You are on page 1of 67

Principles of Tissue Engineering -

eBook PDF
Visit to download the full and correct content document:
https://ebooksecure.com/download/principles-of-tissue-engineering-ebook-pdf/
Principles of Tissue Engineering
Principles of Tissue
Engineering
Fifth Edition

Edited by
Robert Lanza
Astellas Institute for Regenerative Medicine, Westborough, MA, United States;
Institute for Regenerative Medicine, Wake Forest University School of Medicine,
Winston-Salem, NC, United States

Robert Langer
Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology,
Cambridge, MA, United States

Joseph P. Vacanti
Harvard Medical School, Center for Regenerative Medicine,
Massachusetts General Hospital, Cambridge, MA, United States

Anthony Atala
Wake Forest Institute for Regenerative Medicine,
Wake Forest School of Medicine, Winston-Salem, NC, United States
Academic Press is an imprint of Elsevier
125 London Wall, London EC2Y 5AS, United Kingdom
525 B Street, Suite 1650, San Diego, CA 92101, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
Copyright © 2020 Elsevier Inc. All rights reserved.
No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical,
including photocopying, recording, or any information storage and retrieval system, without permission in writing from
the publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our
arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be
found at our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as
may be noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our
understanding, changes in research methods, professional practices, or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any
information, methods, compounds, or experiments described herein. In using such information or methods they should
be mindful of their own safety and the safety of others, including parties for whom they have a professional
responsibility.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for
any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any
use or operation of any methods, products, instructions, or ideas contained in the material herein.
British Library Cataloguing-in-Publication Data
A catalogue record for this book is available from the British Library
Library of Congress Cataloging-in-Publication Data
A catalog record for this book is available from the Library of Congress
ISBN: 978-0-12-818422-6

For Information on all Academic Press publications


visit our website at https://www.elsevier.com/books-and-journals

Publisher: Andre Gerhard Wolff


Acquisitions Editor: Elizabeth Brown
Editorial Project Manager: Pat Gonzalez
Production Project Manager: Sreejith Viswanathan
Cover Designer: Miles Hitchen
Typeset by MPS Limited, Chennai, India
Contents

List of contributors xxix Variability in stem cell populations 40


Preface xli Modeling tissue growth and development 41
Monolayer tissue growth in vitro 42
1. Tissue engineering: current status Tissue growth on complex surfaces in vitro 42
and future perspectives 1 Three-dimensional tissue growth in vitro 43
Pattern formation 44
Prafulla K. Chandra, Shay Soker and Anthony Atala Machine learning in tissue engineering 45
Clinical need 1 Supervised methods 46
Current state of the field 1 Unsupervised methods 46
Smart biomaterials 2 Machine learning of cellular dynamics 47
Cell sources 4 Regulatory network inference 47
Whole organ engineering 8 From mathematical models to clinical reality 47
Biofabrication technologies 9 References 48
Electrospinning 9
Inkjet three-dimensional bioprinting 12 3. Moving into the clinic 53
Extrusion three-dimensional bioprinting 12
Chi Lo, Darren Hickerson, James J. Yoo,
Spheroids and organoids 13
Anthony Atala and Julie Allickson
Imaging technologies 16
Tissue neovascularization 16 Introduction 53
Bioreactors 16 Current state of tissue engineering 53
Organ-on-a-chip and body-on-a-chip 17 Pathway for clinical translation 54
Integration of nanotechnology 18 Regulatory considerations for tissue
Current challenges 19 engineering 58
Future directions 21 Conclusion 60
Smart biomaterials 21 Acknowledgment 60
Cell sources 22 References 60
Whole organ engineering 24 Further reading 60
Biofabrication technologies 24
Tissue neovasculatization 25
Bioreactors 25
Part One
Integration of nanotechnology 25 The basis of growth and
Conclusions and future challenges 26 differentiation 63
References 26
Further reading 35 4. Molecular biology of the cell 65
2. From mathematical modeling and J.M.W. Slack
machine learning to clinical reality 37 The cell nucleus 65
Ben D. MacArthur, Patrick S. Stumpf and Control of gene expression 66
Richard O.C. Oreffo Transcription factors 67
Other controls of gene activity 67
Introduction 37 The cytoplasm 68
Modeling stem cell dynamics 37 The cytoskeleton 69
Positive feedback based molecular switches 38 The cell surface 71

v
vi Contents

Cell adhesion molecules 71 Signal transduction events during


Extracellular matrix 72 cell extracellular matrix interactions 104
Signal transduction 73 Relevance for tissue engineering 111
Growth and death 74 Avoiding a strong immune response that
Culture media 75 can cause chronic inflammation and/or
Cells in tissues and organs 76 rejection 111
Cell types 76 Creating the proper substrate for cell survival
Tissues 77 and differentiation 111
Organs 77 Providing the appropriate environmental
Reference 78 conditions for tissue maintenance 112
Further reading 78 References 113

5. Molecular organization of cells 79 7. Matrix molecules and their ligands 119


Jon D. Ahlstrom Allison P. Drain and Valerie M. Weaver
Introduction 79 Introduction 119
Molecules that organize cells 79 Collagens 120
Changes in cell cell adhesion 80 Fibrillar collagens 121
Changes in celleextracellular matrix adhesion 80 Fibril-associated collagens with interrupted
Changes in cell polarity and stimulation of cell triple helices (FACIT) 122
motility 81 Basement membrane associated collagens 123
Invasion of the basal lamina 81 Other collagens 123
The epithelial mesenchymal transition Major adhesive glycoproteins 123
transcriptional program 82 Fibronectin 123
Transcription factors that regulate Laminin 125
epithelial mesenchymal transition 82 Elastic fibers and microfibrils 126
Regulation at the promoter level 82 Other adhesive glycoproteins and
Posttranscriptional regulation of multifunctional matricellular proteins 126
epithelial mesenchymal transition Vitronectin 126
transcription factors 83 Thrombospondins 126
Molecular control of the Tenascins 126
epithelial mesenchymal transition 83 Proteoglycans 127
Ligand-receptor signaling 83 Hyaluronan and lecticans 127
Additional signaling pathways 85 Perlecan 128
A model for epithelial mesenchymal transition Small leucine-rich repeat proteoglycans and
induction 85 syndecans 128
Conclusion 86 Conclusion 128
List of acronyms and abbreviations 86 References 128
Glossary 86
References 87 8. Morphogenesis and tissue
engineering 133
6. The dynamics of cell extracellular
Priscilla S. Briquez and Jeffrey A. Hubbell
matrix interactions, with implications
for tissue engineering 93 Introduction to tissue morphogenesis 133
Biology of tissue morphogenesis 133
M. Petreaca and M. Martins-Green
Morphogens as bioactive signaling molecules
Introduction 93 during morphogenesis 134
Historical background 93 The extracellular matrix as a key regulator
Extracellular matrix composition 93 of tissue morphogenesis 135
Receptors for extracellular matrix molecules 94 Cell cell interactions during tissue
Cell extracellular matrix interactions 96 morphogenesis 136
Development 96 Tissues as integrated systems in the body 136
Wound healing 100 Engineering tissue morphogenesis 138
Contents vii

Cells as building units in tissue engineering 138 Culture duration 161


Biomaterial scaffolds as artificial extracellular Biomaterials 162
matrices 139 Bioreactors and growth factors 166
Morphogens as signaling cues in tissue Bioreactors and mechanical forces 169
engineering 140 Conclusion 171
Tissue remodeling in healthy and diseased Acknowledgments 172
environments 140 References 172
Current focuses and future challenges 141 Further reading 177
References 141
11. Principles of bioreactor design for
9. Gene expression, cell determination, tissue engineering 179
differentiation, and regeneration 145 Hanry Yu, Seow Khoon Chong,
Frank E. Stockdale Ammar Mansoor Hassanbhai, Yao Teng,
Gowri Balachander, Padmalosini Muthukumaran,
Introduction 145
Feng Wen and Swee Hin Teoh
Determination and differentiation 145
MyoD and the myogenic regulatory factors 147 Introduction 179
Negative regulators of development 148 Macrobioreactors 180
MicroRNAs—regulators of differentiation 148 Design principles 181
Pax in development 149 Sustainable bioreactors 188
Satellite cells in skeletal muscle differentiation and Cell manufacturing quality attributes and
repair 149 process analytics technology 189
Tissue engineering—repairing muscle and fostering Future outlook 189
regeneration by controlling determination and Microbioreactors 191
differentiation 150 Design principles 191
Conclusion 152 Types of microreactors 194
References 152 Components and integration into
microreactors 194
Applications 195
Part Two Summary 197
Acknowledgments 197
In vitro control of tissue
References 197
development 155
12. Regulation of cell behavior by
10. Engineering functional tissues:
extracellular proteins 205
in vitro culture parameters 157
Amy D. Bradshaw
Jennifer J. Bara and Farshid Guilak
Introduction 205
Introduction 157
Thrombospondin-1 205
Key concepts for engineering functional
Thrombospondin-2 207
tissues 158
Tenascin-C 208
Fundamental parameters for engineering
Osteopontin 209
functional tissues 158
Secreted protein acidic and rich in cysteine 210
Fundamental criteria for engineering
Conclusion 212
functional tissues 159
References 212
Importance of in vitro studies for engineering
functional tissues 159
In vitro studies relevant to tissue engineering
13. Cell and matrix dynamics in
and regenerative medicine 159 branching morphogenesis 217
In vitro platforms relevant for high throughput Shaimar R. González Morales and
screening of drugs and other agents 160 Kenneth M. Yamada
Influence of selected in vitro culture
parameters on the development and Introduction 217
performance of engineered tissues 161 The basis of branching morphogenesis 217
viii Contents

Branching morphogenesis in the lung 218 Part Three


Branching morphogenesis in the
In Vivo Synthesis of Tissues and
salivary gland 220
Branching morphogenesis in the kidney 222 Organs 257
Contributions of other cell types 224
MicroRNAs in branching morphogenesis 225 15. In vivo engineering of organs 259
Extracellular matrix components in V. Prasad Shastri
branching morphogenesis 226
Laminin 226 Introduction 259
Collagen 226 Historical context 259
Heparan sulfate proteoglycan 227 Nature’s approach to cellular differentiation
Fibronectin and integrins 228 and organization 260
Basement membrane microperforations 228 Conceptual framework of the in vivo
Mathematical and computational bioreactor 261
models 230 In vivo bone engineering—the bone
Geometry 230 bioreactor 261
Mechanical forces 230 In vivo cartilage engineering 264
Signaling mechanisms 230 Induction of angiogenesis using biophysical
Conclusion 231 cues—organotypic vasculature engineering 265
Acknowledgments 232 De novo liver engineering 267
References 232 Repairing brain tissue through controlled
induction of reactive astrocytes 269
Conclusions and outlook 269
14. Mechanobiology, tissue References 270
development, and tissue
engineering 237
David Li and Yu-li Wang Part Four
Introduction 237
Biomaterials in tissue
Mechanical forces in biological systems 237 engineering 273
Tension 237
Compression 238 16. Cell interactions with polymers 275
Fluid shear 238 W. Mark Saltzman and Themis R. Kyriakides
Cellular mechanosensing 238
The cytoskeleton 239 Methods for characterizing cell interactions
Stretch-activated ion channels 239 with polymers 275
Cell cell adhesions 240 In vitro cell culture methods 275
Cell substrate adhesions 240 In vivo methods 278
The extracellular matrix 241 Cell interactions with polymers 280
Cellular effects of mechanotransduction 243 Protein adsorption to polymers 280
Substrate adhesion, spreading, and Effect of polymer chemistry on cell behavior 280
migration 243 Electrically charged or electrically conducting
Cell cell interactions in collectives 243 polymers 284
Proliferation and differentiation 244 Influence of surface morphology on cell
Mechanotransduction in biological behavior 284
phenomena 245 Use of patterned surfaces to control cell
Wound healing 245 behavior 285
Tissue morphogenesis 247 Cell interactions with polymers in suspension 286
Cancer metastasis 248 Cell interactions with three-dimensional
Mechanobiology in tissue engineering 248 polymer scaffolds and gels 287
Bone-implant design 248 Cell interactions unique to the in vivo setting 287
Organs-on-a-chip 250 Inflammation 287
References 252 Fibrosis and angiogenesis 288
References 289
Contents ix

17. Polymer scaffold fabrication 295 Combinations (hybrids) of synthetic and


biologically derived polymers 333
Matthew L. Bedell, Jason L. Guo, Using polymers to create tissue-engineered
Virginia Y. Xie, Adam M. Navara and products 333
Antonios G. Mikos Barriers: membranes and tubes 334
Introduction 295 Gels 334
Design inputs: materials, processing, and cell Matrices 334
types 297 Conclusion 335
Materials and inks 297 References 335
Processing and cell viability 299
Cell types and biological interactions 300 19. Three-dimensional scaffolds 343
Assessment of cell viability and activity 301
Ying Luo
3D printing systems and printer types 302
Inkjet printing 303 Introduction 343
Extrusion printing 304 Three-dimensional scaffold design and
Laser-assisted bioprinting 305 engineering 343
Stereolithography 305 Mass transport and pore architectures 344
Open source and commercial 3D printing Mechanics 346
systems 306 Electrical conductivity 348
Print outputs: patterning, resolution, and Surface properties 349
porous architecture 307 Temporal control 352
Printing/patterning of multiple inks 308 Spatial control 354
Print resolution 308 Conclusion 355
Porous architecture 309 References 355
Assessment of scaffold fidelity 309
Printing applications: vascularized and
complex, heterogeneous tissues 310
Conclusion 310
Part Five
Acknowledgments 311 Transplantation of engineered
Abbreviations 311 cells and tissues 361
References 311
20. Targeting the host immune
18. Biodegradable polymers 317 response for tissue engineering and
Julian Chesterman, Zheng Zhang,
regenerative medicine applications 363
Ophir Ortiz, Ritu Goyal and Jenna L. Dziki and Stephen F Badylak
Joachim Kohn
Introduction 363
Introduction 317 Immune cells and their roles in building
Biodegradable polymer selection criteria 317 tissues after injury 363
Biologically derived polymers 318 Neutrophils 364
Peptides and proteins 318 Eosinophils 364
Biomimetic materials 322 Macrophages 364
Polysaccharides 322 Dendritic cells 364
Polyhydroxyalkanoates 325 T and B cells 365
Polynucleotides 326 Specialized immune cell functions beyond
Synthetic polymers 326 host defense 365
Aliphatic polyesters 326 Tissue engineering/regenerative medicine
Aliphatic polycarbonates 330 strategies as immunotherapy 365
Biodegradable polyurethanes 330 Future considerations for immune cell targeting
Polyanhydrides 331 tissue engineering/regenerative medicine
Polyphosphazenes 331 therapies 366
Poly(amino acids) and pseudo-poly References 366
(amino acids) 332 Further reading 368
x Contents

21. Tissue engineering and Controlling immune responses to implanted


transplantation in the fetus 369 materials 410
Steps in the foreign body reaction 411
Christopher D. Porada, Anthony Atala and The role of geometry in the foreign body
Graça Almeida-Porada reaction 411
Introduction 369 Tuning chemical composition to prevent
Rationale for in utero therapies 370 attachment 412
In utero transplantation 371 Directing immune cell behavior in the
Early murine experiments with in utero transplant niche 412
transplantation 372 References 412
In utero transplantation experiments in large
preclinical animal models 372
Barriers to in utero transplantation success 373
Part Six
Clinical experience with in utero Stem cells 419
transplantation 376
Rationale for in utero gene therapy 376 23. Embryonic stem cells 421
Hemophilia A as a model genetic disease for Irina Klimanskaya, Erin A. Kimbrel and Robert
correction by in utero gene therapy 377 Lanza
The need for better hemophilia A
treatments 378 Introduction 421
Preclinical animal models for hemophilia A Approaches to human embryonic stem cell
and recent clinical successes 378 derivation 421
Sheep as a preclinical model of hemophilia A 379 Maintenance of human embryonic stem cell 425
Feasibility and justification for treating Subculture of human embryonic stem cell 425
hemophilia A prior to birth 380 Nuances of human embryonic stem cell
Mesenchymal stromal cells as hemophilia A culture 426
therapeutics 383 Directed differentiation 426
Preclinical success with mesenchymal stromal Safety concerns 430
cell based hemophilia A treatment 384 Conclusion 431
Risks of in utero gene therapy 385 Acknowledgment 431
Genomic integration associated insertional References 431
mutagenesis 385
Potential risk to fetal germline 386 24. Induced pluripotent stem cell
Conclusion and future directions 387 technology: venturing into the
References 388 second decade 435
Yanhong Shi, Haruhisa Inoue,
22. Challenges in the development of Jun Takahashi and Shinya Yamanaka
immunoisolation devices 403
Disease modeling 435
Matthew A. Bochenek, Derfogail Delcassian Drug discovery 436
and Daniel G. Anderson Stem cell based therapeutic development 438
Introduction 403 Concluding remarks 440
Rejection and protection of transplanted Acknowledgements 440
cells and materials 403 References 440
Rejection pathways 404
Cellular nutrition 404 25. Applications for stem cells 445
Therapeutic cells 405
Andres M. Bratt-Leal, Ai Zhang, Yanling Wang
Primary cells 405
and Jeanne F. Loring
Immortalized cell lines 406
Stem cells 407 Introduction 445
Device architecture and mass transport 407 Reprogramming of somatic cells into induced
Transplantation site 408 pluripotent stem cells 445
Improving oxygenation of immunoprotected Epigenetic remodeling 446
cells 409 Reprogramming techniques 446
Contents xi

Induced transdifferentiation 448 Microenvironmental cues 472


Genomic stability 448 Three-dimensional versus two-dimensional
Applications of induced pluripotent stem cells 448 cell culture systems 475
Disease modeling 448 High-throughput assays for directing stem cell
Challenges and future possibilities in disease differentiation 475
modeling 450 Physical signals 477
Disease-modifying potential of induced Isolation of specific progenitor cells from
pluripotent stem cells 451 embryonic stem cells 479
Other applications for induced pluripotent Transplantation 480
stem cells 452 Transplantation and immune response 481
Conclusion 452 Future prospects 482
List of acronyms and abbreviations 453 Conclusion 483
References 453 Acknowledgments 483
Conflicts of interest 483
26. Neonatal stem cells in tissue References 483
engineering 457 Further reading 490

Joseph Davidson and Paolo De Coppi


Introduction 457 Part Seven
Stem cells 457 Gene therapy 491
Embryonic stem cells 457
Induced pluripotent stem cells 458 28. Gene therapy 493
Perinatal stem cells 458
Stefan Worgall and Ronald G. Crystal
Scaffolding specifics in fetal and neonatal
tissue engineering 459 Strategies of gene therapy 493
Synthetic materials 459 Ex vivo versus in vivo gene therapy 494
Natural materials 459 Ex vivo 494
Relevance to prenatal therapy 460 In vivo 495
Immunology 460 Chromosomal versus extrachromosomal
Physiology 460 placement of the transferred gene 495
Conditions of interest 461 Gene transfer vectors 495
Spina bifida 461 Nonviral vectors 497
Gastroschisis 461 Adenovirus 497
Congenital diaphragmatic hernia 461 Adeno-associated virus 499
Esophageal atresia 461 Retrovirus 500
Congenital heart disease 462 Lentivirus 501
Congenital airway anomalies 462 Cell-specific targeting strategies 502
Bladder 463 Targeting of Ad vectors 502
Bone and bone marrow 463 Targeting of adeno-associated virus vectors 505
Conclusion 463 Targeting of retroviral and lentiviral vectors 505
References 463 Regulated expression of the transferred gene 505
Using gene transfer vectors for gene editing 507
27. Embryonic stem cells as a cell Combining gene transfer with stem-cell
source for tissue engineering 467 strategies 508
Gene transfer to stem cells 508
Ali Khademhosseini, Nureddin Ashammakhi,
Gene transfer to control uncontrolled
Jeffrey M. Karp, Sharon Gerecht, Lino Ferreira,
stem-cell growth 508
Nasim Annabi, Mohammad Ali Darabi, Dario
Gene transfer to instruct stem-cell
Sirabella, Gordana Vunjak-Novakovic and
differentiation 508
Robert Langer
Gene transfer to regulate gene expression 509
Introduction 467 Challenges to gene therapy for tissue
Maintenance of embryonic stem cells 468 engineering 509
Directed differentiation 471 Acknowledgments 510
Genetic reprogramming 471 References 510
xii Contents

