fibrin is the cryoprecipitation method: after the production of platelet-poor plasma (PPP), the plasma is frozen at -80°C and thawed overnight at +4°C. The precipitate formed contains ~60-70% of fibrinogen. After centrifugation, the supernatant is decanted and the precipitate is subsequently washed twice in rinsed water. After the precipitate is dissolved in water, overnight dialysis against calcium-free TRIS buffer solution is necessary to provide optimal conditions for the embedded cells. The cryoprecipitation method has two major disadvantages: (1) the efficiency of fibrinogen isolation is relatively low with only ~20-25% of the total fibrinogen content removed, and (2) the production process is time-consuming (~2 days). The low isolation efficiency is particularly problematic regarding the use of autologous fibrin gel scaffolds in paediatric
  • 54.
    Tissue Engineering forTissue and Organ Regeneration 38 patients, as the volume of sampled blood needs to be kept to an absolute minimum. Therefore, the use of alternative precipitation methods with different chemicals has been evaluated: (1) ethanol (Kjaergard et al., 1992; Weis-Fogh, 1988), (2) ammonium sulphate alone, and (3) in combination with the cryoprecipitation method (Wolf, 1983), (4) albumin plus cryoprecipitation, and (5) polyethylene glycol (PEG) plus cryoprecipitation (Epstein et al., 1986). Heselhaus investigated each of these different precipitation methods with regard to their efficiency and their use in the development of fibrin scaffold materials for cardiovascular applications (Heselhaus, 2011): Fig. 2. [A] Fibrin gel after polymerisation in a 6-well plate. [B-D] Scanning electron microscopy (SEM) images demonstrating the nano-fibre network structure of the fibrin, which enables the gentle embedding of cells, with a vascular smooth muscle cell (SMC) shown in [D] immediately after the gelation of the fibrin within a web-like network surrounded by cell culture medium Figure 3 demonstrates that all of the reported alternative methods are more efficient than the standard cryoprecipitation method. Here, the technique using ethanol as the precipitation reagent is observed as the most efficient method, with an isolation efficiency of ~80%, ~4-times higher than the efficiency of the standard method. The technique applying both albumin and cryoprecipitation indicates a false positive high result due to contamination of the precipitate with albumin (shown by a significant band in SDS gel electrophoreses) (Heselhaus, 2011).
  • 55.
    Another Random ScribdDocument with Unrelated Content
  • 56.
    TARTALOM. VII. Az amásik 3 VIII. Boszu, a maga módja szerint 56 IX. A két fogoly 76 X. Különböző fogadtatások 131 XI. Az a bizonyos szegény asszony 156 Javítások. Az eredeti szöveg helyesírásán nem változtattunk. A nyomdai hibákat javítottuk. Ezek listája: 14 az kérdezné azt kérdezné 34 Maxenphutsch urat Maxenpfutsch urat 37 a bagoly! a bagoly!“ 42 „Megitati“ csárdában „Megitató“ csárdában 53 hol gyermekrée hol gyermekére 60 gyanuja a Garamvölgyi gyanuja a Garanvölgyi 65 knyszeritnének, hogy kényszeritnének, hogy 74 hogy kit? hogy kit?“ 79 Ezt azt utolsó Ezt az utolsó 80 nagy htrtelen nagy hirtelen 83 Hah! Az égre Ne Hah! Az égre! Ne 110 ön elfo-fogadta ön elfogadta 124 kocsis perse kocsis persze 125 megitató csárdánál Megitató csárdánál 125 szaladt egyesen szaladt egyenesen 125 kisszony meg nem kisasszony meg nem
  • 57.
    126 Hermin afedett Eliz a fedett 127 tudott dükösködni tudott dühösködni 129 ne történjék! ne történjék!“ 131 IX. Különböző fogadtatások X. Különböző fogadtatások 131 lehessen vvnni lehessen venni 133 egyib jobbról egyik jobbról 136 Bizonyosan tévedész Bizonyosan tévedés 141 a ha-hazajövetelét a hazajövetelét 146 Önt azt mondta Ön azt mondta 148 jövök jöbbet jövök többet 156 olyan azszonyra olyan asszonyra 159 ki iudott ki tudott 167 már többet. már többet.“ 170 arrál álmodik arról álmodik 171 megasszony fog lenni menyasszony fog lenni 173 meg nem gyulnék meg nem gyógyulnék
  • 58.
