190
Instructive Matrix Cues for Tissue Engineering Applications by Yun Xiao A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Chemical Engineering and Applied Chemistry Institute of Biomaterials and Biomedical Engineering University of Toronto © Copyright by Yun Xiao 2016

Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

Instructive Matrix Cues for Tissue Engineering Applications

by

Yun Xiao

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Department of Chemical Engineering and Applied Chemistry

Institute of Biomaterials and Biomedical Engineering University of Toronto

© Copyright by Yun Xiao 2016

Page 2: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

ii

Instructive Matrix Cues for Tissue Engineering Applications

Yun Xiao

Doctor of Philosophy

Department of Chemical Engineering and Applied Chemistry

Institute of Biomaterials and Biomedical Engineering

University of Toronto

2016

Abstract

Tissue engineering holds the promise of generating functional tissues to replace, or regenerate

impaired native tissues by combining knowledge from cell biology, material science, and

engineering. Designing instructive biomaterials that harness regeneration potential of native

tissue requires incorporation of biochemical and biophysical cues. Besides, micro-tissues

analogues are generated by emerging microfabrication techniques with precise control. This

thesis aims to facilitate cell-matrix interactions by biochemical and topographical cues in

collagen-based matrix for tissue regeneration in vivo with a focus on cardiac tissue engineering

and wound healing applications, and creating cardiac micro-tissues with better physiological

relevance in vitro.

Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering

applications, with an emphasis on cardiac tissue engineering and wound healing applications.

Chapter 3 describes covalent immobilization of peptides or angiogenic growth factors on

collagen scaffolds. Immobilization efficiency and release profile were characterized and showed

sustained releases. The modification did not affect the porous structure and tensile strength of the

collagen scaffolds.

Page 3: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

iii

Chapter 4 describes the study on protective effect of QHREDGS peptide on primary human

keratinocytes and its efficacy in promoting diabetic wound healing. In vitro studies on normal

and diabetic human keratinocytes showed that immobilized QHREDGS promoted keratinocyte

attachment, collective migration, and survival against H2O2 stress. QHREDGS immobilized in

chitosan-collagen hydrogel accelerated wound healing in diabetic mice by promoting re-

epithelialization in vivo.

Chapter 5 describes the design of a microfabricated bioreactor providing topographical cues to

generate cardiac micro-tissues recapitulating native cardiac bundles. The micro-tissues were

perfusable with micro-tubing in the center, mimicking capillaries. We demonstrated the utility of

this platform by investigating the effects of nitric oxide on electrophysiological properties of the

perfusable cardiac biowires.

We finally conclude with remarks on the future prospects for biochemical and topographical cues

in instructive biomaterials design for tissue engineering applications.

Page 4: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

iv

Acknowledgments

First of all, I would like to thank my supervisor Dr. Milica Radisic for giving me the opportunity

to work on these studies and her support and guidance over the years. Milica has inspired me to

be the best scientist I could be with challenges and encouragement throughout my thesis, and her

enthusiasm and vision in science. I am thankful for every opportunity she provided for me to

grow as a graduate student.

I would also like to thank my committee members, Dr. Michael Sefton and Dr. Alison McGuigan,

for their valuable feedbacks on my thesis. Thanks also to Dr. Edgar Acosta for being my

departmental examiner and to Dr. Todd Hoare for being my external examiner. The invaluable

advices from these experts from different research area made my thesis the best it could be.

It is my greatest pleasure to work with previous and present members of the Laboratory for

Functional Tissue Engineering since 2010: Rohin Iyer, Loraine Chiu, Anne Hsieh, Lewis Reis,

Katherine Chiang, Hannah Song, Nimalan Thavandiran, Larry Meng, Iran Rashedi, Boyang

Zhang, Sara Nunes, Jason Miklas, Lan Dang, Aarash Sofla, Mark Li, Lara Fu, Kujaany Kana,

Nicole Feric, Carol Laschinger, Dario Bogojevic, Miles Montgomery, Yimu Zhao, Aric Pahnke,

Locke Davenport Huyer, Genna Conant, Erica Knee, Anastasia Korolj, Stasja Drecun, Junhao

Gu, Shuwen Cao, Samad Ahadian, and Ben Lai. All of you have provided great support and help

over the years and I am ever so grateful.