29. Gene delivery into cells and tissues 519 Special considerations 565
Breast cancer modeling 565
Christopher E. Nelson, Craig L. Duvall, Aleš Animal models 565
Prokop, Charles A. Gersbach and Jeffrey M. Breast tissue test systems 566
Davidson In silico breast cancer models 570
Introduction 519 Concluding remarks 571
Fundamentals of gene delivery 519 Acknowledgement 571
Biodistribution, targeting, uptake, and References 571
trafficking 521
Tissue biodistribution/targeting 521 Part Nine
Cellular uptake and intracellular trafficking 523
Viral nucleic acid delivery 526
Cardiovascular system 577
Introduction to viral gene therapy 526
Types of viral vectors 527 31. Cardiac progenitor cells, tissue
Engineering viral vectors 528 homeostasis, and regeneration 579
Nonviral nucleic acid delivery 530 Wayne Balkan, Simran Gidwani, Konstantinos
Introduction to nonviral nucleic acid delivery 530 Hatzistergos and Joshua M. Hare
Oligonucleotide modifications 531
Conjugates 531 Origin of cardiac stem/progenitor cells 579
Synthetic polymers 531 Modeling cardiac development with
Polymers derived from natural sources or pluripotent stem cells 581
monomers 534 In vivo fate mapping of cardiac progenitors 582
Lipid-based delivery systems 536 Neonatal cardiac repair 582
Inorganic nanoparticles 537 Reprogramming cardiac fibroblasts 584
High-throughput screening 537 Cardiac resident mesenchymal stem cells 584
Engineering tissues with gene delivery 538 Cardiomyocytes and cardiac repair/
Introduction to engineering tissue with gene regeneration 585
delivery 538 Cell-based therapy 585
Viral delivery to engineer tissues 538 Cardiac progenitor/stem cell therapy 586
Nonviral delivery from scaffolds 540 Combination stem cell therapy 586
Nucleic acid delivery for tissue engineering Pluripotent stem cells 586
advances into the clinic 541 Future directions 588
Future challenges 541 References 588
Outlook 542
Acknowledgments 543 32. Cardiac tissue engineering 593
References 543
Yimu Zhao, George Eng, Benjamin W. Lee,
Milica Radisic and Gordana Vunjak-Novakovic
Part Eight Introduction 593
Breast 555 Clinical problem 593
Engineering cardiac tissue: design principles
30. Breast tissue engineering: and key components 594
implantation and three-dimensional Cell source 594
tissue test system applications 557 Scaffold 598
Biophysical stimulation 599
Karen J.L. Burg and Timothy C. Burg
Directed cardiac differentiation of human
Introduction 557 stem cells 599
Breast anatomy and development 557 Derivation of cardiomyocytes from human
Breast cancer diagnosis and treatments 558 pluripotent stem cells 599
Breast reconstruction 558 Purification and scalable production of stem
Synthetic implants 559 cell derived cardiomyocytes 601
Tissue flaps 559 Scaffolds 601
Cell transplants 559 Decellularization approach 601
Cellular scaffolds 560 Artificial scaffolds 602
Contents xiii

Biophysical cues 604 Bioresorbable grafts 625


Electrical stimulation 604 The living bioreactor 626
Mechanical stimulation 604 Cellular and molecular mediators of graft
Perfusion 606 outcome 626
In vivo applications of cardiac tissue Conclusion and predictions for the future 630
engineering 606 References 630
Engineered heart issue 606
Vascularized cardiac patches 608 34. Heart valve tissue engineering 635
Electrical coupling of cardiomyocytes on the
heart 608 Kevin M. Blum, Jason Zakko, Peter Fong,
Modeling of disease 609 Mark W. Maxfield, Muriel A. Cleary and
Generation of patient-specific Christopher K. Breuer
cardiomyocytes 609 Introduction 635
Engineered heart tissue models 609 Heart valve function and structure 635
Cardiac fibrosis 609 Cellular biology of the heart valve 636
Titin mutation related dilated Heart valve dysfunction and valvular repair
cardiomyopathy 611 and remodeling 637
Diabetes-related cardiomyopathy 611 Heart valve replacement 638
Chronic hypertension induced left ventricle The application of tissue engineering
hypertrophy 611 toward the construction of a replacement
Barth syndrome 611 heart valve 640
Tissue engineering as a platform for Tissue engineering theory 640
pharmacologic studies 611 Biomaterials and scaffolds 640
Summary and challenges 612 The search for appropriate cell sources 643
Acknowledgments 612 Cell seeding techniques 644
References 612 Bioreactors 645
Neotissue development in tissue engineered
33. Blood vessels 617 heart valves 645
Luke Brewster, Eric M. Brey and Clinical applications of the tissue engineered
Howard P. Greisler heart valve 647
Conclusion and future directions 648
Introduction 617 References 649
Normal and pathologic composition of
the vessel wall 617
Developmental biology cues important in
vascular tissue engineering 618
Part Ten
Conduits 618 Endocrinology and metabolism 655
Arteries 618
Veins 618 35. Generation of pancreatic islets
Current status of grafts in patients 618 from stem cells 657
Conduit patency and failure 618
Bárbara Soria-Juan, Javier López-Beas,
Venous reconstruction 619
Bernat Soria and Abdelkrim Hmadcha
Hemodialysis vascular access 619
Inflammation and the host response to Introduction 657
interventions and grafts 620 State-of-the-art 657
Host environment and the critical role of the The challenge of making a β-cell 658
endothelium 621 Recent achievements (first generation of
Prevalent grafts in clinical use 622 pancreatic progenitors used in the clinic) 658
Vascular tissue engineering 623 Need of late maturation: cabimer protocol 659
Early efforts—in vitro tissue-engineered Strategies to maintain cell viability 659
vascular grafts 623 Encapsulation and tolerogenic strategies 661
Endothelial cell seeding 623 The concept of cellular medicament 661
In vitro approaches to tissue-engineered Conclusion 662
vascular grafts 624 Acknowledgments 662
In vivo tissue-engineered vascular grafts 625 References 662
xiv Contents

36. Bioartificial pancreas: challenges Maintenance and regeneration of thymic


and progress 665 epithelial cells: Progenitor/stem cells in
the adult thymus 696
Paul de Vos Strategies for thymus reconstitution 697
Introduction 665 Summary 698
History of the bioartificial pancreas 666 Acknowledgments 699
Replenishable cell sources and encapsulation 666 References 699
Macro- or microedevices 667
Factors contributing to biocompatibility of
encapsulation systems 669 Part Eleven
Avoiding pathogen-associated molecular
patterns in polymers 670
Gastrointestinal system 707
Natural and synthetic polymers 670
Multilayer capsule approaches 670
38. Stem and progenitor cells of the
Antibiofouling approaches 671
gastrointestinal tract: applications for
Formation of polymer brushes 671 tissue engineering the intestine 709
Immunomodulatory materials 672 Kathryn M. Maselli, Christopher R. Schlieve,
Intracapsular environment and longevity Mark R. Frey and Tracy C. Grikscheit
of the encapsulated islet graft 672
Concluding remarks and future Introduction 709
considerations 673 Stem cells of the intestine 709
Acknowledgments 674 Cell types of the epithelial layer 709
References 674 Stem and progenitor cell types 710
Signaling pathways in the intestinal
epithelium 712
37. Thymus and parathyroid
The Wnt pathway 712
organogenesis 681 The Notch pathway 713
Craig Scott Nowell, Kathy E. O’Neill, Paul Rouse, Epidermal growth factor receptor/ErbB
Timothy Henderson, Ellen Rothman Richie, signaling 713
Nancy Ruth Manley and Catherine Clare Blackburn The Hedgehog pathway 714
The BMP pathway 714
Structure and morphology of the thymus 681
Tissue engineering the intestine with stem/
Thymic epithelial cells 682
progenitor cells 714
Complexity of the thymic epithelium
Organ-specific stem cell progenitors versus
compartment 682
pluripotent stem cells 714
Functional diversity 683
Synthetic and biological scaffolds 715
In vitro T cell differentiation 683
Primary intestinal-derived organoid units 716
Thymus organogenesis 685
Pluripotent stem cell approaches—human
Cellular regulation of early thymus
intestinal organoids 717
organogenesis 685
Remaining barriers to the generation of
Origin of thymic epithelial cells 686
tissue-engineered intestine 718
Thymic epithelial progenitor cells 686
Conclusion 718
Human thymus development 688
Acknowledgment 718
Cervical thymus in mouse and human 688
References 718
Molecular regulation of thymus and
parathyroid organogenesis 689
Molecular control of early organogenesis 689
39. Liver stem cells 723
Transcription factors and regulation of third Dagmara Szkolnicka and David C. Hay
pharyngeal pouch outgrowth 691
Introduction 723
Specification of the thymus and
Liver architecture and function 723
parathyroid 692
Liver development 723
Foxn1 and regulation of thymic epithelial cell
Fetal liver stem cells 724
differentiation 695
Hepatocytes and liver progenitors in organ
Medullary development and expansion 696
regeneration 724
Contents xv

Molecular signaling and processes involved in Biliary network engineering 747


liver regeneration 724 Conclusion and outlook 747
Hepatocytes’ role in liver regeneration 725 References 748
Cholangiocytes and liver stem cells in liver
regeneration 725
Pluripotent stem cell derived hepatoblasts Part Twelve
and hepatocytes 726
3D liver organoids and expansion 727
Hematopoietic system 755
Pluripotent stem cell derived liver
organoids 728 41. Hematopoietic stem cells 757
Bile duct derived organoids 728 Qiwei Wang, Yingli Han, Linheng Li and
Hepatocyte-derived organoids 728 Pengxu Qian
Novel scaffolds for liver organoids 729
Organoids as a model to study liver cancer Introduction 757
disease 730 Hematopoietic stem cells and hematopoietic
Reprogramming of human hepatocytes to liver stem cells niche 757
progenitors using different culture Effects of biomaterials on hematopoietic stem
conditions 730 cells 758
Conclusion 731 Applications 759
References 731 Engineering hematopoietic stem cells niche
Further reading 736 for in vitro expansion 759
Manipulation of the multilineage
40. Hepatic tissue engineering 737 differentiation of hematopoietic stem cells 760
In vivo tracking hematopoietic stem cells 761
Amanda X. Chen, Arnav Chhabra, Future perspectives 761
Heather E. Fleming and Sangeeta N. Bhatia Acknowledgments 761
Liver disease burden 737 References 761
Current state of liver therapies 738
Extracorporeal liver support devices 738 42. Blood components from
Biopharmaceuticals 738 pluripotent stem cells 765
Liver transplantation 738
Erin A. Kimbrel and Robert Lanza
Hepatocyte transplantation 740
Current clinical trials 740 Introduction and history of modern
In vitro models 740 hematology 765
Two-dimensional liver culture 741 Red blood cells 765
Three-dimensional liver constructs 741 Megakaryocytes/platelets 769
Physiological microfluidic models of liver 742 White blood cells 770
Controlling three-dimensional architecture Lymphocytes—T cells 770
and cellular organization 742 Lymphocytes—NK cells 773
In vivo models 743 Lymphocytes—NKT cells 775
Cell sourcing 743 Monocyte-derived dendritic cells 776
Cell number requirements 743 Monocyte-derived macrophages 777
Immortalized cell lines 744 Granulocytes—neutrophils 778
Primary cells 744 Perspectives 779
Fetal and adult progenitors 744 References 779
Reprogrammed hepatocytes 744
Extracellular matrix for cell therapies 744 43. Red blood cell substitutes 785
Natural scaffold chemistry and modifications 745
Andre Francis Palmer and Donald Andrew Belcher
Synthetic scaffold chemistry 745
Modifications in scaffold chemistry 745 Introduction 785
Porosity 746 Replicating red blood cell functions 785
Vascular and biliary tissue engineering 746 Hemoglobin-based oxygen carriers 785
Vascular engineering 746 Hemoglobin toxicity 787
Host integration 747 Oxygen delivery 789
xvi Contents

Viscosity and colloid osmotic pressure 789 Conclusion and future perspectives 837
Cross-linked and polymeric hemoglobin 790 Acknowledgment 838
Surface conjugated hemoglobin 790 References 838
Encapsulated hemoglobin 791
Sources of hemoglobin 791 46. Tissue engineering: bladder and
Recombinant hemoglobin 792 urethra 845
Erythrocruorins 792
Perfluorocarbons 793 Yuanyuan Zhang, James J. Yoo and
Perspectives 794 Anthony Atala
Organ transplant preservation 794 Introduction 845
Cancer treatment 795 Cell sources 846
Tissue-engineered construct oxygenation 795 Bladder and ureter cells 846
References 795 Stem cell sources 846
Mechanism of cell therapy 848
Part Thirteen Biodegradable biomaterials 850
Synthetic scaffolds 850
Kidney and genitourinary system 803 Natural collagen matrix 851
Preclinical models 854
44. Stem cells in kidney development Tissue regeneration models 854
and regeneration 805 Fibrotic bladder model 854
Kyle W. McCracken and Joseph V. Bonventre Clinical trials 856
Clinical translation 856
Kidney development 805 Clinical studies 857
Early embryonic origins of nephrogenic Conclusion 858
tissues 806 References 858
Development of the nephric duct and
ureteric bud 808
Maintenance and differentiation of the 47. Tissue engineering for female
nephron progenitor cell 809 reproductive organs 863
Role of stromal lineages in kidney Renata S. Magalhaes, James K. Williams and
organogenesis 811 Anthony Atala
Nephron endowment 812
Kidney repair and regeneration 813 Introduction 863
Stem cells in kidney repair 813 Uterus 863
Sources of nephrogenic cells 814 Acellular tissue engineering approaches
Differentiation of renal tissue from for uterine tissue repair 864
pluripotent stem cells (organoids) 815 Cell-seeded scaffolds for partial uterine repair 864
Conclusion 817 Scaffold-free approaches for partial uterine
Disclosures 818 repair 865
Acknowledgements 818 Uterine cervix tissue engineering 865
References 818 Ovary 865
Tissue engineering ovarian follicles 866
45. Tissue engineering of the kidney 825 Vagina 866
Tissue engineering approaches for neovagina
Ji Hyun Kim, Anthony Atala and James J. Yoo reconstruction 866
Conclusion and future perspectives 867
Introduction 825
References 867
Cell-based tissue engineering of the kidney 826
Cell sources 826
Tissue-engineered cellular three-dimensional 48. Male reproductive organs 871
renal constructs 830
Hooman Sadri-Ardekani, John Jackson and
Cell-free tissue engineering of the kidney 835
Anthony Atala
In situ kidney regeneration 835
Granulocyte-colony stimulating factor 835 Introduction 871
Stromal cell derived factor-1 837 Testes 871
Contents xvii

Spermatogonial stem cell technology 871 50. Bone tissue engineering and bone
Androgen-replacement therapy 873 regeneration 917
Ejaculatory system 874
Engineering vas deferens 874 J.M. Kanczler, J.A. Wells, D.M.R. Gibbs, K.M.
Spinal ejaculation generator 875 Marshall, D.K.O. Tang and Richard O.C. Oreffo
Penis 875 Introduction 917
Penile reconstruction 875 Skeletal stem cells 917
Penile transplantation 876 Fracture repair—the (limited) self-reparative
Stem cell therapy for erectile dysfunction 876 capacity of bone 919
Conclusion 877 A framework for bone repair:
References 877 biomaterial-driven strategies for bone
regeneration 922
Growth factors: biomimetic-driven strategies
Part fourteen for bone regeneration 923
Musculoskeletal system 881 Bone biofabrication 924
Development of vascular bone 925
49. Mesenchymal stem cells in Preclinical development—ex vivo/in vivo
musculoskeletal tissue engineering 883 small and large animal preclinical models 926
Yangzi Jiang, Dan Wang, Anna Blocki and Clinical translation 929
Rocky S. Tuan Summary and future perspectives 931
Acknowledgments 931
Introduction 883 References 931
Mesenchymal stem cell biology relevant to
musculoskeletal tissue engineering 883 51. Tissue engineering for regeneration
Mesenchymal stem cell identification 883
and replacement of the intervertebral
Tissue sources of mesenchymal stem cells 885
disk 937
Mesenchymal stem cell isolation and in vitro
culture 886 Stephen R. Sloan Jr., Niloofar Farhang, Josh Stover,
Mesenchymal stem cell self-renewal and Jake Weston, Robby D. Bowles and Lawrence J.
proliferation capacity 887 Bonassar
Skeletogenic differentiation of mesenchymal
Introduction 937
stem cells 888
Intervertebral disk structure and function 938
Plasticity of mesenchymal stem cells 888
Cell-biomaterial constructs for intervertebral
Mesenchymal stem cell heterogeneity 889
disk regeneration 940
Mesenchymal stem cell effect on host
Nucleus pulposus cell-biomaterial implants 940
immunobiology 889
Annulus fibrosus repair and regeneration 942
Safety of using mesenchymal stem cells for
Composite cell-biomaterial intervertebral disk
transplantation 891
implants 944
Mesenchymal stem cells in musculoskeletal
Cellular engineering for intervertebral disk
tissue engineering 891
regeneration 945
Cartilage tissue engineering 891
Cell therapy preclinical studies 946
General properties of articular cartilage 892
Cell therapy clinical studies 947
Cells for cartilage tissue engineering 892
Growth factors and other biologics for
Bone tissue engineering 897
intervertebral disk regeneration 948
Osteochondral tissue engineering 898
In vitro studies 948
Engineering other skeletal tissues with
In vivo studies: growth factors 952
mesenchymal stem cells 899
In vivo studies: other biologics 953
Tendon/ligament 899
Gene therapy for intervertebral disk
Meniscus 900
regeneration 953
Gene therapy in musculoskeletal tissue
Gene transfer studies: viral 954
engineering 901
Gene transfer studies: nonviral 954
Conclusion and future perspectives 901
Endogenous gene regulation 955
Acknowledgments 902
Gene therapy in summary 955
References 902
xviii Contents

In vivo preclinical models for intervertebral Introduction 989


disk regeneration and replacement 955 Tendon and ligament composition, structure,
Concluding remarks 957 and function 990
Acknowledgment 957 Composition 990
References 957 Structure 990
Function 990
52. Articular cartilage injury 967 Requirements for a tissue-engineered
tendon/ligament 991
J.A. Martin, M. Coleman and J.A. Buckwalter Scaffold 992
Introduction 967 Cell 994
Articular cartilage injury and joint Bioactive factors 995
degeneration 968 Three-dimensional bioprinting and bioink 996
Mechanisms of articular cartilage injuries 968 Bioink inspired from ligament and tendon
Response of articular cartilage to injury 970 structures 997
Matrix and cell injuries 970 Tissue engineering tendon and ligament in
Chondral injuries 971 clinical application 998
Osteochondral injuries 971 Summary 999
Preventing joint degeneration following injury 972 References 1000
Promoting articular surface repair 972
Penetration of subchondral bone 972 55. Skeletal tissue engineering 1007
Periosteal and perichondrial grafts 973
Matthew P. Murphy, Mimi R. Borrelli,
Cell transplantation 973
Daniel T. Montoro, Michael T. Longaker and
Artificial matrices 973
Derrick C. Wan
Growth factors 973
Antiinflammatories 974 Introduction 1007
Conclusion 974 Distraction osteogenesis 1008
Acknowledgments 974 Critical-sized defects 1010
References 974 Cellular therapy 1010
Further reading 977 Cytokines 1013
Scaffolds 1014
53. Engineering cartilage and other Tissue engineering in practice 1016
structural tissues: principals of bone Conclusion 1017
and cartilage reconstruction 979 References 1017

Batzaya Byambaa and Joseph P. Vacanti


Introduction 979 Part Fifteen
Biomaterials for cartilage tissue engineering 979 Nervous system 1023
Cell sources for cartilage tissue engineering 980
Biofabrication of cartilage tissue 981 56. Brain implants 1025
Magnetic resonance imaging and
Lars U. Wahlberg
computerized tomography scans 981
Scaffolds for cartilage tissue engineering 981 Introduction 1025
Bioprinting techniques for fabrication of Cell replacement implants 1025
cartilage constructs 982 Primary tissue implants 1025
Bioinks for cartilage tissue printing 982 Cell line implants 1027
Osteochondral tissue engineering 985 Cell protection and regeneration implants 1028
References 985 Cell implants secreting endogenous factors 1028
Cell implants secreting engineered factors
54. Tendon and ligament tissue (ex vivo gene therapy) 1029
engineering 989 Encapsulated cell brain implants 1029
Controlled-release implants 1030
Spencer P. Lake, Qian Liu, Malcolm Xing, Combined replacement and regeneration
Leanne E. Iannucci, Zhanwen Wang and implants 1030
Chunfeng Zhao Disease targets for brain implants 1031
Contents xix

Surgical considerations 1032 Calcium influx 1094


Conclusion 1032 Endoplasmic reticulum stress 1094
References 1032 Prevention of ototoxicity 1094
Prevention of acoustic trauma 1096
57. Brain machine interfaces 1037 Antiinflammatory agents 1097
Heat shock proteins 1097
José del R. Millán and Serafeim Perdikis Neurotrophic factors 1098
Introduction 1037 Protection from excitotoxicity: “acquired”
Brain machine interface signals 1037 loss of auditory nerve connections to hair
Voluntary activity versus evoked potentials 1038 cells 1098
Mutual learning 1040 Gene transfer for the prevention and
Context-aware brain machine interface 1040 treatment of genetic deafness 1099
Future directions 1041 Interventions for hair cell repair: gene
References 1042 therapy for transdifferentiation 1099
Interventions for repair: hair cell and
58. Spinal cord injury 1047 auditory nerve replacement—exogenous
stem cells 1101
Nicolas N. Madigan and Anthony J. Windebank Interventions for repair/replacement:
Introduction 1047 cochlear prostheses 1101
Epidemiology 1047 Fully implantable cochlear prostheses 1101
Spinal cord organization 1047 Interventions for repair/replacement: central
Spinal cord injury 1048 auditory prostheses 1102
Available clinical interventions 1049 Local delivery to cochlear fluids 1103
The continuum of physical, cellular, and Conclusion 1103
molecular barriers to spinal cord Acknowledgments 1103
regeneration 1049 References 1104
The role of tissue engineering in spinal cord Further reading 1112
injury repair 1051
Bioengineering for integrated spinal cord
biocompatibility 1052 Part Sixteen
Animal models of spinal cord injury 1052 Ophthalmic 1113
Principles of biomaterial fabrication for
spinal cord injury repair 1054
60. Stem cells in the eye 1115
Biomaterials for spinal cord tissue
engineering: natural polymers 1058 Chao Huang, Julie Albon, Alexander Ljubimov
Extracellular matrix polymers 1058 and Maria B. Grant
Polymers from marine or insect life 1065
Introduction 1115
Polymers derived from the blood 1071
Endogenous ocular stem cells 1115
Biomaterials for spinal cord tissue
Corneal stem cells 1115
engineering: synthetic polymers 1072
Stromal stem cells 1119
Poly α-hydroxy acid polymers 1073
Endothelial stem cells 1119
Nonbiodegradable hydrogels 1077
Conjunctival epithelial stem cells 1120
Conclusion and future directions:
The bioengineered cornea 1120
the promise of clinical translation 1080
Retinal progenitor cells 1120
References 1080
Müller stem cells 1121
Retinal pigment epithelium stem cells 1121
59. Protection and repair of hearing 1093 Nonocular stem cells 1121
Su-Hua Sha, Karl Grosh and Richard A. Altschuler Induced pluripotent stem cells (iPSCs) 1121
Embryonic stem cells/iPSCs in retinal
Introduction 1093 regeneration 1121
Protection from “acquired” sensory hair cell Bone marrow stem cells 1124
loss 1093 References 1126
Oxidative stress and stress-related
mitochondrial pathways 1094
xx Contents