    *** END OFTHE PROJECT GUTENBERG EBOOK AZ UJ FÖLDESUR (2. KÖTET) *** Updated editions will replace the previous one—the old editions will be renamed. Creating the works from print editions not protected by U.S. copyright law means that no one owns a United States copyright in these works, so the Foundation (and you!) can copy and distribute it in the United States without permission and without paying copyright royalties. Special rules, set forth in the General Terms of Use part of this license, apply to copying and distributing Project Gutenberg™ electronic works to protect the PROJECT GUTENBERG™ concept and trademark. Project Gutenberg is a registered trademark, and may not be used if you charge for an eBook, except by following the terms of the trademark license, including paying royalties for use of the Project Gutenberg trademark. If you do not charge anything for copies of this eBook, complying with the trademark license is very easy. You may use this eBook for nearly any purpose such as creation of derivative works, reports, performances and research. Project Gutenberg eBooks may be modified and printed and given away—you may do practically ANYTHING in the United States with eBooks not protected by U.S. copyright law. Redistribution is subject to the trademark license, especially commercial redistribution. START: FULL LICENSE
  • 59.
    THE FULL PROJECTGUTENBERG LICENSE
  • 60.
    PLEASE READ THISBEFORE YOU DISTRIBUTE OR USE THIS WORK To protect the Project Gutenberg™ mission of promoting the free distribution of electronic works, by using or distributing this work (or any other work associated in any way with the phrase “Project Gutenberg”), you agree to comply with all the terms of the Full Project Gutenberg™ License available with this file or online at www.gutenberg.org/license. Section 1. General Terms of Use and Redistributing Project Gutenberg™ electronic works 1.A. By reading or using any part of this Project Gutenberg™ electronic work, you indicate that you have read, understand, agree to and accept all the terms of this license and intellectual property (trademark/copyright) agreement. If you do not agree to abide by all the terms of this agreement, you must cease using and return or destroy all copies of Project Gutenberg™ electronic works in your possession. If you paid a fee for obtaining a copy of or access to a Project Gutenberg™ electronic work and you do not agree to be bound by the terms of this agreement, you may obtain a refund from the person or entity to whom you paid the fee as set forth in paragraph 1.E.8. 1.B. “Project Gutenberg” is a registered trademark. It may only be used on or associated in any way with an electronic work by people who agree to be bound by the terms of this agreement. There are a few things that you can do with most Project Gutenberg™ electronic works even without complying with the full terms of this agreement. See paragraph 1.C below. There are a lot of things you can do with Project Gutenberg™ electronic works if you follow the terms of this agreement and help preserve free future access to Project Gutenberg™ electronic works. See paragraph 1.E below.
  • 61.
    1.C. The ProjectGutenberg Literary Archive Foundation (“the Foundation” or PGLAF), owns a compilation copyright in the collection of Project Gutenberg™ electronic works. Nearly all the individual works in the collection are in the public domain in the United States. If an individual work is unprotected by copyright law in the United States and you are located in the United States, we do not claim a right to prevent you from copying, distributing, performing, displaying or creating derivative works based on the work as long as all references to Project Gutenberg are removed. Of course, we hope that you will support the Project Gutenberg™ mission of promoting free access to electronic works by freely sharing Project Gutenberg™ works in compliance with the terms of this agreement for keeping the Project Gutenberg™ name associated with the work. You can easily comply with the terms of this agreement by keeping this work in the same format with its attached full Project Gutenberg™ License when you share it without charge with others. 1.D. The copyright laws of the place where you are located also govern what you can do with this work. Copyright laws in most countries are in a constant state of change. If you are outside the United States, check the laws of your country in addition to the terms of this agreement before downloading, copying, displaying, performing, distributing or creating derivative works based on this work or any other Project Gutenberg™ work. The Foundation makes no representations concerning the copyright status of any work in any country other than the United States. 1.E. Unless you have removed all references to Project Gutenberg: 1.E.1. The following sentence, with active links to, or other immediate access to, the full Project Gutenberg™ License must appear prominently whenever any copy of a Project Gutenberg™ work (any work on which the phrase “Project Gutenberg” appears, or with which the phrase “Project Gutenberg” is associated) is accessed, displayed, performed, viewed, copied or distributed:
  • 62.