I also acknowledge the opportunities to work with and learn from my collaborators: Dr. Kang

Kai and Dr. Lu Sun from Dr. Ren-Ke Li’s group, Haijiao Liu from Dr. Yu Sun and Dr. Craig

Simmons’ group, Dr. Mark Gagliardi from Dr. Dordon Keller’s group, Dr. Lindsay Fitzpatrick

from Dr. Michael Sefton’s group, and Camila Londono from Dr. Alison McGuigan’s group. I

would also like to thank A.J. Wang from the animal facility for his help with my animal studies.

To all the animals who were sacrificed in my studies, I acknowledge your contribution in the

name of science.

I would like to thank my friends here who made Toronto my second home and the others who

have always been there for me despite the thousands of miles between us. To my boyfriend,

Haijiao Liu, without whom I could not have come this far. I am so thankful that you came into

Page 5: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

v

my life at the beginning of my graduate study and changed it throughout. You have been a great

support for me through the stressful times, the person I can share everything with, a great

company when I work overtime, an inspiring young scientist to have a discussion with, and an

example of self-motivation and determination to me.

Finally, I dedicate this thesis to my parents, who love me unconditionally and always allow me

to pursue my dreams. I feel blessed to have such loving and supportive parents and grandparents,

and sorry that I was not always there to be with you.

Page 6: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

vi

Declaration of Co-Authorship

The original scientific content of the thesis is comprised of three previously published, peer

reviewed articles in internationally recognized journals and a fourth article submitted. The

literature review is from one published review paper, one review paper in preparation for

submission, and two published book chapters, the transcribed writing being that of Yun Xiao.

The contributions of co-authors are stated in the thesis, in conformity with the requirements for

the degree of Doctor of Philosophy.

Page 7: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

vii

Abstracts of Published Articles Appearing in the Thesis

Modifications of biomaterials with immobilized growth factors or peptides for tissue

engineering applications.

Xiao Y, Reis L, Zhao Y, Radisic M. Methods 2015;84:44-52.

In order to provide an instructive microenvironment to facilitate cellular behaviors and tissue

regeneration, biomaterials can be modified by immobilizing growth factors or peptides. We

describe here our procedure for modification of collagen-based biomaterials, both porous

scaffolds and hydrogel systems, with growth factors or peptides by covalent immobilization.

Characterizations of the modified biomaterials (immobilization efficiency, release profile,

morphology, mechanical strength, and rheology) and in vitro testing with cells are also discussed.

Contributions: Y.X.-concept and design, performed experiments and data analysis, manuscript

writing; L.R.-concept and design, performed experiments and data analysis, manuscript writing;

Y.Z.-concept and design, manuscript writing; M.R.-concept, data interpretation, final approval of

manuscript.

Page 8: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

viii

Aged human cells rejuvenated by cytokine enhancement of biomaterials for surgical

ventricular restoration.

Kang K*, Sun L*, Xiao Y, Li SH, Wu J, Guo J, Jiang S, Yang L, Yao TM, Weisel RD, Radisic

M, Li RK. J Am Coll Cardiol 2012;60:2237–49. (* equal contribution)

Objectives: This study investigated whether cytokine enhancement of a biodegradable patch

could restore cardiac function after surgical ventricular restoration (SVR) even when seeded with

cells from old donors.

Background: SVR can partially restore heart size and improve function late after an extensive

anterior myocardial infarction. However, 2 limitations include the stiff synthetic patch used and

the limited healing of the infarct scar in aged patients.

Methods: We covalently immobilized 2 proangiogenic cytokines (vascular endothelial growth

factor and basic fibroblast growth factor) onto porous collagen scaffolds. We seeded human

mesenchymal stromal cells from young (50.0 ± 8.0 years, N = 4) or old (74.5 ± 7.4 years, N = 4)

donors into the scaffolds, with or without growth factors. The patches were characterized and

used for SVR in a rat model of myocardial infarction. Cardiac function was assessed.

Results: In vitro results showed that cells from old donors grew slower in the scaffolds.

However, the presence of cytokines modulated the aging-related p16 gene and enhanced cell

proliferation, converting the old cell phenotype to a young phenotype. In vivo studies showed

that 28 days after SVR, patches seeded with cells from old donors did not induce functional

recovery as well as patches seeded with young cells. However, cytokine-enhanced patches

seeded with old cells exhibited preserved patch area, prolonged cell survival, and augmented

angiogenesis, and rats implanted with these patches had better cardiac function. The patch

became an elastic tissue, and the old cells were rejuvenated.