61. Corneal replacement tissue 1135 Optogenetics 1174


Outer retinal cell transplantation 1177
Maria Mirotsou, Masashi Abe and Robert Lanza Cell matrices supporting axonal regrowth 1177
Introduction 1135 Repopulating ischemic or diabetic retina 1178
Corneal anatomy and structure 1135 Assessing the functional outcomes of novel
Epithelium 1136 retinal therapies 1178
Stroma 1138 Conclusion: toward 2020 vision 1179
Endothelium 1139 Acknowledgment 1179
Conclusion 1140 References 1179
References 1141 Further reading 1183

62. Retinal degeneration 1145


Part Seventeen
Erin A. Kimbrel and Robert Lanza
Oral/Dental applications 1185
Epidemiology of visual impairment and
blindness 1145 64. Biological tooth replacement and
Structure/function of the retina and cell types repair 1187
affected in retinal degenerative diseases 1145
Age-related macular degeneration 1147 Anthony J. (Tony) Smith and Paul T. Sharpe
History of retinal pigment epithelium as a Introduction 1187
cellular therapy for age-related macular Tooth development 1187
degeneration 1147 Whole tooth-tissue engineering 1189
Retinal pigment epithelium from pluripotent Stem cell-based tissue engineering of teeth 1189
stem cells 1149 Bioteeth from cell-seeded scaffolds 1189
Retinitis pigmentosa 1150 Root formation 1190
Photoreceptors from pluripotent stem cells 1151 Cell sources 1191
Glaucoma 1153 Dental-tissue regeneration 1191
Stem cell based therapies to treat glaucoma 1154 Natural tissue regeneration 1191
Diabetic retinopathy 1155 Importance of the injury-regeneration
Stem cell based therapies to treat diabetic balance 1192
retinopathy 1155 Signaling events in dental regeneration 1193
Future directions and competing therapies 1156 Control of specificity of dental-tissue
References 1157 regeneration 1193
Dental postnatal stem cells 1194
63. Vision enhancement systems 1163 Directed tissue regeneration 1195
Signaling-based strategies 1195
Gislin Dagnelie, H. Christiaan Stronks and Cell- and gene-based strategies 1196
Michael P. Barry Conclusion 1197
Introduction 1163 References 1197
Visual system, architecture, and (dys)function 1163
Current- and near-term approaches to vision 65. Tissue engineering in oral and
restoration 1166 maxillofacial surgery 1201
Enhancing the stimulus through
Simon Young, F. Kurtis Kasper, James Melville,
optoelectronic and optical means 1166
Ryan Donahue, Kyriacos A. Athanasiou,
Visual prostheses based on electrical tissue
Antonios G. Mikos and Mark Eu-Kien Wong
stimulation 1167
Retinal cell transplantation 1170 Introduction 1201
Optic nerve protection and regeneration 1171 Special challenges in oral and maxillofacial
Drug delivery 1172 reconstruction 1201
Genetic interventions 1172 Current methods of oral and maxillofacial
Emerging application areas for engineered reconstruction 1204
cells and tissues 1173 Mandibular defects 1205
Photosensitive structures 1174 Maxillary defects 1207
Contents xxi

Relevant strategies in oral and maxillofacial Introduction: challenges facing cell and
tissue engineering 1208 tissue-based therapy for the treatment of
Bone applications 1208 lung disease 1253
Cartilage applications 1212 Lung morphogenesis informs the process of
Oral mucosa applications 1214 regeneration 1254
Composite tissue applications 1215 Integration and refinement of signaling and
Animal models 1215 transcriptional pathways during lung
The future of oral and maxillofacial tissue formation 1256
engineering 1216 The mature lung consists of diverse
References 1216 epithelial and mesenchymal cell types 1256
Structure and function of pulmonary
66. Periodontal tissue engineering and vasculature 1257
regeneration 1221 Embryonic development of alveolar
capillaries 1258
Xiao-Tao He, Rui-Xin Wu and Fa-Ming Chen Evidence supporting lung regeneration 1259
Introduction 1221 A diversity of lung epithelial progenitor/stem
Stem cells for periodontal bioengineering 1222 cells is active during regeneration 1260
Intraoral mysenchymal stem cells 1222 Role of lung microvasculature in lung repair 1262
Periodontal tissue derived stem cells 1223 Endothelial progenitor cells in lung repair 1262
Stem cells from apical papilla 1224 Pulmonary cell-replacement strategies for
Dental follicle stem cells 1224 lung regeneration 1263
Hertwig’s epithelial root sheath 1225 Induced pluripotent stem cells for study of
Stem cells from dental pulp or exfoliated treatment of pulmonary disease 1263
deciduous teeth 1225 Differentiation of induced pluripotent stem
Extraoral mysenchymal stem cells 1225 and embryonic stem cells to pulmonary
Bone marrow derived mysenchymal stem epithelial cell lineages 1264
cells 1225 Bioengineering of lung tissues 1265
Adipose-derived stem cells 1226 Mesenchymal stromal cells and mesenchymal
Selection of cell types 1226 stromal cell products for the treatment of
Signaling molecules 1227 lung disease 1265
Types of signals 1228 Important role of the extracellular matrix in
Crucial delivery barriers to progress 1230 lung structure and repair 1265
Gene delivery as an alternative to growth Tissue engineering for conducting airways 1266
factor delivery 1231 Pulmonary macrophage transplantation for the
Scaffolding and biomaterials science 1232 treatment of interstitial lung disease 1266
Requirements of cell scaffolds 1232 Conclusion 1266
Biomaterial-based immune modulation 1233 Acknowledgments 1266
Classes of biomaterials 1233 References 1266
Biomaterial redesign for periodontal
application 1235 68. Lung tissue engineering 1273
Periodontal bioengineering strategies 1236
Micha Sam Brickman Raredon, Yifan Yuan
Cell-free approaches 1237
and Laura E. Niklason
Cell-based approaches 1239
Challenges and future directions 1242 Introduction 1273
Closing remarks 1243 Design criteria for pulmonary engineering 1273
Acknowledgments 1243 Decellularized scaffolds and biofabrication
References 1243 approaches 1274
Pulmonary epithelial engineering 1276
Proximal airway engineering 1276
Part Eighteen Distal airway engineering 1276
Respiratory system 1251 Mesenchymal support of pulmonary
epithelium 1277
Pulmonary endothelial engineering 1277
67. Cell- and tissue-based therapies
Endothelial cell sources for lung tissue
for lung disease 1253 engineering 1278
Jeffrey A. Whitsett, William Zacharias, Endothelial seeding into lung scaffolds 1278
Daniel Swarr and Vladimir V. Kalinichenko Organomimetic endothelial culture 1279
xxii Contents

Mesenchymal support of pulmonary Inflammation 1310


microvasculature 1280 Transition from inflammation to
Bioreactor technologies for pulmonary repair 1310
engineering 1280 Reepithelialization 1310
Conclusion 1281 Granulation tissue 1312
References 1281 Wound contraction and extracellular matrix
organization 1316
Chronic wounds 1317
Part Nineteen Scarring 1318
Skin 1287 Pathological scars 1318
Scarless healing 1319
69. Cutaneous epithelial stem cells 1289 Tissue engineered therapy with skin cells 1320
Denise Gay, Maksim V. Plikus, Iris Lee, Elsa Engineered epidermal constructs 1320
Treffeisen, Anne Wang and George Cotsarelis Engineered dermal constructs 1321
Engineered skin substitutes 1321
Introduction 1289 Skin autograft harvesting without scarring 1322
Interfollicular epidermal stem cells 1289 Tissue-engineered therapy with stem cells,
Models for skin renewal: epidermal bioactives, and biomaterials 1322
proliferative unit versus committed References 1324
progenitor 1290
Hair follicle stem cells 1291 71. Bioengineered skin constructs 1331
The bulge as stem cell source 1291
Vincent Falanga
Defining characteristics of the bulge as a
stem cell source 1292 Introduction 1331
Multiple hair follicle stem cell subpopulations Skin structure and function 1331
by marker expression 1294 The epidermis 1331
Stem cells of other ectodermal appendages 1295 The dermis 1332
Sebaceous glands 1295 The process of wound healing 1333
Sweat glands 1296 Impaired healing and its mechanisms 1333
Nails 1296 Acute versus chronic wound healing 1333
Hair follicle stem cells in skin homeostasis, Bacterial colonization 1333
wound healing, and hair regeneration 1297 Growth factor imbalances 1334
Homeostasis 1297 Matrix metalloproteinase activity 1334
Wound healing 1297 Moist wound healing in chronic wounds 1334
Wound-induced hair follicle neogenesis and Ischemia 1334
regeneration 1298 Abnormalities at the cellular level 1335
Epithelial stem cells in aging 1298 Engineering skin tissue 1335
Role of stem cells in alopecia 1299 Design considerations 1335
Skin as an active immune organ 1300 Commercial considerations 1336
Cross talk between hair follicles and the Process considerations 1337
immune system 1300 Regulatory considerations 1337
The inflammatory memory of skin cells 1301 Immunological considerations 1338
Tissue engineering with epidermal stem cells 1301 Summary: engineering skin tissue 1338
Epidermal stem cells as a therapy: the future 1302 Epidermal regeneration 1338
Conclusion 1302 Dermal replacement 1339
References 1302 Bioengineered living skin equivalents 1339
Bioengineered skin: FDA-approved
70. Wound repair: basic biology to indications 1340
tissue engineering 1309 Cutaneous indications 1340
Oral indications 1341
Richard A.F. Clark, Michael Musillo and Apligraf and Dermagraft: off-label uses 1341
Thomas Stransky The importance of wound bed preparation 1344
Introduction 1309 Proposed mechanisms of action of
Basic biology of wound repair 1310 bioengineered skin 1345
Contents xxiii

Construct priming and a new didactic Tissue engineering of muscle fibers 1378
paradigm for constructs 1347 Scaffolds 1378
Other considerations 1348 Industrial bioreactors 1379
Conclusion 1348 Fields 1380
References 1349 Atrophy and exercise 1380
Further reading 1352 Senescence 1381
Meat processing technology 1381
Associated dangers and risks 1381
Part Twenty
Regulatory issues 1381
Tissue-engineered food 1353 Consumer acceptance and perception 1382
Role of media in publicity of cultured meat 1382
72. Principles of tissue engineering for Market for cultured meat 1382
food 1355 Conclusion 1383
Mark Post and Cor van der Weele References 1384

Introduction 1355
Why tissue engineering of food? 1355
Specifics of tissue engineering for medical
Part Twentyone
application 1356 Emerging technologies 1389
Uniqueness 1356
Function 1356 74. Three-dimensional bioprinting for
Skeletal muscle and fat tissue tissue engineering 1391
engineering 1357
Jun Tae Huh, James J. Yoo, Anthony Atala and
Tissue engineering of skeletal muscle 1357
Sang Jin Lee
Tissue engineering of fat 1359
Specifics of food tissue engineering 1361 Introduction 1391
Scale 1361 3D Bioprinting strategy: from medical
Efficiency 1362 image to printed bioengineered tissue 1391
Taste, texture, juiciness 1362 Three-dimensional bioprinting techniques 1392
Enhanced meat 1363 Jetting-based bioprinting 1392
Other foods 1363 Extrusion-based bioprinting 1394
Consumer acceptance 1364 Laser-assisted bioprinting 1394
Regulatory pathway 1365 Laser-based stereolithography 1395
Conclusion 1365 Digital light processing 1395
References 1365 Hybrid and other techniques 1396
Biomaterials as bioinks for three-dimensional
73. Cultured meat—a humane meat bioprinting 1396
production system 1369 Hydrogel-based bioinks for cell-based
three-dimensional bioprinting 1396
Zuhaib F. Bhat, Hina Bhat and Sunil Kumar Biodegradable synthetic polymers for
Introduction 1369 structure-based three-dimensional
Need and advantages of cultured meat 1370 bioprinting 1399
Cultured meat 1372 Scaffold-free cell printing 1399
Scaffolding techniques 1372 Three-dimensional bioprinting in tissue
Self-organizing tissue culture 1373 engineering applications 1400
Organ printing 1375 Three-dimensional bioprinted vascular
Biophotonics 1375 structures 1400
Nanotechnology 1375 In vitro tissue models 1400
Challenges and requirements for industrial Three-dimensional bioprinted implantable
production 1375 tissue constructs 1403
Generation of suitable stem cell lines from Conclusion and future
farm-animal species 1376 perspectives 1409
Safe media for culturing of stem cells 1377 Abbreviations 1410
Safe differentiation media to produce muscle Glossary 1410
cells 1377 References 1411
xxiv Contents

75. Biofabricated three-dimensional Tissue-specific 1463


tissue models 1417 Cartilage monitoring and real-time control 1463
Skin 1464
David B. Berry, Claire Yu and Shaochen Chen Concluding remarks 1464
Introduction 1417 Acknowledgments 1465
Current methods of three-dimensional References 1465
biofabrication 1418
Biomaterials for three-dimensional 78. Biomanufacturing for regenerative
fabrication 1421 medicine 1469
Three-dimensional tissue models for drug
Joshua G. Hunsberger and Darren H.M. Hickerson
screening, disease modeling, therapeutics,
and toxicology 1425 Current landscape of biomanufacturing 1469
Conclusion and future directions 1435 Highlighting current workflows for
Acknowledgments 1435 biomanufacturing 1470
References 1435 Current challenges in biomanufacturing for
regenerative medicine 1470
76. Body-on-a-chip: three-dimensional Current platform technologies enabling
engineered tissue models 1443 biomanufacturing 1472
Regulatory challenges for biomanufacturing 1473
Thomas Shupe, Aleksander Skardal and Anthony Food and Drug Administration guidance
Atala documents 1474
Introduction 1443 Creating standards 1475
Advanced in vitro modeling The future: envisioned advanced
systems—progression from two-dimensional biomanufacturing 1476
to three-dimensional models 1444 Closed-modular biomanufacturing systems 1476
Organ-on-a-chip technologies and their Off-the-shelf products 1477
applications 1445 Preservation advances 1477
Microengineering and biofabrication 1446 Synthetic biology advances 1477
Liver-on-a-chip 1447 Cell banking advances 1477
Vessel-on-a-chip 1447 Medical applications for biomanufacturing in
Lung-on-a-chip 1448 regenerative medicine 1477
Heart-on-a-chip 1448 Space exploration 1478
Cancer-on-a-chip 1448 References 1479
Body-on-a-chip: integrated multiorgan
systems and future applications 1449 Part Twentytwo
The importance of multiorganoid integration 1449
Cutting edge body-on-a-chip: the first highly
Clinical experience 1481
functional multiorganoid systems 1452
Conclusion and perspectives 1455
79. Tissue-engineered skin products 1483
References 1456 Jonathan Mansbridge
Introduction 1483
77. Monitoring and real-time control of Types of therapeutic tissue-engineered skin
tissue engineering systems 1459 products 1484
Jean F. Welter and Harihara Baskaran Components of tissue-engineered skin
grafts as related to function 1484
Introduction 1459 Scaffold 1484
Current state-of-the-art 1460 Keratinocytes 1485
General environmental monitoring and Fibroblasts 1485
real-time control 1460 Extracellular matrix 1485
Tissue-level monitoring 1462 Subcutaneous fat 1485
Mechanical properties 1462 Components of the immune system 1486
Cell-level monitoring 1463 Melanocytes 1486
Reporter-based gene expression imaging 1463 Adnexal structures 1487
Contents xxv

Commercial production of tissue-engineered Clinical generations of autologous


skin products 1487 chondrocyte implantation 1503
Regulation 1487 Acellular, scaffold-based products 1503
Product development 1487 Particulated autologous or allogenic articular
Overall concept 1487 cartilage 1503
Allogeneic cell source 1488 Commercial autologous chondrocyte
Viability of product and avoidance of a final implantation products 1503
sterile fill 1488 MACI (Vericel, Cambridge, MA,
Shelf life 1488 United States) 1503
Size, user convenience 1489 ChondroCelect (TiGenix, Leuven, Belgium) 1504
The manufacture of Dermagraft and Spherox (Co.don, Berlin, Germany) 1504
TransCyte 1489 Novocart 3D (Tetec, Reutlingen, Germany) 1504
Cells 1489 BioSeed C (Biotissue, Geneva, Switzerland) 1504
Medium 1489 Novocart Inject (Tetec, Reutlingen,
Bioreactor design 1490 Germany) 1504
The Dermagraft and TransCyte production Chondron (Sewon Cellontech, Seoul, Korea) 1505
processes 1490 Cartipatch (Tissue Bank of France, Génie
Release specifications 1491 Tissulaire, Lyon, France) 1505
Distribution and cryopreservation 1491 CARTISTEM (Medipost, Seongnam, Korea) 1505
Problems with commercial culture for tissue Clinical application of autologous chondrocyte
engineering 1492 implantation in reconstructive articular
Clinical trials 1492 cartilage surgery 1505
Immunological properties of Indications for autologous chondrocyte
tissue-engineered skin 1493 implantation 1505
Commercial success 1494 Contraindications 1505
Mechanism of action 1494 Surgical steps 1506
Future developments 1495 Clinical results of autologous chondrocyte
Conclusion 1496 implantation 1506
References 1496 Overview 1506
Data from prospective randomized clinical
80. Tissue-engineered cartilage trials 1507
products 1499 Long-term results of autologous chondrocyte
implantation 1508
Henning Madry Clinical factors affecting the clinical
Introduction 1499 outcomes of autologous chondrocyte
Cartilage defects, osteoarthritis, and implantation 1508
reconstructive surgical options 1499 Conflict of interest 1509
Cartilage defects pathophysiology 1499 References 1509
Surgical treatment options for articular
cartilage defects 1500 81. Bone tissue engineering 1511
Tissue-engineered cartilage products for
Hani A. Awad, Regis J. O’Keefe and Jeremy J. Mao
orthopedic reconstruction 1500
Cells for tissue-engineered cartilage repair 1500 Introduction 1511
Scaffolds for clinical tissue-engineered Conventional bone tissue engineering
cartilage repair 1501 strategies: cells, scaffolds, and biofactors 1511
Collagen scaffolds 1501 Delivery of molecules and/or scaffolds to
Hyaluronan 1502 augment endogenous bone regeneration 1512
Synthetic polymers 1502 Biomaterials development and
Agarose and alginate 1502 three-dimensional printing 1513
Scaffold-free three-dimensional systems 1502 Clinical successes and opportunities in
Bioreactors for tissue-engineered cartilage regenerative repair of craniofacial defects 1516
repair 1502 Conclusion 1517
Clinical nomenclature of scaffold-based Acknowledgments 1517
techniques 1503 References 1517
xxvi Contents

82. Tissue-engineered cardiovascular Introduction 1553


products 1521 Regulatory background 1553
Overview of development and approval
Doris A. Taylor, Camila Hochman-Mendez, Joern process 1554
Huelsmann, Abdelmotagaly Elgalad and Early-stage development 1554
Luiz C. Sampaio Chemistry, manufacturing, and controls 1555
Clinical situation/reality 1521 Pharmacology and toxicology 1555
Considerations for tissue-engineered Clinical 1556
cardiovascular constructs 1521 US Food and Drug Administration/sponsor
Components for tissue-engineered meetings 1557
cardiovascular constructs 1521 Submitting an investigational new drug
Cell sources 1521 application 1557
Scaffolds 1524 Required US Food and Drug Administration
Tissue-engineered cardiovascular constructs 1525 forms 1557
Vascular grafts 1525 Investigational new drug application
Valves 1526 contents 1558
Cardiac patches 1527 US Food and Drug Administration review
Building the next level of complexity: whole of an original investigational new drug
heart 1529 application submission 1559
Pathway to approval and commercialization 1530 Later-stage development topics 1559
Future perspectives 1532 Compliance with current good
References 1532 manufacturing practice 1559
Product readiness for Phase 3 1559
83. Tissue organoid models and Potency assay 1560
Pharmacology and toxicology 1560
applications 1537
Phase 3 clinical development 1560
Timothy S. Leach, Anthony Dominijanni, Combination products 1561
Sean V. Murphy and Anthony Atala Tissue-engineered and regenerative medicine
products 1562
Introduction 1537
3D bio-printed tissue-engineered/
Cell sources 1537
regenerative-medicine products 1563
Types of organoid models 1538
Medical devices 1563
Cardiac organoid 1539
Least burdensome principles 1563
Liver organoid 1540
Breakthrough device program 1563
Brain organoid 1540
Evaluation of devices used with regenerative
Lung organoid 1541
medicine advanced therapy 1564
Gastrointestinal tract organoid 1541
Expedited review programs 1564
Other organoid models 1542
Other regulatory topics 1565
Applications 1542
Minimal manipulation and homologous
Tumor and disease models 1542
use of human cells, tissues, and cellular and
Drug analysis 1543
tissue-based products 1565
Organ-on-a-chip 1544
Clinical research involving children 1566
Developmental biology 1544
Expanded access to investigational drugs for
Conclusion 1545
treatment use 1566
References 1545
Charging for investigational drugs under an
investigational new drug application 1566
Part Twenty three Responsibilities of sponsors and investigators 1566
Regulation, commercialization Clinical research conducted outside of the
United States 1568
and ethics 1551 Use of standards 1568
US Food and Drug Administration
84. The regulatory process from concept to international regulatory activities 1568
market 1553 The role of cell-based products in medical
Kyung Eun Sung, Judith Arcidiacono, Donald W. product testing 1568
Fink Jr., Andrea Gray, Johnny Lam, Winson Tang, Conclusion 1568
Iwen Wu and Raj K. Puri Acknowledgments 1568
Contents xxvii