    This eBook isfor the use of anyone anywhere in the United States and most other parts of the world at no cost and with almost no restrictions whatsoever. You may copy it, give it away or re-use it under the terms of the Project Gutenberg License included with this eBook or online at www.gutenberg.org. If you are not located in the United States, you will have to check the laws of the country where you are located before using this eBook. 1.E.2. If an individual Project Gutenberg™ electronic work is derived from texts not protected by U.S. copyright law (does not contain a notice indicating that it is posted with permission of the copyright holder), the work can be copied and distributed to anyone in the United States without paying any fees or charges. If you are redistributing or providing access to a work with the phrase “Project Gutenberg” associated with or appearing on the work, you must comply either with the requirements of paragraphs 1.E.1 through 1.E.7 or obtain permission for the use of the work and the Project Gutenberg™ trademark as set forth in paragraphs 1.E.8 or 1.E.9. 1.E.3. If an individual Project Gutenberg™ electronic work is posted with the permission of the copyright holder, your use and distribution must comply with both paragraphs 1.E.1 through 1.E.7 and any additional terms imposed by the copyright holder. Additional terms will be linked to the Project Gutenberg™ License for all works posted with the permission of the copyright holder found at the beginning of this work. 1.E.4. Do not unlink or detach or remove the full Project Gutenberg™ License terms from this work, or any files containing a part of this work or any other work associated with Project Gutenberg™. 1.E.5. Do not copy, display, perform, distribute or redistribute this electronic work, or any part of this electronic work, without prominently displaying the sentence set forth in paragraph 1.E.1
  • 63.
    with active linksor immediate access to the full terms of the Project Gutenberg™ License. 1.E.6. You may convert to and distribute this work in any binary, compressed, marked up, nonproprietary or proprietary form, including any word processing or hypertext form. However, if you provide access to or distribute copies of a Project Gutenberg™ work in a format other than “Plain Vanilla ASCII” or other format used in the official version posted on the official Project Gutenberg™ website (www.gutenberg.org), you must, at no additional cost, fee or expense to the user, provide a copy, a means of exporting a copy, or a means of obtaining a copy upon request, of the work in its original “Plain Vanilla ASCII” or other form. Any alternate format must include the full Project Gutenberg™ License as specified in paragraph 1.E.1. 1.E.7. Do not charge a fee for access to, viewing, displaying, performing, copying or distributing any Project Gutenberg™ works unless you comply with paragraph 1.E.8 or 1.E.9. 1.E.8. You may charge a reasonable fee for copies of or providing access to or distributing Project Gutenberg™ electronic works provided that: • You pay a royalty fee of 20% of the gross profits you derive from the use of Project Gutenberg™ works calculated using the method you already use to calculate your applicable taxes. The fee is owed to the owner of the Project Gutenberg™ trademark, but he has agreed to donate royalties under this paragraph to the Project Gutenberg Literary Archive Foundation. Royalty payments must be paid within 60 days following each date on which you prepare (or are legally required to prepare) your periodic tax returns. Royalty payments should be clearly marked as such and sent to the Project Gutenberg Literary Archive Foundation at the address specified in Section 4, “Information
  • 64.