Conclusions: This sustained-release, cytokine-conjugated system provides a promising platform

for engineering myocardial tissue for aged patients with heart failure.

Page 9: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

ix

Contributions: K.K., L.S.-concept and design, performed in vitro and in vivo experiments and

data analysis, manuscript writing; Y.X.-design, scaffold preparation and characterization,

manuscript writing; S.H.L., J.W., J.G., S.J., L.Y., T.M.Y.-performed surgical work and

evaluation, R.D.W., M.R., R.K.L.-concept, data interpretation, final approval of manuscript.

Page 10: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

x

Diabetic wound regeneration using peptide-modified hydrogels targeting the epithelium

Xiao Y, Feric N, Knee EJ, Gu J, Cao S, Laschinger CA, Londono C, McGuigan AP, Radisic M.

Submitted to Proceedings of the National Academy of Sciences.

There is a clinical need for new, more effective treatments for chronic wounds in diabetic

patients. Lack of epithelial cell migration is a hallmark of non-healing wounds and diabetes often

involves endothelial dysfunction. Therefore, targeting re-epithelialization, which mainly involves

keratinocytes, may improve therapeutic outcomes of current treatments that mostly focus on

angiogenesis. In this study, we present an integrin-binding prosurvival peptide derived from

angiopoietin-1, QHREDGS, as a novel therapeutic candidate for diabetic wound treatments by

demonstrating its efficacy in promoting human primary keratinocytes attachment, survival,

collective migration, and Akt and MAPKp42/44 activation. The QHREDGS peptide, both as a

soluble supplement and when immobilized in a substrate, protected keratinocytes against

hydrogen peroxide stress in a dose dependent manner. Collective migration of both normal and

diabetic human keratinocytes was promoted on chitosan-collagen films immobilized with the

QHREDGS peptide. The clinical relevance was further demonstrated by assessing the

QHREDGS-immobilized chitosan-collagen hydrogel in full-thickness excisional wounds in a

db/db diabetic mouse model, which showed accelerated wound closure compared to peptide-free

hydrogel and blank wound controls. Furthermore, the accelerated wound closure was primarily

due to faster re-epithelialization and increased granulation tissue formation. There were no

observable differences in blood vessel density or size within the wound. Together, these findings

indicate that QHREDGS is a promising candidate for new wound-healing interventions that

enhance re-epithelialization and granulation tissue formation.

Contributions: Y.X. designed and performed experiments, analyzed data and prepared the

manuscript. N.F. analyzed data and prepared the manuscript. E.J.K. analyzed data. J.G. and S.C.

preformed peptide conjugation and analyzed data. C.A.L. performed Western blotting and

analyzed data. C.L. performed a preliminary migration experiment. A.P.M. provided feedback on

the design of the collective migration experiments, suggested appropriate controls and helped

with data presentation. M.R. supervised the work and wrote the manuscript.

Page 11: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xi

Microfabricated perfusable cardiac biowire: a platform that mimics native cardiac bundle.

Xiao Y, Zhang B, Liu H, Miklas JW, Gagliardi M, Pahnke AQ, Thavandiran N, Sun Y,

Simmons C, Keller G, Radisic M. Lab Chip 2014;14:869–82.

Tissue engineering enables the generation of three-dimensional (3D) functional cardiac tissue for

pre-clinical testing in vitro, which is critical for new drug development. However, current tissue

engineering methods poorly recapitulate the architecture of oriented cardiac bundles with

supporting capillaries. In this study, we designed a microfabricated bioreactor to generate 3D

micro-tissues, termed biowires, using both primary neonatal rat cardiomyocytes and human

embryonic stem cell (hESC) derived cardiomyocytes. Perfusable cardiac biowires were

generated with polytetrafluoroethylene (PTFE) tubing template, and were integrated with

electrical field stimulation using carbon rod electrodes. To demonstrate the feasibility of this

platform for pharmaceutical testing, nitric oxide (NO) was released from perfused sodium

nitroprusside (SNP) solution and diffused through the tubing. The NO treatment slowed down

the spontaneous beating of cardiac biowires based on hESC derived cardiomyocytes and

degraded the myofibrillar cytoskeleton of the cardiomyocytes within the biowires. The biowires

were also integrated with electrical stimulation using carbon rod electrodes to further improve

phenotype of cardiomyocytes, as indicated by organized contractile apparatus, higher Young's

modulus, and improved electrical properties. This microfabricated platform provides a unique

opportunity to assess pharmacological effects on cardiac tissue in vitro by perfusion in a cardiac

bundle model, which could provide improved physiological relevance.