Appendix I: Code of Federal Regulations 86. Ethical issues 1585


citations relevant to cellular product
development 1569 Laurie Zoloth
Appendix II: The list of acronyms 1569 Introduction 1585
References 1570 Duty and healing: natural makers in a broken
world 1587
85. Business issues 1573 To make is to know: notes on an old problem
about knowledge 1587
Matthew Vincent
What is a thing? The perils of deconstruction 1588
Introduction 1573 What contextual factors should be taken into
The aging population 1573 account, and do any of these prevent the
Rise of regenerative medicine 1575 development and use of the technology? 1588
Product development 1577 What purposes, techniques, or applications
Embryonic stem cells 1578 would be permissible and under what
Induced pluripotent stem cells 1579 circumstances? 1589
Direct reprogramming of differentiated cells 1580 On what procedures and structures, involving
Small molecule-induced differentiation 1580 what policies, should decisions on
Reimbursement 1580 appropriate techniques and uses be based? 1590
Conclusion 1582 Conclusion 1590
References 1582 References 1590
Index 1593
List of contributors

Masashi Abe Astellas Institute for Regenerative Nureddin Ashammakhi Center for Minimally Invasive
Medicine, Westborough, MA, United States Therapeutics, University of California, Los Angeles,
Los Angeles, CA, United States; Department of
Jon D. Ahlstrom PolarityTE, Salt Lake City, UT, United
Bioengineering, University of California, Los Angeles,
States
Los Angeles, CA, United States; Department of
Julie Albon School of Optometry and Vision Sciences, Radiological Sciences, David Geffen School of
Cardiff University, Cardiff, United Kingdom Medicine, University of California, Los Angeles, Los
Angeles, CA, United States
Julie Allickson Wake Forest Institute for Regenerative
Medicine, Wake Forest School of Medicine, Winston- Anthony Atala Wake Forest Institute for Regenerative
Salem, NC, United States Medicine, Wake Forest University, Winston-Salem,
Graça Almeida-Porada Wake Forest Institute for NC, United States
Regenerative Medicine, Fetal Research and Therapy Kyriacos A. Athanasiou Department of Biomedical
Program, Wake Forest School of Medicine, Winston Engineering, University of California, Irvine, CA,
Salem, NC, United States United States
Richard A. Altschuler Department of Otolaryngology, Hani A. Awad Department of Biomedical Engineering,
Kresge Hearing Research Institute, University of The Center for Musculoskeletal Research, University
Michigan, Ann Arbor, MI, United States; Department of Rochester, Rochester, NY, United States
of Cell and Developmental Biology, Kresge Hearing Stephen F Badylak McGowan Institute for Regenerative
Research Institute, University of Michigan, Ann Medicine, University of Pittsburgh, Pittsburgh, PA,
Arbor, MI, United States; VA Ann Arbor Health Care United States; Department of Surgery, University of
System, Ann Arbor, MI, United States Pittsburgh School of Medicine, Pittsburgh, PA, United
Daniel G. Anderson Department of Chemical States; Department of Bioengineering, University of
Engineering, Massachusetts Institute of Technology, Pittsburgh, Pittsburgh, PA, United States
Cambridge, MA, United States; David H Koch Gowri Balachander National University of Singapore,
Institute for Integrative Cancer Research, Singapore, Singapore
Massachusetts Institute of Technology, Cambridge,
Wayne Balkan Interdisciplinary Stem Cell Institute,
MA, United States; Department of Anesthesiology,
Miller School of Medicine, University of Miami,
Boston Children’s Hospital, Boston, MA, United
Miami, FL, United States; Department of Medicine,
States; Division of Health Science Technology,
Miller School of Medicine, University of Miami,
Massachusetts Institute of Technology, Cambridge,
Miami, FL, United States
MA, United States; Institute for Medical Engineering
and Science, Massachusetts Institute of Technology, Jennifer J. Bara Center of Regenerative Medicine,
Cambridge, MA, United States Washington University, St. Louis, MO, United States

Nasim Annabi Department of Chemical Engineering, Michael P. Barry Second Sight Medical Products, Los
University of California, Los Angeles, Los Angeles, Angeles, CA, United States
CA, United States Harihara Baskaran Department of Chemical
Engineering, Case Western Reserve University,
Judith Arcidiacono Office of Tissues and Advanced Cleveland, OH, United States; Case Center for
Therapies, Center for Biologics Evaluation and Multimodal Evaluation of Tissue Engineered
Research, Food and Drug Administration, Silver Cartilage, Cleveland, OH, United States
Spring, MD, United States

xxix
xxx List of contributors

Matthew L. Bedell Department of Bioengineering, Rice Boston Children’s Hospital, Boston, MA, United
University, Houston, TX, United States States
Donald Andrew Belcher William G. Lowrie Department Lawrence J. Bonassar Meinig School of Biomedical
of Chemical and Biomolecular Engineering, The Ohio Engineering, Sibley School of Mechanical and
State University, OH, United States Aerospace Engineering, Cornell University, Ithaca,
David B. Berry Department of NanoEngineering, NY, United States
University of California, San Diego, La Jolla, CA, Joseph V. Bonventre Renal Division, Brigham and
United States Women’s Hospital, Department of Medicine, Harvard
Hina Bhat Division of Biotechnology, Faculty of Medical School, Boston, MA, United States
Veterinary Sciences and Animal Husbandry, SKUAST
Mimi R. Borrelli Hagey Laboratory for Pediatric
of Kashmir, Srinagar, India
Regenerative Medicine, Division of Plastic and
Zuhaib F. Bhat Department of Wine Food and Molecular Reconstructive Surgery, Department of Surgery,
Biosciences, Faculty of Agriculture and Life Sciences, Stanford University School of Medicine, Stanford,
Lincoln University, Lincoln, New Zealand CA, United States
Sangeeta N. Bhatia David H. Koch Institute for Robby D. Bowles Department Bioengineering, University
Integrative Cancer Research, Massachusetts Institute of Utah, Salt Lake City, UT, United States
of Technology, Cambridge, MA, United States;
Harvard-MIT Health Sciences and Technology, Amy D. Bradshaw Deptartment of Medicine, Medical
Massachusetts Institute of Technology, Cambridge, University of South Carolina, Charleston, SC, United
MA, United States; Institute for Medical Engineering States; The Ralph H. Johnson Department of Veteran’s
and Science, Massachusetts Institute of Technology, Affair Medical Center, Charleston, SC, United States
Cambridge, MA, United States; Wyss Institute for Andres M. Bratt-Leal Department of Molecular
Biologically Inspired Engineering, Boston, MA, Medicine, The Scripps Research Institute, La Jolla,
United States; Department of Electrical Engineering CA, United States; Aspen Neuroscience, Inc., San
and Computer Science, Massachusetts Institute of Diego, CA, United States
Technology, Cambridge, MA, United States; Howard Christopher K. Breuer Center for Regenerative
Hughes Medical Institute, Chevy Chase, MD, United Medicine, Nationwide Children’s Hospital, Columbus,
States OH, United States
Catherine Clare Blackburn MRC Centre for Luke Brewster Department of Surgery, Emory
Regenerative Medicine, Institute for Stem Cell University School of Medicine, Atlanta, GA, United
Research, School of Biological Sciences, University of States; Georgia Institute of Technology, Parker H.
Edinburgh, Edinburgh, United Kingdom Petit Institute for Bioengineering and Biosciences,
Anna Blocki Institute for Tissue Engineering and Atlanta, GA, United States; Atlanta VA Hospital,
Regenerative Medicine, The Chinese University of Decatur, GA, United States
Hong Kong, Hong Kong SAR, P.R. China; School of Eric M. Brey Surgical and Research Services, Edward J.
Biomedical Sciences, Faculty of Medicine, The Hines, Jr. VA Hospital, Hines, IL, United States;
Chinese University of Hong Kong, Hong Kong SAR, Department of Biomedical Engineering, University of
P.R. China Texas at San Antonio, San Antonio, TX, United States
Kevin M. Blum Center for Regenerative Medicine, Priscilla S. Briquez Pritzker School of Molecular
Nationwide Children’s Hospital, Columbus, OH, Engineering, University of Chicago, Chicago, IL,
United States; Department of Biomedical Engineering, United States
The Ohio State University, Columbus, OH, United
States J.A. Buckwalter Department of Orthopedics and
Rehabilitation, Iowa City Veterans Administration
Matthew A. Bochenek Department of Chemical Medical Center, University of Iowa College of
Engineering, Massachusetts Institute of Technology, Medicine, Iowa City, IA, United States
Cambridge, MA, United States; David H Koch
Institute for Integrative Cancer Research, Karen J.L. Burg Department of Small Animal Medicine
Massachusetts Institute of Technology, Cambridge, and Surgery, University of Georgia, Athens, GA,
MA, United States; Department of Anesthesiology, United States
List of contributors xxxi

Timothy C. Burg Department of Veterinary Biosciences Medical Center, University of Iowa College of
and Diagnostic Imaging, University of Georgia, Medicine, Iowa City, IA, United States
Athens, GA, United States
George Cotsarelis Department of Dermatology, Kligman
Batzaya Byambaa 3D BioLabs, LLC, Cambridge, MA, Laboratories, University of Pennsylvania School of
United States Medicine, Philadelphia, Pennsylvania, United States
Prafulla K. Chandra Wake Forest Institute for Ronald G. Crystal Department of Genetic Medicine,
Regenerative Medicine, Wake Forest School of Weill Medical College of Cornell University, New
Medicine, Winston Salem, NC, United States York, NY, United States
Amanda X. Chen Department of Biological Engineering, Gislin Dagnelie Department of Ophthalmology, Johns
Massachusetts Institute of Technology, Cambridge, Hopkins University, Baltimore, MD, United States
MA, United States; David H. Koch Institute for
Integrative Cancer Research, Massachusetts Institute Mohammad Ali Darabi Center for Minimally Invasive
of Technology, Cambridge, MA, United States Therapeutics, University of California, Los Angeles,
Los Angeles, CA, United States; Department of
Fa-Ming Chen State Key Laboratory of Military
Bioengineering, University of California, Los Angeles,
Stomatology and National Clinical Research Center
Los Angeles, CA, United States
for Oral Diseases, Department of Periodontology,
School of Stomatology, Fourth Military Medical Jeffrey M. Davidson Department of Pathology,
University, Xi’an, P.R. China Microbiology and Immunology, Vanderbilt University
Medical Center, Nashville, TN, United States
Shaochen Chen Department of NanoEngineering,
University of California, San Diego, La Jolla, CA, Joseph Davidson Stem Cell and Regenerative Medicine
United States; Department of Bioengineering, Section, Great Ormond Street Institute of Child
University of California, San Diego, La Jolla, CA, Health, University College London, London, United
United States; Materials Science and Engineering Kingdom
Program, University of California, San Diego, La Paolo De Coppi Stem Cell and Regenerative Medicine
Jolla, CA, United States; Chemical Engineering Section, Great Ormond Street Institute of Child
Program, University of California, San Diego, La Health, University College London, London, United
Jolla, CA, United States Kingdom
Julian Chesterman New Jersey Center for Biomaterials, Derfogail Delcassian Department of Chemical
Rutgers, The State University of New Jersey, Engineering, Massachusetts Institute of Technology,
Piscataway, NJ, United States Cambridge, MA, United States; David H Koch
Arnav Chhabra David H. Koch Institute for Integrative Institute for Integrative Cancer Research,
Cancer Research, Massachusetts Institute of Massachusetts Institute of Technology, Cambridge,
Technology, Cambridge, MA, United States; Harvard- MA, United States; Division of Regenerative
MIT Health Sciences and Technology, Massachusetts Medicine and Cellular Therapies, School of Pharmacy,
Institute of Technology, Cambridge, MA, United University of Nottingham, Nottingham, United
States; Institute for Medical Engineering and Science, Kingdom
Massachusetts Institute of Technology, Cambridge, Paul de Vos Section of Immunoendocrinology,
MA, United States Department of Pathology and Medical Biology,
Seow Khoon Chong Nanyang Technological University, University Medical Center Groningen, University of
Singapore, Singapore Groningen, Groningen, The Netherlands
Richard A.F. Clark Departments of Biomedical Anthony Dominijanni Wake Forest School of Medicine,
Engineering, Stony Brook University, Stony Brook, Wake Forest Institute for Regenerative Medicine,
NY, United States; Dermatology and Medicine, Stony Winston-Salem, NC, United States
Brook University, Stony Brook, NY, United States Ryan Donahue Department of Biomedical Engineering,
Muriel A. Cleary University of Massachusetts Medical University of California, Irvine, CA, United States
School, Worcester, MA, United States Allison P. Drain Center for Bioengineering and Tissue
M. Coleman Department of Orthopedics and Regeneration, Department of Surgery, University of
Rehabilitation, Iowa City Veterans Administration California, San Francisco, CA, United States
xxxii List of contributors

Craig L. Duvall Department of Biomedical Engineering, Sharon Gerecht Department of Chemical and
Vanderbilt University, Nashville, TN, United States Biomolecular Engineering, The Institute for
Jenna L. Dziki McGowan Institute for Regenerative NanoBioTechnology, Physical Sciences-Oncology
Medicine, University of Pittsburgh, Pittsburgh, PA, Center, Johns Hopkins University, Baltimore, MD,
United States; Department of Surgery, University of United States
Pittsburgh School of Medicine, Pittsburgh, PA, United Charles A. Gersbach Department of Biomedical
States Engineering, Duke University, Durham, NC, United
Abdelmotagaly Elgalad Regenerative Medicine States
Research, Texas Heart Institute, Houston, TX, United D.M.R. Gibbs Bone & Joint Research Group, Centre for
States Human Development, Stem Cells and Regeneration,
George Eng Department of Biomedical Engineering, Human Development and Health, Faculty of
Columbia University, New York, NY, United States; Medicine, University of Southampton, Southampton,
College of Physicians and Surgeons, Columbia United Kingdom
University, New York, NY, United States
Simran Gidwani Interdisciplinary Stem Cell Institute,
Vincent Falanga Department of Dermatology, Boston Miller School of Medicine, University of Miami,
University School of Medicine, Boston, MA, United Miami, FL, United States
States; Department of Biochemistry, Boston
University School of Medicine, Boston, MA, United Shaimar R. González Morales Cell Biology Section,
States; Wound Biotechnology Foundation, Boston, National Institute of Dental and Craniofacial
MA, United States Research, National Institutes of Health, Bethesda,
MD, United States; Greehey Children’s Cancer
Niloofar Farhang Department Bioengineering, Research Institute, UT Health Science Center at San
University of Utah, Salt Lake City, UT, United States Antonio, San Antonio, TX, United States; Department
Lino Ferreira Faculty of Medicine, Coimbra and Center of Cell Systems & Anatomy, UT Health Science
for Neurosciences and Cell Biology, University of Center at San Antonio, San Antonio, TX, United
Coimbra, Coimbra, Portugal States
Donald W. Fink, Jr. Division of Cellular and Gene Ritu Goyal New Jersey Center for Biomaterials, Rutgers,
Therapies, Office of Tissues and Advanced Therapies, The State University of New Jersey, Piscataway, NJ,
Center for Biologics Evaluation and Research, Food United States
and Drug Administration, Silver Spring, MD, United
States Maria B. Grant Department of Ophthalmology and
Visual Sciences, University of Alabama at
Heather E. Fleming David H. Koch Institute for Birmingham, Birmingham, AL, United States
Integrative Cancer Research, Massachusetts Institute
of Technology, Cambridge, MA, United States; Andrea Gray Division of Cellular and Gene Therapies,
Harvard-MIT Health Sciences and Technology, Office of Tissues and Advanced Therapies, Center for
Massachusetts Institute of Technology, Cambridge, Biologics Evaluation and Research, Food and Drug
MA, United States Administration, Silver Spring, MD, United States
Peter Fong Flagship Pioneering, Cambridge, MA, United Howard P. Greisler Cell Biology, Neurobiology, &
States Anatomy, Departments of Surgery, Loyola University
Mark R. Frey Department of Pediatrics, Division of Medical Center, Maywood, IL, United States
Gastroenterology, Hepatology and Nutrition, Tracy C. Grikscheit Developmental Biology and
Children’s Hospital Los Angeles, Los Angeles, CA, Regenerative Medicine Program, The Saban Research
United States; Department of Biochemistry and Institute, Children’s Hospital Los Angeles, Los
Molecular Biology, University of Southern California Angeles, CA, United States; Department of Surgery,
Keck School of Medicine, Los Angeles, CA, United Division of Pediatric Surgery, Children’s Hospital Los
States; The Saban Research Institute, Children’s Angeles, Los Angeles, CA, United States
Hospital Los Angeles, Los Angeles, CA, United States Karl Grosh Department of Mechanical Engineering,
Denise Gay Department of Dermatology, Kligman University of Michigan, Ann Arbor, MI, United
Laboratories, University of Pennsylvania School of States; Department of Biomedical Engineering,
Medicine, Philadelphia, Pennsylvania, United States University of Michigan, Ann Arbor, MI, United States
List of contributors xxxiii

Farshid Guilak Department of Orthopaedic Surgery, Camila Hochman-Mendez Regenerative Medicine


Washington University, St. Louis, MO, United States; Research, Texas Heart Institute, Houston, TX, United
Shriners Hospitals for Children—St. Louis, St. Louis, States
MO, United States; Center of Regenerative Medicine, Chao Huang Department of Ophthalmology and Visual
Washington University, St. Louis, MO, United States Sciences, University of Alabama at Birmingham,
Jason L. Guo Department of Bioengineering, Rice Birmingham, AL, United States
University, Houston, TX, United States Jeffrey A. Hubbell Pritzker School of Molecular
Yingli Han Center of Stem Cell and Regenerative Engineering, University of Chicago, Chicago, IL,
Medicine, and Bone Marrow Transplantation Center United States
of the First Affiliated Hospital, Zhejiang University Joern Huelsmann Regenerative Medicine Research,
School of Medicine, Hangzhou, P.R. China; Institute Texas Heart Institute, Houston, TX, United States
of Hematology, Zhejiang University & Zhejiang Jun Tae Huh Wake Forest Institute for Regenerative
Engineering Laboratory for Stem Cell and Medicine, Wake Forest School of Medicine, Winston-
Immunotherapy, Hangzhou, P.R. China Salem, NC, United States
Joshua M. Hare Interdisciplinary Stem Cell Institute, Joshua G. Hunsberger Regenerative Medicine
Miller School of Medicine, University of Miami, Manufacturing Society, Winston-Salem, NC, United States
Miami, FL, United States; Department of Medicine,
Leanne E. Iannucci Department of Biomedical
Miller School of Medicine, University of Miami,
Engineering, Washington University in St. Louis, St.
Miami, FL, United States
Louis, MO, United States
Ammar Mansoor Hassanbhai Nanyang Technological
Haruhisa Inoue Center for iPS Cell Research and
University, Singapore, Singapore
Application (CiRA), Kyoto University, Kyoto, Japan;
Konstantinos Hatzistergos Interdisciplinary Stem Cell iPSC-based Drug Discovery and Development Team,
Institute, Miller School of Medicine, University of RIKEN BioResource Research Center (BRC), Kyoto,
Miami, Miami, FL, United States; Department of Cell Japan; Medical-risk Avoidance based on iPS Cells
Biology and Physiology, Miller School of Medicine, Team, RIKEN Center for Advanced Intelligence
University of Miami, Miami, FL, United States Project (AIP), Kyoto, Japan
David C. Hay MRC Centre for Regenerative Medicine, John Jackson Wake Forest Institute for Regenerative
University of Edinburgh, United Kingdom Medicine, Wake Forest School of Medicine, Winston-
Salem, NC, United States
Xiao-Tao He State Key Laboratory of Military
Stomatology and National Clinical Research Center Yangzi Jiang Institute for Tissue Engineering and
for Oral Diseases, Department of Periodontology, Regenerative Medicine, The Chinese University of Hong
School of Stomatology, Fourth Military Medical Kong, Hong Kong SAR, P.R. China; School of
University, Xi’an, P.R. China Biomedical Sciences, Faculty of Medicine, The Chinese
University of Hong Kong, Hong Kong SAR, P.R. China
Timothy Henderson MRC Centre for Regenerative
Medicine, Institute for Stem Cell Research, School of Vladimir V. Kalinichenko Division of Neonatology,
Biological Sciences, University of Edinburgh, Perinatal and Pulmonary Biology, Cincinnati
Edinburgh, United Kingdom Children’s Hospital Medical Center, Perinatal
Institute, University of Cincinnati College of
Darren Hickerson Wake Forest Institute for Medicine, Cincinnati, OH, United States
Regenerative Medicine, Wake Forest School of
J.M. Kanczler Bone & Joint Research Group, Centre for
Medicine, Winston-Salem, NC, United States
Human Development, Stem Cells and Regeneration,
Darren H.M. Hickerson Wake Forest Institute for Human Development and Health, Faculty of
Regenerative Medicine, Winston-Salem, NC, United Medicine, University of Southampton, Southampton,
States United Kingdom
Abdelkrim Hmadcha Andalusian Center for Molecular Jeffrey M. Karp Harvard-Massachusetts Institute of
Biology and Regenerative Medicine (CABIMER), Technology Division of Health Sciences and
University of Pablo de Olavide-University of Seville- Technology, Massachusetts Institute of Technology,
CSIC, Sevilla, Spain; Spanish Biomedical Research Cambridge, MA, United States; Department of
Centre in Diabetes and Associated Metabolic Medicine, Brigham and Women’s Hospital, Harvard
Disorders (CIBERDEM), Madrid, Spain Medical School, Boston, MA, United States
xxxiv List of contributors