    about donations tothe Project Gutenberg Literary Archive Foundation.” • You provide a full refund of any money paid by a user who notifies you in writing (or by e-mail) within 30 days of receipt that s/he does not agree to the terms of the full Project Gutenberg™ License. You must require such a user to return or destroy all copies of the works possessed in a physical medium and discontinue all use of and all access to other copies of Project Gutenberg™ works. • You provide, in accordance with paragraph 1.F.3, a full refund of any money paid for a work or a replacement copy, if a defect in the electronic work is discovered and reported to you within 90 days of receipt of the work. • You comply with all other terms of this agreement for free distribution of Project Gutenberg™ works. 1.E.9. If you wish to charge a fee or distribute a Project Gutenberg™ electronic work or group of works on different terms than are set forth in this agreement, you must obtain permission in writing from the Project Gutenberg Literary Archive Foundation, the manager of the Project Gutenberg™ trademark. Contact the Foundation as set forth in Section 3 below. 1.F. 1.F.1. Project Gutenberg volunteers and employees expend considerable effort to identify, do copyright research on, transcribe and proofread works not protected by U.S. copyright law in creating the Project Gutenberg™ collection. Despite these efforts, Project Gutenberg™ electronic works, and the medium on which they may be stored, may contain “Defects,” such as, but not limited to, incomplete, inaccurate or corrupt data, transcription errors, a copyright or other intellectual property infringement, a defective or
  • 65.
    damaged disk orother medium, a computer virus, or computer codes that damage or cannot be read by your equipment. 1.F.2. LIMITED WARRANTY, DISCLAIMER OF DAMAGES - Except for the “Right of Replacement or Refund” described in paragraph 1.F.3, the Project Gutenberg Literary Archive Foundation, the owner of the Project Gutenberg™ trademark, and any other party distributing a Project Gutenberg™ electronic work under this agreement, disclaim all liability to you for damages, costs and expenses, including legal fees. YOU AGREE THAT YOU HAVE NO REMEDIES FOR NEGLIGENCE, STRICT LIABILITY, BREACH OF WARRANTY OR BREACH OF CONTRACT EXCEPT THOSE PROVIDED IN PARAGRAPH 1.F.3. YOU AGREE THAT THE FOUNDATION, THE TRADEMARK OWNER, AND ANY DISTRIBUTOR UNDER THIS AGREEMENT WILL NOT BE LIABLE TO YOU FOR ACTUAL, DIRECT, INDIRECT, CONSEQUENTIAL, PUNITIVE OR INCIDENTAL DAMAGES EVEN IF YOU GIVE NOTICE OF THE POSSIBILITY OF SUCH DAMAGE. 1.F.3. LIMITED RIGHT OF REPLACEMENT OR REFUND - If you discover a defect in this electronic work within 90 days of receiving it, you can receive a refund of the money (if any) you paid for it by sending a written explanation to the person you received the work from. If you received the work on a physical medium, you must return the medium with your written explanation. The person or entity that provided you with the defective work may elect to provide a replacement copy in lieu of a refund. If you received the work electronically, the person or entity providing it to you may choose to give you a second opportunity to receive the work electronically in lieu of a refund. If the second copy is also defective, you may demand a refund in writing without further opportunities to fix the problem. 1.F.4. Except for the limited right of replacement or refund set forth in paragraph 1.F.3, this work is provided to you ‘AS-IS’, WITH NO OTHER WARRANTIES OF ANY KIND, EXPRESS OR IMPLIED,
  • 66.
    INCLUDING BUT NOTLIMITED TO WARRANTIES OF MERCHANTABILITY OR FITNESS FOR ANY PURPOSE. 1.F.5. Some states do not allow disclaimers of certain implied warranties or the exclusion or limitation of certain types of damages. If any disclaimer or limitation set forth in this agreement violates the law of the state applicable to this agreement, the agreement shall be interpreted to make the maximum disclaimer or limitation permitted by the applicable state law. The invalidity or unenforceability of any provision of this agreement shall not void the remaining provisions. 1.F.6. INDEMNITY - You agree to indemnify and hold the Foundation, the trademark owner, any agent or employee of the Foundation, anyone providing copies of Project Gutenberg™ electronic works in accordance with this agreement, and any volunteers associated with the production, promotion and distribution of Project Gutenberg™ electronic works, harmless from all liability, costs and expenses, including legal fees, that arise directly or indirectly from any of the following which you do or cause to occur: (a) distribution of this or any Project Gutenberg™ work, (b) alteration, modification, or additions or deletions to any Project Gutenberg™ work, and (c) any Defect you cause. Section 2. Information about the Mission of Project Gutenberg™ Project Gutenberg™ is synonymous with the free distribution of electronic works in formats readable by the widest variety of computers including obsolete, old, middle-aged and new computers. It exists because of the efforts of hundreds of volunteers and donations from people in all walks of life. Volunteers and financial support to provide volunteers with the assistance they need are critical to reaching Project Gutenberg™’s goals and ensuring that the Project Gutenberg™ collection will
  • 67.