F. Kurtis Kasper Department of Orthodontics, Wake Forest School of Biomedical Engineering and
University of Texas Health Science Center Sciences, Wake Forest School of Medicine, Winston-
Houston, Houston, TX, United States Salem, NC, United States
Ali Khademhosseini Center for Minimally Invasive Benjamin W. Lee Department of Biomedical
Therapeutics, University of California, Los Angeles, Engineering, Columbia University, New York, NY,
Los Angeles, CA, United States; Department of United States; College of Physicians and Surgeons,
Bioengineering, University of California, Los Angeles, Columbia University, New York, NY, United States
Los Angeles, CA, United States; Department of Iris Lee Bioengineering, University of Pennsylvania
Chemical Engineering, University of California, Los School of Engineering, Philadelphia, PA, United States
Angeles, Los Angeles, CA, United States; Department
of Radiological Sciences, David Geffen School of Sang Jin Lee Wake Forest Institute for Regenerative
Medicine, University of California, Los Angeles, Los Medicine, Wake Forest School of Medicine, Winston-
Angeles, CA, United States; California NanoSystems Salem, NC, United States
Institute (CNSI), University of California, Los David Li Department of Biomedical Engineering,
Angeles, Los Angeles, CA, United States Carnegie Mellon University, Pittsburgh, PA, United
Ji Hyun Kim Wake Forest Institute for Regenerative States
Medicine, Wake Forest School of Medicine, Winston- Linheng Li Stowers Institute for Medical Research,
Salem, NC, United States Kansas City, MO, United States
Erin A. Kimbrel Astellas Institute for Regenerative Qian Liu Department of Orthopaedics, The Second
Medicine, Westborough, MA, United States Xiangya Hospital, Central South University,
Irina Klimanskaya Astellas Institute for Regenerative Changsha, P.R. China
Medicine, Westborough, MA, United States Alexander Ljubimov Department of Biomedical
Joachim Kohn New Jersey Center for Biomaterials, Sciences, Cedars-Sinai Medical Center, Los Angeles,
Rutgers, The State University of New Jersey, CA, United States
Piscataway, NJ, United States Chi Lo Wake Forest Institute for Regenerative Medicine,
Sunil Kumar Division of Livestock Products Wake Forest School of Medicine, Winston-Salem,
Technology, Faculty of Veterinary Sciences and NC, United States
Animal Husbandry, SKUAST of Jammu, Jammu, Michael T. Longaker Hagey Laboratory for Pediatric
India Regenerative Medicine, Division of Plastic and
Themis R. Kyriakides Department of Pathology, Yale Reconstructive Surgery, Department of Surgery,
University, New Haven, CT, United States Stanford University School of Medicine, Stanford,
CA, United States
Spencer P. Lake Department of Mechanical Engineering
& Materials Science, Washington University in St. Javier López-Beas Andalusian Center for Molecular
Louis, St. Louis, MO, United States Biology and Regenerative Medicine (CABIMER),
University of Pablo de Olavide-University of Seville-
Johnny Lam Division of Cellular and Gene Therapies, CSIC, Sevilla, Spain
Office of Tissues and Advanced Therapies, Center
for Biologics Evaluation and Research, Food and Jeanne F. Loring Department of Molecular Medicine,
Drug Administration, Silver Spring, MD, United The Scripps Research Institute, La Jolla, CA, United
States States; Aspen Neuroscience, Inc., San Diego, CA,
United States
Robert Langer Koch Institute for Integrative Cancer
Research, Massachusetts Institute of Technology, Ying Luo Lyndra Therapeutics, Watertown, MA, United
Cambridge, MA, United States States
Robert Lanza Astellas Institute for Regenerative Ben D. MacArthur Faculty of Medicine, School of
Medicine, Westborough, MA, United States; Institute Mathematics & Institute for Life Sciences, University
for Regenerative Medicine, Wake Forest University of Southampton, Southampton, United Kingdom
School of Medicine, Winston-Salem, NC, United Nicolas N. Madigan Department of Neurology,
States Regenerative Neurobiology Laboratory, Mayo Clinic,
Timothy S. Leach Wake Forest School of Medicine, Rochester, MN, United States
Wake Forest Institute for Regenerative Medicine, Henning Madry Center of Experimental Orthopaedics,
Winston-Salem, NC, United States; Virginia Tech- Saarland University, Homburg, Germany
List of contributors xxxv

Renata S. Magalhaes Wake Forest Institute for University of Texas at Austin, Austin, TX, United
Regenerative Medicine, Wake Forest University School States
of Medicine, Winston-Salem, NC, United States
Maria Mirotsou Astellas Institute for Regenerative
Nancy Ruth Manley Department of Genetics, University Medicine, Westborough, MA, United States
of Georgia, Athens, GA, United States
Daniel T. Montoro Hagey Laboratory for Pediatric
Jonathan Mansbridge California Way, Woodside, Regenerative Medicine, Division of Plastic and
California, United States Reconstructive Surgery, Department of Surgery,
Jeremy J. Mao Center for Craniofacial Regeneration, Stanford University School of Medicine, Stanford,
Columbia University Medical Center, New York, NY, CA, United States
United States; Department of Pathology and Cell
Matthew P. Murphy Hagey Laboratory for Pediatric
Biology, Columbia University, New York, NY, United
Regenerative Medicine, Division of Plastic and
States; Department of Orthopedic Surgery, Columbia
Reconstructive Surgery, Department of Surgery,
University Physician and Surgeons, New York, NY,
Stanford University School of Medicine, Stanford,
United States; Department of Biomedical Engineering,
CA, United States
Columbia University, New York, NY, United States
K.M. Marshall Bone & Joint Research Group, Centre for Sean V. Murphy Wake Forest School of Medicine,
Human Development, Stem Cells and Regeneration, Wake Forest Institute for Regenerative Medicine,
Human Development and Health, Faculty of Winston-Salem, NC, United States
Medicine, University of Southampton, Southampton, Michael Musillo Departments of Biomedical
United Kingdom Engineering, Stony Brook University, Stony Brook,
J.A. Martin Department of Orthopedics and NY, United States
Rehabilitation, Iowa City Veterans Administration Padmalosini Muthukumaran Nanyang Technological
Medical Center, University of Iowa College of University, Singapore, Singapore
Medicine, Iowa City, IA, United States
Adam M. Navara Department of Bioengineering, Rice
M. Martins-Green Department of Molecular, Cell and
University, Houston, TX, United States
Systems Biology, University of California, Riverside,
CA, United States Christopher E. Nelson Department of Biomedical
Engineering, University of Arkansas, Fayetteville, AR,
Kathryn M. Maselli Developmental Biology and
United States
Regenerative Medicine Program, The Saban Research
Institute, Children’s Hospital Los Angeles, Los Laura E. Niklason Department of Biomedical
Angeles, CA, United States; Department of Surgery, Engineering, Yale University, New Haven, CT, United
Division of Pediatric Surgery, Children’s Hospital Los States; Department of Anesthesiology, Yale
Angeles, Los Angeles, CA, United States University, New Haven, CT, United States
Mark W. Maxfield University of Massachusetts Medical Craig Scott Nowell MRC Centre for Regenerative
School, Worcester, MA, United States Medicine, Institute for Stem Cell Research, School of
Biological Sciences, University of Edinburgh,
Kyle W. McCracken Division of Pediatric Nephrology,
Edinburgh, United Kingdom
Boston Children’s Hospital, Boston, MA, United
States; Renal Division, Brigham and Women’s Regis J. O’Keefe Department of Orthopaedic Surgery,
Hospital, Department of Medicine, Harvard Medical Washington University School of Medicine, St. Louis,
School, Boston, MA, United States MO, United States
Kathy E. O’Neill MRC Centre for Regenerative
James Melville Department of Oral and Maxillofacial
Medicine, Institute for Stem Cell Research, School of
Surgery, University of Texas Health Science Center
Biological Sciences, University of Edinburgh,
Houston, Houston, TX, United States
Edinburgh, United Kingdom
Antonios G. Mikos Department of Bioengineering, Rice
Richard O.C. Oreffo Bone & Joint Research Group,
University, Houston, TX, United States
Centre for Human Development, Stem Cells and
José del R. Millán Department of Electrical and Regeneration, Human Development and Health,
Computer Engineering, University of Texas at Austin, Faculty of Medicine, University of Southampton,
Austin, TX, United States; Department of Neurology, Southampton, United Kingdom
xxxvi List of contributors

Ophir Ortiz New Jersey Center for Biomaterials, Paul Rouse MRC Centre for Regenerative Medicine, Institute
Rutgers, The State University of New Jersey, for Stem Cell Research, School of Biological Sciences,
Piscataway, NJ, United States University of Edinburgh, Edinburgh, United Kingdom
Andre Francis Palmer William G. Lowrie Department Hooman Sadri-Ardekani Wake Forest Institute for
of Chemical and Biomolecular Engineering, The Ohio Regenerative Medicine, Wake Forest School of
State University, OH, United States Medicine, Winston-Salem, NC, United States;
Serafeim Perdikis Brain Computer Interfaces and Department of Urology, Wake Forest School of
Neural Engineering Laboratory, School of Computer Medicine, Winston-Salem, NC, United States
Science and Electronic Engineering, University of W. Mark Saltzman Deaprtment of Biomedical
Essex, Colchester, United Kingdom Engineering, Yale University, New Haven, CT, United
M. Petreaca Department of Biology, DePauw University, States
Greencastle, IN, United States Luiz C. Sampaio Regenerative Medicine Research,
Maksim V. Plikus Department of Developmental and Texas Heart Institute, Houston, TX, United States
Cell Biology, Sue and Bill Gross Stem Cell Research Christopher R. Schlieve Developmental Biology and
Center, University of California, Irvine, CA, United Regenerative Medicine Program, The Saban Research
States Institute, Children’s Hospital Los Angeles, Los
Christopher D. Porada Wake Forest Institute for Angeles, CA, United States; Department of Surgery,
Regenerative Medicine, Fetal Research and Therapy Division of Pediatric Surgery, Children’s Hospital Los
Program, Wake Forest School of Medicine, Winston Angeles, Los Angeles, CA, United States
Salem, NC, United States Su-Hua Sha Department of Pathology & Laboratory
Mark Post Department of Physiology, Maastricht Medicine, Medical University of South Carolina,
University, Maastricht, The Netherlands Charleston, SC, United States

Aleš Prokop Department of Chemical and Biomolecular Paul T. Sharpe Centre for Craniofacial and Regenerative
Engineering, Vanderbilt University, Nashville, TN, Biology, Faculty of Dentistry, Oral & Craniofacial
United States Sciences, King’s College London, London, United
Kingdom
Raj K. Puri Division of Cellular and Gene Therapies,
Office of Tissues and Advanced Therapies, Center for V. Prasad Shastri Institute for Macromolecular
Biologics Evaluation and Research, Food and Drug Chemistry and Centre for Biological Signalling
Administration, Silver Spring, MD, United States Studies, University of Freiburg, Freiburg, Germany
Pengxu Qian Center of Stem Cell and Regenerative Yanhong Shi Division of Stem Cell Biology Research,
Medicine, and Bone Marrow Transplantation Center Department of Developmental and Stem Cell Biology,
of the First Affiliated Hospital, Zhejiang University Beckman Research Institute of City of Hope, Duarte,
School of Medicine, Hangzhou, P.R. China; Institute CA, United States
of Hematology, Zhejiang University & Zhejiang Thomas Shupe Wake Forest Institute for Regenerative
Engineering Laboratory for Stem Cell and Medicine, Wake Forest School of Medicine, Medical
Immunotherapy, Hangzhou, P.R. China; Dr. Li Dak Center Boulevard, Winston-Salem, NC, United States
Sum & Yip Yio Chin Center for Stem Cell and Dario Sirabella Department of Biomedical Engineering,
Regenerative Medicine, Zhejiang University, Columbia University, New York, NY, United States;
Hangzhou, P.R. China Department of Medicine, Columbia University, New
Milica Radisic Department of Chemical Engineering and York, NY, United States
Applied Chemistry, University of Toronto, Toronto, Aleksander Skardal The Ohio State University College
ON, Canada of Engineering, Columbus, OH, United States
Micha Sam Brickman Raredon Department of J.M.W. Slack Department of Biology and Biochemistry,
Biomedical Engineering, Yale University, New University of Bath, Bath, United Kingdom
Haven, CT, United States
Stephen R. Sloan, Jr. Meinig School of Biomedical
Ellen Rothman Richie Department of Epigenetics and Engineering, Sibley School of Mechanical and
Molecular Carcinogenesis, University of Texas MD Aerospace Engineering, Cornell University, Ithaca,
Anderson Cancer Center, Smithville, TX, United States NY, United States
List of contributors xxxvii

Shay Soker Wake Forest Institute for Regenerative Winson Tang Division of Clinical Evaluation and
Medicine, Wake Forest School of Medicine, Winston Pharmacology/Toxicology, Office of Tissues and
Salem, NC, United States Advanced Therapies, Center for Biologics Evaluation
and Research, Food and Drug Administration, Silver
Bernat Soria Department of Physiology, School of
Spring, MD, United States
Medicine, University Miguel Hernandez, Alicante,
Spain; Institute of Bioengineering Avenida de la Doris A. Taylor Regenerative Medicine Research, Texas
Universidad s/n, Alicante, Spain; Department of Heart Institute, Houston, TX, United States
Regenerative Medicine, University Pablo de Olavide, Yao Teng National University of Singapore, Singapore,
Sevilla, Spain Singapore
Bárbara Soria-Juan University of Pablo de Olavide, Swee Hin Teoh Nanyang Technological University,
Sevilla, Spain; Fundación Jiménez Dı́az Health Singapore, Singapore
Research Institute, Madrid, Spain Anthony J. (Tony) Smith University of Birmingham,
Frank E. Stockdale School of Medicine, Stanford Birmingham, United Kingdom
University, Stanford, CA, United States Elsa Treffeisen Department of Dermatology, Kligman
Josh Stover Department Bioengineering, University of Laboratories, University of Pennsylvania School of
Utah, Salt Lake City, UT, United States Medicine, Philadelphia, Pennsylvania, United States
Thomas Stransky Departments of Biomedical Rocky S. Tuan Institute for Tissue Engineering and
Engineering, Stony Brook University, Stony Brook, Regenerative Medicine, The Chinese University of
NY, United States Hong Kong, Hong Kong SAR, P.R. China; School of
Biomedical Sciences, Faculty of Medicine, The Chinese
H. Christiaan Stronks Department of Ophthalmology, University of Hong Kong, Hong Kong SAR, P.R. China
Johns Hopkins University, Baltimore, MD, United
States; Department of Otorhinolaryngology, Leiden Joseph P. Vacanti Harvard Medical School, Center for
University, Leiden, The Netherlands Regenerative Medicine, Massachusetts General
Hospital, Cambridge, MA, United States
Patrick S. Stumpf Faculty of Medicine, Centre for
Human Development, Stem Cells and Regeneration, Cor van der Weele Department of Social Sciences,
Human Development and Health, Institute of Wageningen University, Wageningen, The
Developmental Sciences, University of Southampton, Netherlands
Southampton, United Kingdom Matthew Vincent Avacta Life Sciences, Cambridge,
Kyung Eun Sung Division of Cellular and Gene United Kingdom
Therapies, Office of Tissues and Advanced Therapies, Gordana Vunjak-Novakovic Department of Biomedical
Center for Biologics Evaluation and Research, Food Engineering, Columbia University, New York, NY,
and Drug Administration, Silver Spring, MD, United United States; Department of Medicine, Columbia
States University, New York, NY, United States
Daniel Swarr Division of Neonatology, Perinatal and Lars U. Wahlberg Gloriana Therapeutics, Inc.,
Pulmonary Biology, Cincinnati Children’s Hospital Providence, RI, United States
Medical Center, Perinatal Institute, University of Derrick C. Wan Hagey Laboratory for Pediatric
Cincinnati College of Medicine, Cincinnati, OH, Regenerative Medicine, Division of Plastic and
United States Reconstructive Surgery, Department of Surgery,
Dagmara Szkolnicka Division of Gastroenterology and Stanford University School of Medicine, Stanford,
Hepatology, Centre Hospitalier Universitaire Vaudois, CA, United States
University of Lausanne, Lausanne, Switzerland Anne Wang Department of Dermatology, Kligman
Jun Takahashi Center for iPS Cell Research and Laboratories, University of Pennsylvania School of
Application (CiRA), Kyoto University, Kyoto, Japan Medicine, Philadelphia, Pennsylvania, United States
D.K.O. Tang Bone & Joint Research Group, Centre for Dan Wang Institute for Tissue Engineering and
Human Development, Stem Cells and Regeneration, Regenerative Medicine, The Chinese University of
Human Development and Health, Faculty of Hong Kong, Hong Kong SAR, P.R. China; School of
Medicine, University of Southampton, Southampton, Biomedical Sciences, Faculty of Medicine, The Chinese
United Kingdom University of Hong Kong, Hong Kong SAR, P.R. China
xxxviii List of contributors

Qiwei Wang Center of Stem Cell and Regenerative Anthony J. Windebank Department of Neurology,
Medicine, and Bone Marrow Transplantation Center Regenerative Neurobiology Laboratory, Mayo Clinic,
of the First Affiliated Hospital, Zhejiang University Rochester, MN, United States
School of Medicine, Hangzhou, P.R. China; Institute Mark Eu-Kien Wong Department of Oral and
of Hematology, Zhejiang University & Zhejiang Maxillofacial Surgery, University of Texas Health
Engineering Laboratory for Stem Cell and Science Center Houston, Houston, TX, United
Immunotherapy, Hangzhou, P.R. China States
Yanling Wang Department of Molecular Medicine, The Stefan Worgall Department of Pediatrics, Weill Medical
Scripps Research Institute, La Jolla, CA, United College of Cornell University, New York, NY, United
States; Department of Neurological Sciences, Rush States; Department of Genetic Medicine, Weill
Medical Center, Chicago, IL, United States Medical College of Cornell University, New York,
Yu-li Wang Department of Biomedical Engineering, NY, United States
Carnegie Mellon University, Pittsburgh, PA, United Iwen Wu Division of Clinical Evaluation and
States Pharmacology/Toxicology, Office of Tissues and
Zhanwen Wang Department of Sports Medicine, Advanced Therapies, Center for Biologics Evaluation
Xiangya Hospital, Central South University, and Research, Food and Drug Administration, Silver
Changsha, P.R. China Spring, MD, United States
Valerie M. Weaver Center for Bioengineering and Rui-Xin Wu State Key Laboratory of Military
Tissue Regeneration, Department of Surgery, Stomatology and National Clinical Research Center
University of California, San Francisco, CA, United for Oral Diseases, Department of Periodontology,
States; UCSF Helen Diller Comprehensive Cancer School of Stomatology, Fourth Military Medical
Center, University of California, San Francisco, CA, University, Xi’an, P.R. China
United States; Departments of Bioengineering and
Virginia Y. Xie Department of Bioengineering, Rice
Therapeutic Sciences, and Radiation Oncology, Eli
University, Houston, TX, United States
and Edythe Broad Center of Regeneration Medicine
and Stem Cell Research, University of California, San Malcolm Xing Department of Mechanical Engineering,
Francisco, CA, United States Children’s Hospital Research Institute of Manitoba,
Winnipeg, MB, Canada; Department of Biochemistry
J.A. Wells Bone & Joint Research Group, Centre for
& Genetics, Children’s Hospital Research Institute of
Human Development, Stem Cells and Regeneration,
Manitoba, Winnipeg, MB, Canada
Human Development and Health, Faculty of
Medicine, University of Southampton, Southampton, Kenneth M. Yamada Cell Biology Section, National Institute
United Kingdom of Dental and Craniofacial Research, National Institutes of
Jean F. Welter Department of Biology, Case Western Health, Bethesda, MD, United States; Department of Cell
Reserve University, Cleveland, OH, United States; Systems & Anatomy, UT Health Science Center at San
Case Center for Multimodal Evaluation of Tissue Antonio, San Antonio, TX, United States
Engineered Cartilage, Cleveland, OH, United States Shinya Yamanaka Center for iPS Cell Research and
Feng Wen Nanyang Technological University, Application (CiRA), Kyoto University, Kyoto, Japan;
Singapore, Singapore Gladstone Institute of Cardiovascular Disease, San
Francisco, CA, United States
Jake Weston Department Bioengineering, University of
Utah, Salt Lake City, UT, United States James J. Yoo Wake Forest Institute for Regenerative
Medicine, Wake Forest School of Medicine, Winston-
Jeffrey A. Whitsett Division of Neonatology, Perinatal
Salem, NC, United States
and Pulmonary Biology, Cincinnati Children’s
Hospital Medical Center, Perinatal Institute, Simon Young Department of Oral and Maxillofacial
University of Cincinnati College of Medicine, Surgery, University of Texas Health Science Center
Cincinnati, OH, United States Houston, Houston, TX, United States
James K. Williams Wake Forest Institute for Claire Yu Department of NanoEngineering, University of
Regenerative Medicine, Wake Forest University California, San Diego, La Jolla, CA, United States
School of Medicine, Winston-Salem, NC, United Hanry Yu National University of Singapore, Singapore,
States Singapore
List of contributors xxxix

Yifan Yuan Department of Anesthesiology, Yale Yuanyuan Zhang Wake Forest Institute for Regenerative
University, New Haven, CT, United States Medicine, Wake Forest School of Medicine, Winston-
William Zacharias Division of Neonatology, Perinatal and Salem, NC, United States
Pulmonary Biology, Cincinnati Children’s Hospital Zheng Zhang New Jersey Center for Biomaterials,
Medical Center, Perinatal Institute, University of Rutgers, The State University of New Jersey,
Cincinnati College of Medicine, Cincinnati, OH, United Piscataway, NJ, United States
States Chunfeng Zhao Department of Orthopedic Surgery and
Jason Zakko Center for Regenerative Medicine, Department of Biomedical Engineering, Mayo Clinic,
Nationwide Children’s Hospital, Columbus, OH, Rochester, MN, United States
United States; Department of Surgery, Ohio State Yimu Zhao Department of Chemical Engineering and
University, Wexner Medical Center, Columbus, OH, Applied Chemistry, University of Toronto, Toronto,
United States ON, Canada
Ai Zhang Department of Molecular Medicine, The Laurie Zoloth University of Chicago, Chicago, IL,
Scripps Research Institute, La Jolla, CA, United United States
States; Aspen Neuroscience, Inc., San Diego, CA,
United States
Preface

The first edition of Principles of Tissue Engineering was remains largely unchanged, combining the prerequisites
published almost a quarter-of-a-century ago—back in the for a general understanding of cellular differentiation and
1990s when the term “tissue engineering” was first coine- tissue growth and development, the tools and theoretical
d—and quickly became the most widely relevant and information needed to design tissues and organs, as well
cited textbook in the field. Since that time there have as a presentation by the world’s experts of what is cur-
been powerful developments, including breakthroughs at rently known about each specific organ system, including
all stages of development, ranging from two Nobel Prizes breast, endocrine and metabolism, ophthalmic, oral/dental
for pioneering work in the area of stem cells, which could applications, skin, and the cardiovascular, gastrointestinal,
be used as an unlimited source of cells for repair and hematopoietic, kidney and genitourinary, musculoskeletal,
engineering of tissues and organs, to actual clinical thera- nervous, and respiratory systems. We have again striven
pies, ranging from skin and bladder replacement to carti- to create a comprehensive book that, on one hand, strikes
lage, bone, and cardiovascular repair. a balance among the diversity of subjects that are related
The fifth edition of “Principles” covers all of this tre- to tissue engineering, including biology, chemistry, mate-
mendous progress as well as the latest advances in the rial science, medicine, and engineering, while emphasiz-
biology and design of functional tissues and organs for ing those research areas that are likely to be of clinical
repair and replacement, from mathematical models to value in the future.
clinical reality. We have also added Anthony Atala, the While we cannot describe all of the new and updated
W.H. Boyce Professor and Director of the Wake Forest material of the fifth edition, we continue to provide
Institute for Regenerative Medicine, as a new editor and expanded coverage of stem cells, including neonatal, post-
have expanded the book to include a new section on natal, embryonic, and induced pluripotent stem cells and
emerging technologies, including 3D bioprinting and bio- progenitor populations that may soon lead to new tissue-
manufacturing for tissue-engineering products. As in the engineering therapies for cardiovascular disease, diabetes,
previous editions, the book attempts to simultaneously and a wide variety of other diseases that afflict humanity.
connect the basic sciences with the potential application This up-to-date coverage of stem cell biology and other
of tissue engineering to diseases affecting specific organ emerging technologies is complemented by updated chap-
systems. While the fifth edition furnishes a much needed ters on gene therapy, the regulatory process, and the chal-
update of the rapid progress that has been achieved in the lenges of tissue engineering for food and in vitro meat
field in the last 6 years, we have retained the fundamen- production, which someday may end up a routine part of
tals of tissue engineering, as well as those facts and sec- our food system, potentially reducing environmental pol-
tions which, while not new, will assist scientists, lution and land use. As with previous editions, we believe
clinicians, and students in understanding this exciting area the result is a comprehensive textbook that will be useful
of biology and medicine. to students and experts alike.
The fifth edition of “Principles” is divided into an
introductory section, followed by 23 parts starting with
Robert Lanza, Robert Langer, Joseph Vacanti and
the basic science of the field and moving upward into
Anthony Atala
applications and clinical experience. The organization

xli
Chapter 1

Tissue engineering: current status and


future perspectives
Prafulla K. Chandra, Shay Soker and Anthony Atala
Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, United States

Clinical need congestive heart failure [5], and around 17.9 million
people die or cardiovascular diseases globally (World
Tissue and organ failure due to disease, injury, and Health Organization data on Cardiovascular disease;
developmental defects has become a major economical https://www.who.int/cardiovascular_diseases/en/). TE can
and healthcare concerns [1]. At present, use of donated help such patients by providing healthy engineered tis-
tissues and organs is the clinical practice to address this sues (and possibly whole organ in future) to replace their
situation. However, due to the shortage of organ donors, diseased tissue for restoring function. For example,
the increasing number of people on the transplant waiting chronic kidney disease (CKD) is a worldwide health
lists, and an ever-increasing aging population, dependence crisis that can be treated, but it also depends on organ
on donated tissues and organs is not a practical approach. donation. In the United States alone, around 30 million
In addition, due to severe logistical constraints, many people are suffering from CKD (Center for Disease
organs from donors cannot be matched, transported, and Control & Prevention; National Chronic Kidney Disease
successfully transplanted into a patient within the very Fact Sheet 2017; https://www.cdc.gov/kidneydisease/pdf/
limited time available. In the United States alone, more kidney factsheet), while close to 10% of the population is
than 113,000 people are on the National Transplant affected worldwide. Liver disease is another healthcare
Waiting list and around 17,000 people have been waiting problem, which is responsible for approximately 2 mil-
for more than 5 years for an organ transplant (US lion deaths per year worldwide [6]. Other diseases or
Department of Health and Human Services, Organ conditions that can benefit from TE technologies include
Procurement and Transplantation network; https://optn. skin burns, bone defects, nervous system repair, craniofa-
transplant.hrsa.gov; data as of February, 2019). To cial reconstruction, cornea replacement, volumetric mus-
address this critical medical need, tissue engineering (TE) cle loss, cartilage repair, vascular disease, pulmonary
has become a promising option. TE and regenerative disease, gastrointestinal tissue repair, genitourinary tissue
medicine (RM) are multidisciplinary fields that combine repair, and cosmetic procedures. The field of TE, with its
knowledge and technologies from different fields such as goal and promise of providing bioengineered, functional
biology, chemistry, engineering, medicine, pharmaceuti- tissues, and organs for repair or replacement could trans-
cal, and material science to develop therapies and pro- form clinical medicine in the coming years.
ducts for repair or replacement of damaged tissues and
organs [2,3].
The process of TE is multistep and involves engineer-
ing of different components that will be combined to
Current state of the field
generate the desired neo-tissue or organ (Fig. 1.1). TE has seen continuous evolution since the past two dec-
Today, this field has advanced so much that it is being ades. It has also seen assimilating of knowledge and tech-
used to develop therapies for patients that have severe nical advancements from related fields such as material
chronic disease affecting major organs such as the kid- science, rapid prototyping, nanotechnology, cell biology,
ney, heart, and liver. For example, in the United States and developmental biology. Specific advancements that
alone, around 5.7 million people are suffering from have benefited TE as a field in recent years include novel

Principles of Tissue Engineering. DOI: https://doi.org/10.1016/B978-0-12-818422-6.00004-6


Copyright © 2020 Elsevier Inc. All rights reserved. 1
2 CHAPTER | 1 Tissue engineering: current status and future perspectives

innervation in bioengineered tissue is a continuing chal-


lenge essential to warrant sustained efforts success of tis-
sues implanted in vivo would be very low. Therefore
there is a need for greater understanding of vasculariza-
tion and innervation as applied to bioengineered tissues.
This is an ongoing effort, and the results we are seeing
from various studies are encouraging. Biofabrication tech-
nologies are playing a great role in this regards.
Several engineered tissues are moving toward clinical
translation or are already being used in patients. These
include cartilage, bone, skin, bladder, vascular grafts, car-
diac tissues, etc. [12]. Although, complex tissues such as
liver, lung, kidney, and heart have been recreated in the
lab and are being tested in animals, their clinical transla-
tion still has many challenges to overcome. For in vitro
use, miniature versions of tissues called organoids are
being created and used for research in disease modeling,
drug screening, and drug development. They are also
being applied in a diagnostic format called organ-on-a-
chip or body-on-a-chip, which can also be used for the
FIGURE 1.1 Schematic representation of different aspects of tissue
above stated applications. Indeed, the development of 3D
engineering. Each component (materials, cells, and tissue architectures) tissue models that closely resemble in vivo tissue struc-
can be engineered separately or in combination to achieve the therapeu- ture and physiology are revolutionizing our understanding
tic goals. Reprinted with permission from Khademhosseini A., Langer R. of diseases such as cancer and Alzheimer and can also
A decade of progress in tissue engineering. Nat Protoc 2016;11 accelerate development of new and improved therapies
(10):1775 81. doi: 10.1038/nprot.2016.123 [4]. r2016 Springer
Nature Publishing AG.
for multiple diseases and disorders. This approach is also
expected to drastically reduce the number of animals that
are currently being used for testing and research. In addi-
biomaterials [7], three-dimensional (3D) bioprinting tion, 3D tissue models and organ-on-a-chip or body-on-a-
technologies [8], integration of nanotechnology [9], stem- chip platforms can support advancement of personalized
cell technologies such as induced pluripotent stem medicine by offering patient-specific information on the
cells (iPSCs) [9,10], and gene editing technology such as effects of drugs, therapies, environmental factors, etc.
Clustered Regularly Interspaced Short Palindromic Development of advanced bioreactors represent
Repeats (CRISPR) [11]. All these have led to promising another recent developments that are supporting clinical
developments in the field that include smart biomaterials, translation of TE technologies. Such bioreactors can bet-
organoids, and 3D tissue for disease modeling and drug ter mimic in vivo environments by provide physical and
development, whole organ engineering, precise control biochemical control of regulatory signals to cells and tis-
and manipulation of cells and their environments, and sue being cultured. Examples of such control include
personalized TE therapies. application of mechanical forces, control of electrical pac-
Biomaterials are critical components of many current ing, dynamic culture components, induction of cell differ-
TE strategies. Recent developments in this field that are entiation. Incorporation of advanced sensors and imaging
benefiting TE include synthesis of new biomaterials that capabilities within these bioreactors are also allowing for
can respond to their local environment and cues (smart real-time monitoring of culture parameters such as pH,
biomaterials). Advancements in 3D bioprinting technolo- oxygen consumption, cell proliferation, and factor secre-
gies are at the core of many developments in TE. It is tion from a growing tissue. 3D modeling is also a new
now possible to print multiple biocompatible materials tool relevant to TE that provides great opportunities and
(both natural and synthetic), cells, and growth factors better productivity for translational research, with wide
together into complex 3D tissues, many with functional clinical applicability [13]. Recent advancements in spe-
vascular networks, which match their counterparts cific field that are helping advance TE are discussed next.
in vivo. We have also learned a great deal about cell
sourcing, culture, expansion, and control of differentia-
Smart biomaterials
tion. This is also true for stem cells, where new sources
such as placenta, amniotic fluid, and iPSCs have been Smart biomaterials are biomaterials that can be designed
explored and optimized for use. Vascularization and to modulate their physical, chemical, and mechanical
Tissue engineering: current status and future perspectives Chapter | 1 3

properties in response to changes in external stimuli or linking through physical methods, self-assembly, or ther-
local physiological environment (Fig. 1.2) [14,15]. mally induced polymer chain entanglement is creating
Advances in polymer synthesis, protein engineering, hydrogels that undergo structural changes in response to
molecular self-assembly, and microfabrication technolo- external stimuli [19,20]. Another class of hydrogels that
gies have made producing these next-generation biomater- are recent developments is called self-healing and shear
ials possible. These biomaterials can respond to a variety thinning hydrogels. These materials are now being used
of physical, chemical, and biological cues such as temper- to develop injectable biomaterials, which have low vis-
ature, sound, light, humidity, redox potential, pH, and cosity during application (injection) due to shear thinning
enzyme activity [16,17]. Other unique characteristics dis- and once at their target site, they self-crosslink (or heal)
played by some smart biomaterials are self-healing or to fill the defect site [21]. Injectable biomaterials are also
shape-memory behavior [18]. The development of bioma- often loaded with drugs, biologics, and cells. For exam-
terials with highly tunable properties has been driven by ple, Montgomery et al. created an injectable shape-
the desire to replicate the structure and function of extra- memory biomaterial for minimally invasive delivery of
cellular matrix (ECM). Such materials can enable control functional tissues [22]. In other applications, tissue glues
of chemical and mechanical properties of the engineered are being developed using smart biomaterials, where they
tissue, including stiffness, porosity, cell attachment sites, are used to bond and allow the tissue to self-heal. An
and water uptake. For hydrogels, use of reversible cross- example of this approach is a study by Bhagat and Becker

FIGURE 1.2 Different applications of smart biomaterials in the fields of tissue engineering and related fields. (A) Stimuli-responsive material that
can promote cell differentiation and tissue growth; (B) injectable biomaterial loaded with cells, drugs, or bioactive molecules can be delivered less-
invasively and can promote healing of tissue at the target damage site; (C) swelling polymer can be delivered as small scaffolds but can expand
in vivo to achieve 3D structure of the target defect after exposure to water; (D) shape-memory and temperature-responsive soft material can be used
as a tissue adhesive; (E) star-shaped delivery system for sustained drug release in the gastrointestinal tract; (F) nanoparticle-based stimuli-responsive
drug delivery system for systemic application; (G) materials for enhanced cancer immunotherapy using targeted delivery of chimeric antigen receptor
T cell. 3D, Three-dimensional. Reprinted with permission from Kowlaski PS, Bhattacharya C, Afewerki S, Langer R. Smart biomaterials: recent
advances and future directions. ACS Biomater Sci Eng 2018;4(11):3809 17 [14]. r2018 American Chemical Society.
4 CHAPTER | 1 Tissue engineering: current status and future perspectives

who created a chondroitin-based tissue glue that helps can also control cellular behavior. For example, in a
direct improved tissue repair [23]. mouse study by Moore et al., MDPs alone were found to
The ECM is a complex and dynamic structural scaf- be biocompatible and had prohealing effects in vivo [39].
fold for cells within tissues and plays an important role in Hydrogel have also been prepared from multiple ECM
regulating cell function [1]. Given the role of the ECM in mimetic peptides for the purpose of enhancing the viabil-
structural support of tissues, there has been significant ity of the biomaterial in vivo. Smart biomaterials are
effort in developing ECM-based scaffolds for TE and RM going to have a big impact on 3D printing of tissues and
[24,25]. However, as with all materials implanted into the organs. By combining smart biomaterials with 3D bio-
body, the immune response significantly influences the printing, a wide variety of architectures can be created
ability of scaffold-containing engineered tissues to inte- which can further offer control over how these materials
grate and functionally interact with the host [26]. Thus an perform in a biological environments. Smart biomaterials
emerging strategy in TE is to design materials that can can also be made from proteins. Some protein protein
directly control the host immune response [27]. For exam- interactions can be utilized to physically crosslink protein
ple, the Arg-Gly-Asp (RGD) of ECM proteins can exert chains, while small coiled-coil domains within some pro-
immunomodulatory effects on both innate and adaptive teins (called leucine zippers) can self-assemble into super-
immune cells while also having an inhibitory effect on helical structures. Leucine zippers have been used to
phagocytosis and neutrophil chemotaxis [28]. In the con- make hydrogels by physically crosslinking protein
text of TE, synthetic ECM-mimetic hydrogels containing domains [40]. The stability of the leucine zipper self-
the RGD sequence have been shown to cause increased assembly (and hence the hydrogel) can be controlled by
cellular adhesion on polymer scaffolds and also have an changing the temperature. Another way to control the sta-
antiinflammatory effects from macrophages [29,30]. bility of some protein-based hydrogels is by arrangement
Under certain conditions, the RGD peptides have also of the interacting domains [41].
been found to effect cytokine secretion from T cells [31]. One drawback of hydrogels made of self-interacting
Therefore use of RGD as part of TE scaffolds or hydro- protein domains is their low-to-moderate mechanical
gels can be used to enhance cells adhesion in addition to properties, which is not ideal for TE applications.
controlling the ability of macrophages to degrade and However, these week interactions can be reinforced by
remodel the surrounding tissue environment. introducing covalent bonds into the network (e.g., disul-
Matrix metalloproteinases (MMPs) are a family of fide bonds between cysteine in the protein chains). This
proteases that not only selective degrade a wide variety of will not only improve the mechanical properties of the
ECM proteins but also interact with bioactive molecules, hydrogel but also its stability [42].
some of which have immunomodulatory effects [32,33].
So, another strategy to control the extent of matrix remo-
Cell sources
deling, integration of engineered tissues into native host
tissues or invasion of immune cell into implanted materi- For TE, a variety of cell types are now being used. They
als could be by incorporating MMP-sensitive peptides include autologous, allogeneic, progenitors, adult unipo-
into the TE constructs. Examples of this approach include tent or multipotent stem cells and iPSCs (Fig. 1.3). For
studies by Patterson and Hubbell, who showed that the some applications, the ability to expand a sufficient
rate of scaffold material degradation depends on the number of autologous cells from a small biopsy is well-
MMP-sensitive peptide sequence, the type of MMP, and established [44]. A good example is bladder augmenta-
also the MMP concentrations [34]. In a separate study, tion, where smooth muscle and urothelium can be easily
West and Hubbell created biomimetic poly(ethylene gly- isolated from then native tissue, expanded in culture and
col) (PEG) hydrogels that incorporated peptides that could used for engineering a new bladder tissue. However, in
be degraded by either a fibrinolytic protease (plasmin) or many cases, it is challenging to harvest and/or expand
a fibroblast collagenase (MMP-1) [35,36]. One drawback enough appropriate autologous cells for this purpose.
of this using MMP-sensitive peptides in TE constructs is Examples of such cell types include hepatocytes, kidney
their immunogenicity and more work will be needed to cells, insulin-producing pancreatic beta cells, cardiomyo-
get around this issue. Possibly, use of immunomodulatory cytes, neurons. New sources or methods to obtain these
domains along with MMP-sensitive peptides could sup- cell types in quantities can advance engineering of these
port long-term viability and integration within native host tissues/organs and significantly benefit treatment of asso-
tissues. ciated diseases. Immature precursor cells present within
Another category of smart biomaterials is multidomain tissue such as skin, cartilage, muscle, and bladder are
peptides (MDPs) hydrogels. These are injectable ECM essential for the expansion of corresponding cells from
mimetic materials that are engineered to form self- biopsies and enabling engineering of neo-tissues [45].
assembling meshes at the target site [37,38]. These MDPs The extension of this approach to other tissue and organ
Tissue engineering: current status and future perspectives Chapter | 1 5

FIGURE 1.3 Different sources of


Cell sources
for cells for tissue engineering. Fetus-
tissue engineering derived and induced pluripotent
stem cells are gaining more atten-
tion for tissue engineering applica-
tions. Reprinted from Al-Himdani
Embryonic stem Fetus-derived stem Adult stem Induced pluripotent
S, Jessop ZM, Al-Sabah A,
cells cells cells stem cells
Combellack E, Ibrahim A, Doak
SH, et al. Tissue-engineered solu-
E.g., ESC
E.g., UC-MSC/EPC, CV-
EPC,AF-MSC/EPC
E.g., EPCs, BMSC, ADSC E.g., iPSC tion in plastic and reconstructive
Totipotent Pluripotent or Pluripotent or
Pluripotent surgery: principles and practice.
multipotent multipotent
Front Surg 2017;4:4. doi: 10.3389/
fsurg.2017.00004. [43]. r2017 Al-
Morula Chorionic villi Amniotic fluid Blood Bone marrow Somatic cells Himdani, Jessop, Al-Sabah,
Combellack, Ibrahim, Doak, Hart,
Archer, Thornton and Whitaker.
Open-access article distributed
Adipose tissue under the terms of the Creative
Umbilical cord Commons Attribution License (CC
BY). Some portions of the original
artwork have been modified.

systems will depend greatly on finding sources of appro- The first clinical tissue-engineered products to achieve
priate stem and progenitor cells. marketing approval from the US Food and Drug
Three major stem-cell sources are currently under Administration (FDA) were skin substitutes that were
intensive investigation: used for wound healing. Examples of such products
include Dermagraft (Shire Regenerative Medicine Inc.,
1. embryonic stem (ES) cells, which are derived from
CT, United States) and Apligraf (Organogenesis, MA,
discarded human embryos, and the equivalent embry-
United States), which were off-the-shelf products that
onic germ (EG) cells;
used cells (fibroblasts for Dermagraft and fibroblasts plus
2. iPSCs derived by genetic reprograming of somatic
keratinocytes for Apligraf) expanded from donated human
cells; and
foreskins. Whereas fibroblasts have been cultured in vitro
3. Autologous or allogeneic adult tissue stem cells
since the early 20th century, the successful large-scale
(sourced from fetal, neonatal, pediatric, or adult donor
culture of human keratinocytes represented an important
tissue).
breakthrough for RM [46]. The success of off-the-shelf
Shared features of all stem cells include their capacity skin substitutes can be attributed to the lack of antigen-
self-renewal and their ability to give rise to particular presenting cells, because of which they were not acutely
classes of differentiated cells. The ES, EG, and iPSCs can rejected despite the inevitable histocompatibility mis-
serve as precursors for many specialized cell type found matches between donors and recipients [47,48].
during normal development and therefore are pluripotent. Eventually, the cells in the skin substitutes could be
Adult stem cells are generally restricted to limited sets of rejected, but the grafts has enough time for patients’ own
cell lineages, hence called unipotent (constrained to a sin- skin cells to regenerate. This stands in contrast to standard
gle fate) or multipotent (can give rise to multiple cell tissue/organ transplantation in which immune rejection is
types). It appears likely that multiple tissue-engineered a major concern and immunosuppressive drug therapy is
products based on each class of stem-cell source will be generally part of the application of allogeneic grafts [49].
tested in the clinic in the coming years. Previous clinical Tissue-engineered products based on harvesting and
and commercial experience sheds light on key differences expanding autologous cells containing stem and/or pro-
between personalized products containing autologous genitor populations have also been developed success-
cells and off-the-shelf products containing allogeneic fully. Prominent examples include Epicel (Genzyme, MA,
cells. The vast majority of human studies till date have United States), a permanent skin replacement product
focused on using either adult stem or progenitor cells. based on expanded keratinocytes for patients with life-
More recently, clinical trials have begun with tissue- threatening burns, and Carticel (Genzyme, MA, United
engineered products derived from pluripotent stem cells States), a chondrocyte-based treatment for large articular
and their future looks promising. cartilage lesions [50,51].
6 CHAPTER | 1 Tissue engineering: current status and future perspectives