    remain freely availablefor generations to come. In 2001, the Project Gutenberg Literary Archive Foundation was created to provide a secure and permanent future for Project Gutenberg™ and future generations. To learn more about the Project Gutenberg Literary Archive Foundation and how your efforts and donations can help, see Sections 3 and 4 and the Foundation information page at www.gutenberg.org. Section 3. Information about the Project Gutenberg Literary Archive Foundation The Project Gutenberg Literary Archive Foundation is a non-profit 501(c)(3) educational corporation organized under the laws of the state of Mississippi and granted tax exempt status by the Internal Revenue Service. The Foundation’s EIN or federal tax identification number is 64-6221541. Contributions to the Project Gutenberg Literary Archive Foundation are tax deductible to the full extent permitted by U.S. federal laws and your state’s laws. The Foundation’s business office is located at 809 North 1500 West, Salt Lake City, UT 84116, (801) 596-1887. Email contact links and up to date contact information can be found at the Foundation’s website and official page at www.gutenberg.org/contact Section 4. Information about Donations to the Project Gutenberg Literary Archive Foundation Project Gutenberg™ depends upon and cannot survive without widespread public support and donations to carry out its mission of increasing the number of public domain and licensed works that can be freely distributed in machine-readable form accessible by the widest array of equipment including outdated equipment. Many
  • 68.
    small donations ($1to $5,000) are particularly important to maintaining tax exempt status with the IRS. The Foundation is committed to complying with the laws regulating charities and charitable donations in all 50 states of the United States. Compliance requirements are not uniform and it takes a considerable effort, much paperwork and many fees to meet and keep up with these requirements. We do not solicit donations in locations where we have not received written confirmation of compliance. To SEND DONATIONS or determine the status of compliance for any particular state visit www.gutenberg.org/donate. While we cannot and do not solicit contributions from states where we have not met the solicitation requirements, we know of no prohibition against accepting unsolicited donations from donors in such states who approach us with offers to donate. International donations are gratefully accepted, but we cannot make any statements concerning tax treatment of donations received from outside the United States. U.S. laws alone swamp our small staff. Please check the Project Gutenberg web pages for current donation methods and addresses. Donations are accepted in a number of other ways including checks, online payments and credit card donations. To donate, please visit: www.gutenberg.org/donate. Section 5. General Information About Project Gutenberg™ electronic works Professor Michael S. Hart was the originator of the Project Gutenberg™ concept of a library of electronic works that could be freely shared with anyone. For forty years, he produced and distributed Project Gutenberg™ eBooks with only a loose network of volunteer support.
  • 69.
    Project Gutenberg™ eBooksare often created from several printed editions, all of which are confirmed as not protected by copyright in the U.S. unless a copyright notice is included. Thus, we do not necessarily keep eBooks in compliance with any particular paper edition. Most people start at our website which has the main PG search facility: www.gutenberg.org. This website includes information about Project Gutenberg™, including how to make donations to the Project Gutenberg Literary Archive Foundation, how to help produce our new eBooks, and how to subscribe to our email newsletter to hear about new eBooks.
  • 70.
    Welcome to ourwebsite – the perfect destination for book lovers and knowledge seekers. We believe that every book holds a new world, offering opportunities for learning, discovery, and personal growth. That’s why we are dedicated to bringing you a diverse collection of books, ranging from classic literature and specialized publications to self-development guides and children's books. More than just a book-buying platform, we strive to be a bridge connecting you with timeless cultural and intellectual values. With an elegant, user-friendly interface and a smart search system, you can quickly find the books that best suit your interests. Additionally, our special promotions and home delivery services help you save time and fully enjoy the joy of reading. Join us on a journey of knowledge exploration, passion nurturing, and personal growth every day! ebookbell.com