Embryonic stem cells Institute for Regenerative Medicine; CA, United States)
ES cells and EG cells are indeed quite similar to early were used in clinical trials in patients to treat Stargardt’s
germ cells, with an apparently unlimited self-renewal macular dystrophy and dry age-related macular degenera-
capacity and pluripotency. Their great degree of plasticity tion. Encouraging results from such clinical studies using
represents both a strongest virtue and a significant poten- ES cell-derived product will have a positive impact to
tial limitation to their use in TE. A major ongoing chal- develop tissue-engineered products from pluripotent stem
lenge is in efficiently obtaining pure populations of cells in the near future. Areas of clear unmet medical
specific desired specialized cell types from human ES need that might benefit from stem-cell-derived products
cells [52,53]. Efforts during recent years have yielded include type 1 diabetes and Parkinson’s disease. For type
more robust methods to isolate and grow ES cells under 1 diabetes, research at a biotech company called Viacyte
conditions consistent with Good Manufacturing Practice Inc. (CA, United States) similarly pursued the produced
(GMP) and to generate differentiated cell products. While progenitors of pancreatic endocrine cells from human ES
initial efforts have focused on cell therapies, these cells using growth factors and hormones [63]. The pro-
advances will positively impact production of tissue- genitor cells from the final-stage differentiation in vitro
engineered constructs using ES cells. Human ES cells are were able to mature further in vivo to yield glucose-
considerably more difficult to isolate and maintain stably responsive β-like cells [64]. As a potential therapy for
in culture than the cell types that have previously been Parkinson’s disease, significant advances have been made
used in clinical testing. However, they can now be in the production of functional midbrain dopaminergic
derived, grown, and cryopreserved without exposure to neurons by staged differentiation from ES cells [65,66].
nonhuman cells or proteins, even under a GMP environ- Studies in the past few years have demonstrated that effi-
ment [54,55]. In the future, use of bioreactors, microcar- cient grafting of these cells can lead to physiological cor-
riers, along with improved xeno-free and serum-free rection of symptoms in several animal models, including
media and possibly small molecules that inhibit spontane- nonhuman primates [67]. A particular safety concern is
ous differentiation of these cells would facilitate expan- that undifferentiated pluripotent ES and iPS cells form
sion of these stem cells to population sizes that are teratomas in vivo. The risk of tumorigenicity makes it
normally required for product development and clinical essential to rigorously determine the residual level of
application [56,57]. undifferentiated stem-cell population in any therapeutic
Human tissues include more than 200 distinct cell product derived from ES or iPS cells [68]. It will also be
types, and ES cells, in principle, can give rise to all of valuable to determine whether a small number of undif-
them. The historical approach of allowing ES cells to dif- ferentiated pluripotent stem cells can be introduced into
ferentiate spontaneously has now been supplanted. human patients without significant risk of tumor growth
Current strategies employ staged differentiation guided by and if this threshold is influenced by use of immune sup-
knowledge of signaling events that regulate normal pressive drugs during treatment.
embryonic development [58]. For example, fine tuning of
the exposure of early embryonic cells to the growth factor Induced pluripotent stem cells
Nodal (a member of the transforming growth factor beta Theoretically, the development of iPSCs represent the
or TGF-β family) or its analog Activin A, in conjunction most direct way to ensure immune compatibility of
with other growth factors or small molecules, can now tissue-engineered products when the recipient themselves
allow consistent generation of endoderm-specific cells serve as the donor. Generation of iPSCs through repro-
from ES cells in vitro [59,60]. This is an early, but graming of mature somatic cells to a pluripotent state was
key milestone in a multistep process to generate differen- first accomplished by ectopic expression of four transcrip-
tiated cells that can eventually be used for TE of tissues/ tion factors: OCT4 and SOX2, both with KLF4 and c-
organs like the liver and pancreas. Conversely, inhibition MYC [69] or NANOG and LIN28 [70]. The resulting
of Nodal/Activin signaling favors the production of ecto- iPSCs closely resembled ES cells in key properties such
derm specific cells, a precursor for neural lineage cells as the capacity for extensive self-renewal, ability to dif-
[61]. ferentiate to multiple cell lineages, and generation of tera-
Despite substantial challenges, the first ES-cell- tomas in vivo. Initial studies on reprograming of
derived therapeutic product to enter clinical trials was the fibroblasts soon were extended to a variety of other cell
human ES-cell-derived oligodendrocyte progenitors types such as peripheral blood cells [71], cord blood cells
(Geron Corporation; CA, United States) for stimulating [72], keratinocytes from hair shafts [73], and urine-
nerve process growth in subjects with spinal cord injury derived cells [74]. Many recent developments have
[62]. Similarly, ES-cell-derived retinal pigment epithe- advanced this reprograming technology toward a safer,
lium cells (Advanced Cell Technology, now Astellas efficient translation toward therapeutic products. Also,
Another random document with
no related content on Scribd:
hours you have been spending alone.” “Alone!” he exclaimed, in a
joyful tone, “I am never alone, and never weary. How should I be
either, when my days are passed in the company of innocent
animals, and time is given me to think of God!” The priest smiled
even more approvingly than before; and I remarked to him, “We are
here in Arcadia.” “But not without human sin,” said he, and pointing
to a woman at a distance, who was in the employ of the farmer’s
wife, he asked the latter how she could still have anything to do with
a well-known thief. “Eh, father,” was the comment of a woman whom
John Howard would have kissed, “starving her in idleness would not
cure her of pilfering; and between working and being well-watched,
she will soon leave her evil habits.” “You are a good Christian,” I said
to her, “be you of what community you may.” “She is a good
Catholic,” added the priest. “I am what the good God has made me,”
was the simple reply of the Walloon wife; “and my religion is this to
go on my knees when all the house is asleep, and then pray for the
whole world.” “Ay, ay,” was the chorus of those around her, “that is
true religion.” “It is a part of true religion,” interposed the priest; but I
could not help thinking that he would have done as well had he left
Marie Justine’s text without his comment. We walked together down
to the bank of the river opposite the Chateau of the young Count de
Levignon the proprietor and burgomaster of Houx. I looked up from
the modern chateau to the ruins of the vast castle where the sons of
Aymon once held barbaric state, maintained continual war, and
affected a reverence for the mother of Him who was the Prince of
Peace. The good priest seemed to guess my thoughts, for he
remarked, “We live now in better times; the church is less splendid,
and chivalry less ‘glorious,’ if not extinct; but there is a closer
brotherhood of all men—at least,” he added hesitatingly—“at least I
hope so.” “I can not remember,” said I, “a single virtue possessed by
either Aymon or his sons, except brute courage, and a rude sort of
generosity, not based on principle, but born of impulse. It is a pity
that Belgium can not boast of more perfect chevaliers than the old
proprietors of Poilvache, and that you have not a hero to match with
Bayard.” “Belgium,” was his answer, “can make such boast, and had
a hero who had finished his heroic career long before Bayard was
born. Have you never heard of ‘the Good Knight without fear and
without doubt’?” “I have heard of one without fear and without
reproach.” “That title,” he remarked, “was but a plagiarism from that
conferred on Jacques de Lelaing, by his contemporaries.” And then
he sketched the outline of the good knight’s career, and directed me
to sources where I might gather more detailed intelligence. I was
interested in what I learned, and it is because I hope also to interest
readers at home, that I venture to place before them, however
imperfectly rendered, a sketch of the career of a brave man before
the time of Bayard; one who illustrates the old saying that—

“Vixere fortes ante Agamemnona.”

Jacques de Lelaing, the good knight, without fear and without doubt,
was born in the château of Lelaing, in the first quarter of the fifteenth
century. The precise year is not known, but it was full half a century
before the birth of Bayard. He came of a noble race; that is, of a
race, the male portion of which saw more honor in slaughter than in
science. His mother was celebrated for her beauty as well as nobility.
She was wise, courteous, and débonnaire; well-mannered, and full
of all good virtues. So, at least, in nearly similar terms, wrote George
Chastellan of her, just two centuries ago.
Jacques de Lelaing was as precocious a boy as the Duke of
Wharton in his youth. At the age of seven, a priestly tutor had
perfected him in French and Latin, and the good man had so imbued
him with literary tastes that, in after life, the good knight found time to
cultivate the acquaintance of Captain Pen, as well as of Captain
Sword; and specimens of his handiwork are yet said to exist in the
libraries of Flanders and Brabant.
Jacques, however, was never a mere student, “sicklied o’er with the
pale cast of thought.” He loved manly sports; and he was yet but a
blooming youth when the “demoiseau of Clèves,” nephew of that
great Duke whom men, for no earthly reason, called Philip the Good,
carried off his young friend from the castle of Lelaing, and made of
him a squire, not of dames, but of knights, in the turbulent court of
the ducal Philip, with the benevolent qualification to his name.
The youth entered upon his career with a paternal provision which
bespoke at once the liberality and the wisdom of his father, stout
William de Lelaing. The sire bestowed upon his son four splendid
horses, a well-skilled groom, and a “gentleman of service” which, in
common phrase, means a valet, or “gentleman’s gentleman.” But the
young soldier had more than this in his brain; namely, a well-lettered
cleric, commissioned to be for ever expounding and instructing, with
a special object, to boot, that Jacques should not forget his Latin!
Excellent sire thus to care for his son! If modern fathers only might
send into barracks with their sons, when the latter first join their
regiments, reverend clerks, whose office it should be to keep their
pupils well up in their catechism, the Eton grammar, and English
orthography, what a blessing it would be to the young gentlemen and
to all acquainted with them! As it is, we have officers worse
instructed and less intelligent than the sons of the artists who make
their uniforms.
When Jacques went forth into the world, his sire gave him as good
advice as Polonius threw away on his son Laertes. The sum of it was
according to the old French maxim, “Noblesse oblige”—“Inasmuch,”
said the old man, “as you are more noble than others by birth, so,”
said he, “should you be more noble than they by virtues.” The hearty
old father added an assurance, that “few great men gained renown
for prowess and virtue who did not entertain love for some dame or
damoiselle.” This last, however, was but an equivocal assurance, for
by counselling Jacques to fall in love with “some dame or
damoiselle,” he simply advised him to do so with any man’s wife or
daughter. But it was advice commonly given to young gentlemen in
arms, and is, to this day, commonly followed by them. Jacques
bettered the paternal instruction, by falling in love with two ladies at
the same time. As ambitious youths are wont to do, he passed by
the white and pink young ladies whom he met, and paid his
addresses, with remarkable success, to two married duchesses.
Neither of these suspected that the smooth-chinned young “squire”
was swearing eternal fidelity to the other, or that this light-mailed
Macheath wooed his madiæval Polly with his pockets full of “favors,”
just bestowed on him by an unsuspecting Lucy. Thus has love ever
been made by officers and highwaymen.
But if Jacques loved two, there was not a lady at the Court of
Burgundy who did not love him. The most virtuous of them sighingly
expressed a wish that their husbands, or their lovers, were only like
him. The men hated him, while they affected to admire his grace, his
bearing, and his irresistible bravery. Jacques very complacently
accepted the love of the women and the envy of the men; and
feeling that he had something to be thankful for, he repaired to the
shrine of the Virgin at Hal, and thanked “Our Lady,” accordingly.
Now Philip the Good was good only just as Nicholas the Czar was
“good.” He had a fair face and a black heart. Philip, like Nicholas,
joined an outward display of conjugal decency with some private but
very crapulous indecency; and the Duke, like the Czar, was the
appalling liar of his day. Philip had increased the ducal territory of
Burgundy by such means as secured Finland to Muscovy, by
treachery of the most fiendish quality; and in 1442, affecting to think
that Luxembourg was in the sick condition which Nicholas described
as the condition of Turkey—when the imperial felon thought he was
making a confederate of Sir Hamilton Seymour, the Duke resolved to
seize on the territory in question, and young Jacques de Lelaing was
in an ecstacy of delight at being permitted to join in this most rascally
of expeditions.
Within a year, desolation was spread throughout a wide district. Fire
and sword did their devastating work, and the earth was swept of the
crops, dwellings, and human beings, which lay between the invaders
and Luxembourg. The city was ultimately taken by surprise, and the
good Philip delivered it up to pillage; then ensued a scene which hell
itself could not equal; and the Duke and his followers having enacted
horrors from which devils would have recoiled, they returned to
Brussels, where they were received with ten times more delight than
if they had come back from an expedition which had been
undertaken for the benefit of humanity.
What was called peace now followed, and Jacques de Lelaing,
having fleshed his maiden sword, and gained the praise of brave
men, and the love of fair women, resolved to commence a series of
provincial excursions for his own especial benefit. As, in modern
times, professors without scholars, and actors without engagements,
wander from town to town, and give lectures at “the King’s Arms,” so
Jacques de Lelaing went forth upon his way, offering to fight all
comers, in presence of kings themselves.
His first appearance on this provincial tour was at Nancy, in 1445,
where a brilliant French Court was holding joyous festival while
awaiting the coming of Suffolk, who was commissioned to escort to
England a royal bride, in the person of Margaret of Anjou. The
French knights made light of the soldier of Burgundy; but Jacques,
when announcing that he was the holder of the tournament, added
that no French knight should unhorse him, unless God and his good
lady decreed otherwise.
The latter was not likely, and he felt himself secure, doubly so, for he
rode into the lists decorated with favors, gold embroidery, and rich
jewels, the gifts of the Duchesses of Orleans and Calabria, each of
whom fondly believed that she was the sole fair one by whose bright
eyes Jacques de Lelaing swore his prettiest oath. Accordingly, there
was not a cavalier who rode against him in that passage of arms,
who left the field otherwise than with broken or bruised bones. “What
manner of man will this be?” cried they, “if, even as a lad, he lays on
so lustily?”
The lad, at the subsequent banquet, to which he was borne in
triumph, again proved that he had the capacity of a man. He was
fresh as a rose just blown; gay as a lark in early spring. The queens
of France and Sicily conversed with him by the half hour, while ladies
of lower degree gazed at him till they sighed; and sighed, knowing
full well why, and caring very much, wherefore. Charles VII. too,
treated him with especial distinction, and conferred on him the rich
prizes he had won as victor in the rough tourney of the day. But there
were other guerdons awarded him that night, which he more highly
prized. Jacques visited the Duchess of Orleans in her bower, and
carried away with him, on leaving, the richest diamond she had to
bestow. He then passed to the pavilion of the Duchess of Calabria, a
lady who, among other gifts willingly made by her, placed upon his
finger a brilliant ruby set in a gorgeous gold ring. He went to his own
bed that night as impudently happy as a modern Lifeguardsman who
is successfully fooling two ladies’ maids. His cleric had left him, and
Jacques had ceased to care for the keeping-up of his Latin, except,
perhaps, the conjugation of the imperative mood of amo. “Amemus,”
let us love, was the favorite part of the mood, and the most
frequently repeated by him and his brace of duchesses.
Sometime after this very successful first appearance, and toward the
end of 1445, our doughty squire was traversing the cathedral of
Notre Dame of Antwerp, and was on the point of cursing the singers
for their bad voices, just as one might be almost justified in doing
now, so execrable are they; he was there and thus engaged, when a
Sicilian knight, named Bonifazio, came jingling his spurs along the
transept, and looking jauntingly and impertinently as he passed by.
Jacques looked boldly at this “pretty fellow” of the time, and
remarked that he wore a golden fetter ring on his left leg, held up by
a chain of the same metal fastened to a circlet above his knee. His
shield bore the device, “Who has fair lady, let him look to her well!”
“It’s an impertinent device,” said Jacques, touching the shield, by
way of token that he would fight the bearer for carrying it. “Thou art
but a poor squire, albeit a bold man,” said the Sicilian, with the air of
one who was half inclined to chastise the Hainaulter for his
insolence. Toison d’Or, the herald, whispered in the ear of the
Hainaulter; thereupon, Jacques exclaimed, “If my master, Duke
Philip, will give me permission to fight, thou darest not deny me, on
his Grace’s territory.” Bonifazio bowed by way of assent. The
permission was gained, and the encounter came off at Ghent. The
first day’s combat was a species of preliminary struggle on
horseback, in which Jacques showed himself so worthy of the spurs
he did not yet wear, that Philip fastened them to his heels the next
day, and dubbed him Knight in solemn form. As the combatants
strode into the lists, on the second day, the Duke of Orleans
remarked to his Duchess, that Jacques was not so “gent as the
Sicilian.” The Duchess smiled, as Guinever smiled when she looked
on Sir Launcelot, while her husband, King Arthur, commented upon
him; and she said, in phrase known to all who read Spenser, “he
loves a lady gent;” and she added, with more of the smile and less of
the blush, “he is a better man than the Sicilian, and, to my thinking,
he will this day prove it.”
“We shall see,” remarked the Duke carelessly.
“We shall see,” re-echoed the Duchess, with the sunniest of smiles.
Jacques, like the chivalric “gent” that he was, did honor to the
testimony of the Duchess. The combatants went at it, like stout men.
Jacques belabored his antagonist with a staff, the Sicilian answered
by thrusting a javelin at his adversary’s uncovered face. They then
flung away their arms and their shields, and hewed at each other
with their battle-axes. Having spoiled the edges of these, and
loosened them from their handles, by battering at each other’s skulls,
they finally drew their lusty and well-tempered swords, and fought so
fiercely that the gleaming of their swiftly manœuvred blades made
them seem as if they were smiting each other with lightning. Jacques
had well-nigh dealt a mortal thrust at the Sicilian, when, at the
intervention of the Duke of Orleans, Philip the Good flung his
truncheon into the lists, and so saved the foreign knight, by ending
the fray. The Duchess reproved her consort for being over-intrusive,
but she smiled more gleesomely than before. “Whither away, Sir
Jacques?” asked she, as the latter modestly bowed on passing her
—the multitude the while rending the welkin with their approving
shout. “To the chapel in the wood,” replied Jacques, “to render
thanks for the aid vouchsafed to me by our Lady.” “Marry,” murmured
the Duchess, “we will be there too.” She thought it not less edifying
to see knight at his devotions than at beholding him in the duello. “I
am grateful to the Lady of Good Succor,” said Jacques. “And thou
doest right loyally,” was the comment of the Duchess.
The victory of the Belgian cavalier over the Sicilian gained for him
the distinctive name which he never lost, that of “the Good Knight.”
To maintain it, he proceeded to travel from court to court, as pugilists
itinerate it from fair to fair, to exhibit prowess and to gather praise.
The minor pugilist looks to pence as well as praise, and the ancient
knight had an eye to profit also—he invariably carried off the horse,
armor, and jewels of the vanquished. As Sir Jacques deemed
himself invincible, he looked to the realization of a lucrative tour. “Go
on thy way, with God’s blessing,” exclaimed his sire. “Go on thy way,
Jacques,” murmured his mother through her tears; “thou wilt find
ointment in thy valise, to cure all bruises. Heaven send thee a
surgeon, and thou break thy bones.”
Across the French frontier merrily rode Sir Jacques, followed by his
squire, and attended by his page. From his left arm hung a
splendidly-wrought helmet, by a chain of gold—the prize offered by
him to any one who could overcome him in single combat. Jacques
announced that, in addition, he would give a diamond to any lady or
demoiselle indicated to him by his conqueror. He stipulated that
whichever combatant first dropped his axe, he should bestow a
bracelet upon his adversary; and Jacques would only fight upon the
condition that neither knight should be fastened in his saddle—a
regulation which I should never think of seeing insisted upon
anywhere, except by equestrian aldermen when they amble on Mr.
Batty’s horses, to meet the Sovereign at Temple Bar. For the rest
Jacques put his trust in God, and relied upon the strength given him
in the love of “the fair lady who had more power over him than aught
besides throughout the entire world.” A hundred ladies fair, matrons
and maids, who heard of this well-advertised confidence, did not
hesitate to exclaim, “Delicious fellow! He means me!”
It was the proud boast of Jacques, that he traversed the capital, and
the provincial cities of France, without meeting with a knight who
would accept his defiance. It would be more correct to say—a knight
who could take up his challenge. Charles VII. forbade his chivalry
from encountering the fierce Hainaulter anywhere but at the festive
board. In the South of France, then held by the English, he met with
the same civility; and he rode fairly into Spain, his lance in rest,
before his onward career was checked by the presence of an
adversary. That adversary was Don Diego de Guzman, Grand-
master of Calatrava, and, although he knew it not, ancestor to a
future Empress of the French. The Don met the Belgian on the
borders of Castile, and accepted his published challenge out of mere
love, as the one silly fellow said of the other, out of mere love for his
“très aimée dame.” The “dames” of those days enjoyed nothing so
much as seeing the gentlemen thwack each other; and considering
what a worthless set these latter, for the most part, were, the ladies
had logically comic reasons to support their argument.
It was necessary, however, for Don Diego to obtain the consent of
his sovereign to encounter in mortal combat a knight of the
household of Burgundy, then in alliance with Spain. The Sovereign
was absent from the country, and while an answer was being
expected from him to the application duly made, Jacques, at the
head of a most splendid retinue, trotted leisurely into Portugal, to
tempt the Lusitanian knights to set their lances against him. He rode
forward to the capital, and was greeted by the way, as if he had been
as illustrious a monarch as his ducal master. It was one ovation, from
the frontier to Lisbon, where he was welcomed by the most crowded
of royal balls, at which the King (Alphonso XV.) invited him to foot it
with the Queen. The King, however, was but an indifferent master of
the ceremonies. The late Mr. Simpson of Vauxhall, or the illustrious
Baron Nathan of Rosherville, would never have dreamed of taking
the lady to introduce her to the gentleman. This uncourteous process
was, however, the one followed by Alphonso, who taking his consort
by the hand, led her to Sire Jacques, and bad him tread a measure
with her. Messire Jacques consented, and there was more than
enough of dancing, and feasting, and pleasure-seeking, but no
fighting. Lisbon was as dull to the Belgian as Donnybrook Fair
without a skrimmage used to be to all its lively habitués. “I have had
a turn with the Queen,” said Jacques, “let me now have a tourney
with your captains.” “Burgundy is my good friend,” answered the
King—and he was right in a double sense, for Burgundy was as dear
to him as Champagne is to the Czar’s valet, Frederick William, who
resides at Berlin. “Burgundy is our good friend,” answered Alphonso,
“and Heaven forbid that a knight from such a court should be roughly
treated by any knights at mine.” “By St. George! I defy them!”
exclaimed Jacques. “And even so let it rest,” said the monarch; “ride
back to Castile, and do thy worst upon the hard ribs of the Guzman.”
Jacques adopted the suggestion; and on the 3d of February, 1447,
there was not a bed in Valladolid to be had “for love or money;” so
crowded was that strong-smelling city with stronger-smelling
Spaniards, whose curiosity was even stronger than the odors they
distilled, to witness the “set-to” between the Belgian Chicken and the
Castile Shaver!
I will not detail the preliminary ceremonies, the processions to the
field, the entry of the sovereigns, the fluttering of the ladies, the
excitement of the knights, and the eagerness of the countless
multitude. Jacques was on the ground by ten o’clock, where Guzman
kept him waiting till three; and then the latter came with an axe so
much longer than that wielded by the Belgian, that even the Spanish
umpires forbade its being employed. Don Diego’s own “godfather”
for the occasion was almost minded to thump him with the handle;
and there was all the trouble in the world to induce him to select
another. This being effected, each knight was conducted to his tent,
with the understanding that he was not to issue therefrom until the
clarions had thrice sounded by way of signal. At the very first blast,
however, out rushed the Guzman, looking as ferocious as a stage
Richard who has killed five false Richmonds, and is anxiously
inquiring for the real one wherewith to finish the half-dozen. The too
volatile Don was beckoned back by the chief herald as haughtily as
when the sempiternal Widdicombe points out with his whip some
obvious duty to be performed by Mr. Merryman. Diego retired
muttering, but he again appeared in front of his tent at the second
note of summons from the trumpet, and only withdrew after the king
had assailed him “with an ugly word.” At the third “flourish,” the two
champions flew at each other, battle-axe in hand. With this weapon
they hammered at each other’s head, until there was little sense left
in either of them. At length, Diego was disarmed; then ensued a
contest made up partly of wrestling and partly of boxing; finally, they
had recourse to their swords, when the king, perceiving that murder
was likely to ensue, to one or both, threw his bâton into the lists, put
an end to the combat, and refused permission to the adversaries to
continue the struggle on horseback. The antagonists shook hands,
and the people shouted. The Spanish knight is deemed, by Belgian
chroniclers, as having come off “second best” in the struggle; but it is
also clear that Diego de Guzman was by far the “toughest customer”
that ever confronted Jacques de Lelaing. There was some jealousy
on the part of the Iberian, but his behavior was, altogether, marked
by generosity. He praised the prowess of Jacques, and presented
him with an Andalusian horse covered with the richest trappings; and
de Lelaing, as unwilling to be outdone in liberality as in fight, sent to
Guzman, by a herald, a magnificent charger, with coverings of blue
velvet embroidered in gold, and a saddle of violet velvet, to be
seated in which, was of itself a luxury. Much dancing at court
followed; and finally, the “good knight” left Valladolid loaded with gifts
from the king, praises from men, and love from the ladies, who made
surrender of more hearts than he had time to accept.
In Navarre and in Aragon he challenged all comers, but in vain.
Swords slept in scabbards, and battle-axes hung quietly from
saddle-bows, and there was more feasting than fighting. At length
Jacques, after passing through Perpignan and Narbonne, arrived at
Montpelier, where he became the guest of the famous Jacques
Cœur, the silversmith and banker of Charles VII. Old Cœur was a
hearty old host, for he offered the knight any amount of money he
would honor him by accepting; and he intimated that if De Lelaing, in
the course of his travels had found it necessary to pawn any of his
plate or jewelry, he (Jacques Cœur) would redeem it free of
expense. “My good master, the Duke of Burgundy,” replied the errant
chevalier, “provides all that is necessary for me, and allows me to
want for nothing;” and thereupon he went on his way to the court of
Burgundy, where he was received with more honor than if he had
been executing a mission for the especial benefit of humanity.
But these honors were little, compared with the rejoicings which took
place when the “good knight” revisited his native château, and the
parents who therein resided. His sire hugged him till his armor was
warm again; and his lady mother walked about the halls in a state of
ecstacy and thanksgiving. Finally, the rafters shook at the efforts of
the joyous dancers, and many a judicious matron instructed her
daughter how Jacques, who subdued the stoutest knights, might be
himself subdued by the very gentlest of ladies. The instruction was
given in vain. The good chevalier made love alike to young widows,
wives, and daughters, and having broken more hearts than he ever
broke lances, he suddenly left home in search of new adventures.
Great was the astonishment, and that altogether of a pleasurable
sort, when the herald Charolais appeared at the Scottish court in
July, 1449, and delivered a challenge from Jacques to the whole of
the Douglases. It was accepted in their name by James Douglas, the
brother of the lieutenant-general of the kingdom; and in December of
the year last named, Jacques, with a retinue of fighting uncles,
cousins, and friends, embarked at Ecluse and set sail for Caledonia.
The party were more battered about by the sea than ever they had
been by enemy on land; and when they arrived at Leith, they looked
so “shaky,” were so pale and haggard, and had so little of a
“slashing” look, wrapped up as they were in surcoats and comforters,
that the Scottish cavaliers, observing the draggled condition of the
strangers and of the plumes which seemed to be moulting from their
helmets, fairly asked them what motive induced them to come so far
in so sorry a plight, for the mere sake of getting bruised by knights
ashore after having been tossed about, sick and sorry, during whole
nights at sea. When the northern cavaliers heard that honor and not
profit had moved the Belgian company, they marvelled much thereat,
but prepared themselves, nevertheless, to meet the new-comers in
dread encounter at Stirling.
James II. presided at the bloody fray, in which three fought against
three. What the Scottish chroniclers say of the struggle, I can not
learn, but the Belgian historians describe their champions as having
been eminently victorious with every arm; and, according to them,
the Douglases were not only soundly drubbed, but took their beating
with considerable sulkiness. But there is much poetry in Belgian
history, and probably the doughty Douglas party may not have been
so thoroughly worsted as the pleasant chroniclers in question
describe them to have been. No doubt the conquerors behaved well,
as we know “les braves Belges” have never failed to do, if history
may be credited. However this may be, Jacques and his friends
hurried from Scotland, appeared at London before the meek
Lancastrian king, Henry VI.; and as the latter would not license his
knights to meet the Burgundians in the lists, the foreign fighting
gentlemen had their passports visé, and taking passage in the fast
sailer “Flower of Hainault,” duly arrived at home, where they were
hailed with enthusiasm.
Jacques had short space wherein to breathe. An English knight,
named Thomas Karr, speedily appeared at the court of Philip the
Duke, and challenged De Lelaing, for the honor of old England. This
affair caused a great sensation, and the lists were dressed in a field
near Bruges. The English knight was the heavier man in flesh and
armor, but Jacques, of course, was the favorite. Dire was the conflict.
The adversaries strove to fell each other with their axes, as butchers
do oxen. Karr paralyzed, if he did not break, the arm of Jacques; but
the Belgian, dropping his axe, closed with his foe, and after a
struggle, fell with and upon him. Karr was required, as a defeated
man, to carry the gauntlet of the victor to the lady pointed out by him.
But obstinate Tom Karr protested against this, as he had only fallen
on his elbow. The umpires declared that he had had a full fall, “head,
belly, arms, and legs;” Jacques, however, was generous and would
not insist. On the contrary, adverting to the fact that he had himself
been the first to drop his own axe, he presented Karr with a rich
diamond, as the forfeit due by him who first lost a weapon in the
combat.
Karr had terribly wounded Jacques, and the wound of the latter took
long to cure. The Duke Philip hastened his convalescence by
naming him counsellor and chamberlain; and as soon as the man so
honored by his master, had recovered from his wounds, he repaired
to Chalons on Saone, where he opened a “tourney,” which was
talked of in the country for many a long year afterward. Jacques had
vowed that he would appear in the closed lists thirty times before he
had attained his thirtieth year; and this tourney at Chalons was held
by him against all comers, in order the better to enable him to fulfil
his vow. The detail would be tedious; suffice it to say that the affair
was of barbarian magnificence, and that knights smashed one
another’s limbs, for personal honor, ladies’ love, and the glory of Our
Lady in Tears! Rich prizes were awarded to the victors, as rich
forfeits were exacted from the vanquished, and there was not only a
sea of good blood spilt in this splendidly atrocious fray, but as much
bad blood made as there was good blood shed. But then there was
empty honor acquired, a frail sort of affection gained, and an
impalpable glory added to the non-existent crown of an imaginary
Venus Victrix, decorated with the name of Our Lady of Tears! What
more could true knights desire? Chivalry was satisfied; and
commonplace men, with only common sense to direct them, had to
look on in admiring silence, at risk of being cudgelled if they dared to
speak out.
Jacques was now at the height of his renown. He was “the good
knight without fear and without doubt;” and Duke Philip placed the
last rose in his chaplet of honor, by creating him a knight of the
illustrious order of the Golden Fleece. Thus distinguished, he rode
about Europe, inviting adversaries to measure swords with him, and
meeting with none willing to accept the invitation. In 1451 he was the
embassador of Burgundy at Rome, charged to negotiate a project of
crusade against the Turks. M. Alexander Henne, the author of the
best compendium, gathered from the chronicles, of the deeds of
Jacques de Lelaing—says that after the knight’s mission to Rome,
he appeared at a passage of arms held in the park at Brussels, in
honor of the Duke of Burgundy’s son, the Count of Charolais, then
eighteen years of age, and about to mate his first appearance in the
lists. The Duchess, tender of her son as the Dowager Czarina who
kept her boys at home, and had not a tear for other mothers, whose
children have been bloodily sacrificed to the savage ambition of
Nicholas—the Duchess careful of the young Count, was desirous
that he should make essay before he appeared in the lists. Jacques
de Lelaing was accordingly selected to run a lance with him. “Three
days before the fete, the Duke, the Duchess, and the Court repaired
to the park of Brussels, where the trial was to be made. In the first
onset, the Count de Charolais shattered his lance against the shield
of Jacques, who raised his own weapon, and passed without
touching his adversary. The Duke perceived that the good knight had
spared his young adversary; he was displeased thereat, and sent
Jacques word that if he intended to continue the same course, he
would do well to meddle no further in the matter. Other lances were
then brought, and Jacques, running straight against the Count, both
lances flew into splinters. At this incident, the Duchess, in her turn,
gave expression to her discontent; but the Duke only laughed; and
thus mother and father were of different opinions; the one desiring a
fair trial, the other security for her son.” On the day of the great
tourney, there were assembled, with the multitude, on the great
square at Brussels, not less than two hundred and twenty-five
princes, barons, knights, and squires. Some of the noblest of these
broke a lance with, and perhaps the limbs of, their adversaries. The
Count de Charolais broke eighteen lances on that day, and he
carried off the the prize, which was conferred upon him by the ladies.
This was the last of the show-fights in which Jacques de Lelaing
exhibited himself. The bloodier conflicts in which he was
subsequently engaged, were far less to his credit. They formed a
part of the savage war which the despotic Duke and the nobles
carried on against the free and opulent cities, whose spirit of liberty
was an object of hatred, and whose wealth was an object of
covetous desire, to the Duke and his body of gentleman-like
assassins. Many a fair town was devastated by the Duke and his
followers, who affected to be inspired by religious feelings, a desire
for peace, and a disinclination to make conquests. Whereby it may
be seen that the late Czar was only a Burgundian duke enlarged,
impelled by much the same principle, and addicted to a similar sort
of veracity. It was a time of unmitigated horrors, when crimes enough
were committed by the nobles to render the name of aristocracy for
ever execrable throughout Belgium; and atrocities were practised by
the enraged commons, sufficient to insure, for the plebeians, the
undying hatred of their patrician oppressors. There was no respect
on either side for age, sex, or condition. The people, of every
degree, were transformed into the worst of fiends—slaying, burning,
violating, and plundering; and turning from their accursed work to
kneel at the shrine of that Mary whose blessed Son was the Prince
of Peace. Each side slaughtered, hung, or drowned its prisoners; but
the nobles gave the provocation by first setting the example, and the
commons were not cruel till the nobility showed itself alike destitute
of honor and of mercy. The arms of the popular party were nerved by
the infamy of their adversaries, but many an innocent man on either
side was condemned to suffer, undeservedly, for the sins of others.
The greatest efforts were made against the people of the district and
city of Ghent, but all Flanders sympathized with them in a war which
was considered national. In the struggle, the Duke won no victory
over the people for which the latter did not compel him to pay a
frightful price; he was heartily sick of the war before it was half
concluded—even when his banner was being most successfully
upheld by the strong arm and slender scruples of Jacques de
Lelaing.
The good knight was however, it must be confessed, among the few
—if he were not the only one—of the betterminded nobles. He had
been commissioned by the Duke to set fire to the Abbey of
Eenaeme, and he obeyed without hesitation, and yet with reluctance.
He destroyed the religious edifice with all which it contained, and
which could be made to burn; but having thus performed his duty as
a soldier, he forthwith accomplished his equally bounden duty, as a
Christian—and, after paying for three masses, at which he devoutly
assisted, he confessed himself to a predicant friar, “making a case of
conscience,” says one of his biographers, “of having, out of respect
for discipline, committed an act which the uprightness of his heart
compelled him to condemn as criminal.” Never was there a better
illustration of that so-called diverse condition of things which is said
to represent a distinction without a difference.
The repentance of Jacques de Lelaing came, it is hoped, in time. He
did well, at all events, not to defer it any longer, for he was soon on
the threshold of that world where faith ceases and belief begins. He
was engaged, although badly wounded, in inspecting the siege-
works in the front of the Chateau de Pouckes, that Flemish cradle of
the Pooks settled in England. It was on a June afternoon of the year
1453, that Jacques, with a crowd of nobles half-encircling him, rode
out, in spite of the protest of his doctors (because, as he said, if he
were to remain doing nothing he should certainly die), in order that
he might have something to do. There was a famous piece of
artillery on the Burgundian side, which was sorely troublesome to the
stout little band that was defending Pouckes. It was called the
“Shepherdess,” but never did shepherdess speak with so
thundering-unlovely a voice, or fling her favors about her with such
dire destruction to those upon whom they were showered. Jacques
drew up behind the manteau of this cannon, to watch (like our gallant
seamen at Sebastopol) the effects of the shot discharged from it. At
the same moment a stone projectile, discharged from a culverin by
the hand of a young artilleryman of Ghent, who was known as the
son of Henry the Blindman, struck Jacques on the forehead, carrying
away the upper part of his head, and stretched him dead upon the
field. A Carmelite brother rushed up to him to offer the succor and
consolation of religion, but it was too late. Jacques had sighed out
his last breath, and the friar decently folded the dead warrior’s arms
over his breast. A mournful troop carried the body back to the camp.
The hero of his day died in harness. He had virtues that fitted him for
a more refined, a more honest, in short, a more Christian, period.
These he exercised whenever he could find opportunity, but such
opportunity was rare. He lived at a period when, as M. de Sismondi
has remarked, “Knights thought of nothing but equalling the Rolands
and Olivers of the days of Charlemagne, by the destruction of the
vile canaille”—a sort of pastime which has been recently
recommended in our senate, although the days of chivalry be gone.
The noble comrades of Jacques, as M. Henne observes,
acknowledged but one species of supreme pleasure and glory, which
consisted in making flow abundantly the blood of villains—or, as they
are now called, the lower orders. But in truth the modern “villain” or
the low-class man is not exclusively to be found in the ranks which
have had such names applied to them. As Bosquier-Gavaudan used
so joyously to sing, some thirty years ago, in the Ermite de St.
Avelle:—

“Les gens de bien


Sont souvent des gens de rien;
Et les gens de rien
Sont souvent des gens de bien!”

For a knight, Jacques was really a respectable man, and so


disgusted with his butcher-like occupation, that, just before his death,
he had resolved to surrender his estate to a younger brother, and,
since fate had made of him a licensed murderer, to henceforth
murder none but eastern infidels—to slay whom was held to be more
of a virtue than a sin. Let us add of him, that he was too honest to
earn a reputation by being compassionate to half-a-dozen helpless
foes, after directing his men to slaughter a score of the mutilated and
defenceless enemy. Jacques de Lelaing would sooner have sent his
dagger up to the hilt in his own heart, than have violated the
safeguard of a flag of truce. Such days and such doings of chivalry
are not those most agreeable to Russian chivalry. Witness Odessa,
where the pious governor directed the fire on a flag of truce which he
swore he could not see; and witness the massacre of Hango, the
assassins concerned in which exploit were defended by their worthy
superior De Berg.
Jacques de Lelaing, however, it must not be forgotten, fell in a most
unworthy cause—that of a despot armed against free people. His
excellent master swore to avenge him; and he kept his word. When
the Château de Pouckes was compelled to surrender, Philip the
Good ordered every one found alive in it to be hung from the walls.
He made exception only of a priest or two, one soldier afflicted with
what was called leprosy, but which has now another name in the
catalogue of avenging maladies, and a couple of boys. It was
precisely one of these lads who had, by his well-laid shot, slain “the
good knight without fear and without doubt;” but Philip was not aware
of this till the lad was far beyond his reach, and in safety at Ghent.
Those who may be curious to know the course taken by the war until
it was terminated by the treaty of Lille, are recommended to study
the Chronicles of De Lettenhooe, of Olivier de la Marche, of
Chastellain, and Du Clery. I had no intention, at setting out, to paint a
battle-piece, but simply to sketch a single figure. My task is done,
however imperfectly, and, as old chroniclers were wont to say, May
Heaven bless the gentle reader, and send pistoles and abounding
grace to the unworthy author.
Such is the history of an individual; let us now trace the fortunes of a
knightly house. The story of the Guises belongs entirely to chivalry
and statesmanship.
THE FORTUNES OF A KNIGHTLY FAMILY.
“This deals with nobler knights and monarchs,
Full of great fears, great hopes, great enterprises.”
Antony Brewer, “Lingua.”

In the pleasant spring-time of the year 1506, a little boy, mounted on


a mule, and accompanied by a serving man on foot, crossed over
the frontier from Lorraine into France. The boy was a pretty child,
some ten years old. He was soberly clad, but a merry heart beat
under his gray jerkin; and his spirits were as light as the feather in
his bonnet. The servant who walked at his side was a simple yet
faithful follower of his house; but there was no more speculation in
his face than there was in that of the mule. Nothing could have
looked more harmless and innocent than the trio in question; and yet
the whole—joyous child, plodding servitor, and the mule whose bells
rang music as he trod—formed one of the most remarkable
invasions of which the kingdom of France has ever been the victim.
The boy was the fifth child of René and Philippa de Gueldres, the
ducal sovereigns of Lorraine. This duchy, a portion of the old
kingdom of Lotharingia—in disputes for the possession of which the
children of Charlemagne had shed rivers of blood—had maintained
its independence, despite the repeated attempts of Germany and
France to reduce it to subjection. At the opening of the sixteenth
century, it had seen a legal succession of sovereign and
independent masters during seven centuries. The reigning duke was
René, the second of that name. He had acquired estates in France,
and he had inherited the hatred of Lorraine to the Capetian race
which had dethroned the heirs of Charlemagne. It was for this double
reason that he unostentatiously sent into the kingdom of France one
of his sons, a boy of fair promise. The mission of the yet
unconscious child was to increase the territorial possessions of his
family within the French dominions, and ultimately to rule both
Church and State—if not from the throne, why then from behind it.
The merry boy proved himself in course of time to be no unfitting
instrument for this especial purpose. He was brought up at the
French court, studied chivalry, and practised passages of arms with
French knights; was the first up at réveillée, the last at a feast, the
most devout at mass, and the most winning in ladies’ bower. The
princes of the blood loved him, and so did the princesses. The army
hailed him with delight; and the church beheld in him and his brother,
Cardinal John, two of those champions whom it records with
gladness, and canonizes with alacrity.
Such was Claude of Lorraine, who won the heart and lands of
Antoinette de Bourbon, and who received from Francis I. not only
letters of naturalization, but the title of Duke of Guise. The locality so
named is in Picardy. It had fallen to the house of Lorraine by
marriage, and the dignity of Count which accompanied it was now
changed for that of Duke. It was not long before Claude made the
title famous. The sword of Guise was never from his grasp, and its
point was unceasingly directed against the enemies of his new
country. He shed his own blood, and spilled that of others, with a
ferocious joy. Francis saw in him the warmest of his friends and the
bravest of his soldiers. His bravery helped to the glory that was
reaped at Marignan, at Fontarabia, and in Picardy. Against internal
revolt or foreign invasion he was equally irresistible. His sword drove
back the Imperialists of Germany within their own frontier; and when
on the night of Pavia the warriors of France sat weeping like girls
amid the wide ruin around them, his heart alone throbbed with
hopeful impulses, and his mind only was filled with bright visions of
victories to come.
These came indeed, but they were sometimes triumphs that earned
for him an immortality of infamy. The crest of his house was a double
cross, and this device, though it was no emblem of the intensity of
religion felt by these who bore it, was significant of the double
sanguinary zeal of the family—a zeal employed solely for selfish
ends. The apostolic reformers of France were, at this period, in a
position of some power. Their preachers were in the pulpit, and their
people in the field. They heard the gospel leaning on their swords;

You might also like