Contributions: Y.X.-concept and design, performed experiments and data analysis, manuscript

writing; B.Z.-design, device fabrication; H.L.-performed AFM experiments and data analysis;

J.W.M., M.G., A.Q.P.-cardiomyocyte differentiation from human embryonic stem cells (hESCs);

N.T.-hydrogel composition development; Y.S., C.S., K.G., M.R.-concept, data interpretation,

final approval of manuscript.

Page 12: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xii

Table of Contents

Abstract ........................................................................................................................................... ii

Acknowledgments .......................................................................................................................... iv

Declaration of Co-Authorship ........................................................................................................ vi

Abstracts of Published Articles Appearing in the Thesis ............................................................. vii

Table of Contents .......................................................................................................................... xii

List of Tables .............................................................................................................................. xvii

List of Figures ............................................................................................................................ xviii

List of Abbreviations .................................................................................................................... xx

Chapter 1 ......................................................................................................................................... 1

1 Introduction ................................................................................................................................ 1

1.1 Overview ............................................................................................................................. 1

1.2 Hypothesis ........................................................................................................................... 2

1.3 Specific aims ....................................................................................................................... 3

Chapter 2 ......................................................................................................................................... 5

2 Literature review ........................................................................................................................ 5

2.1 Instructive biomaterials for tissue engineering ................................................................... 5

2.1.1 Motivation for instructive biomaterials .................................................................. 5

2.1.2 Naturally derived biomaterials ................................................................................ 7

2.1.3 Instructive biochemical cues provided by biomaterials ........................................ 10

2.1.4 Biomechanical instructions provided by biomaterials .......................................... 12

2.1.5 Mesenchymal stromal cells ................................................................................... 13

2.2 Cardiac tissue engineering ................................................................................................ 15

2.2.1 Motivation for cardiac tissue engineering ............................................................. 15

Page 13: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xiii

2.2.2 Cell sources ........................................................................................................... 15

2.2.3 Biomaterials .......................................................................................................... 20

2.3 Tissue engineering for wound healing .............................................................................. 23

2.3.1 Wound healing process ......................................................................................... 23

2.3.2 Diabetic wound healing ........................................................................................ 26

2.3.3 Current tissue engineering products for topical wounds ....................................... 27

2.3.4 Instructive biochemical cues for wound healing .................................................. 28

2.4 Cardiac tissue engineering in vitro ................................................................................... 36

2.4.1 Motivation for generating cardiac micro-tissues .................................................. 36

2.4.2 Cardiac micro-tissues as research platform .......................................................... 39

Chapter 3 ....................................................................................................................................... 42

3 Collagen patches immobilized with growth factors or peptides for cardiac regeneration ....... 42

3.1 Introduction ....................................................................................................................... 42

3.2 Materials and methods ...................................................................................................... 44

3.2.1 Materials ............................................................................................................... 44

3.2.2 Covalent immobilization of growth factors and peptides on collagen scaffolds .. 44

3.2.3 Quantification of growth factor immobilization efficiency .................................. 45

3.2.4 Quantification of QHREDGS peptide immobilization efficiency ........................ 46

3.2.5 Characterization of release profile ........................................................................ 47

3.2.6 Scanning electron microscopy .............................................................................. 47

3.2.7 Tensile testing of porous collagen scaffolds ......................................................... 48

3.3 Results and discussion ...................................................................................................... 50

3.4 Conclusion ........................................................................................................................ 54

3.5 Acknowledgments ............................................................................................................. 54

Chapter 4 ....................................................................................................................................... 55

Page 14: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xiv

4 Diabetic wound regeneration using peptide-modified hydrogel targeting the epithelium ....... 55

4.1 Introduction ....................................................................................................................... 55

4.2 Materials and methods ...................................................................................................... 56

4.2.1 Primary human keratinocytes cell culture ............................................................. 56

4.2.2 Evaluation of soluble QHREDGS in vitro ............................................................ 57

4.2.3 Proliferation assay ................................................................................................. 57

4.2.4 H2O2 treatment on HEKs with soluble QHREDGS peptide ................................. 57

4.2.5 Conjugation of QHREDGS to chitosan ................................................................ 58

4.2.6 Solvent casting of chitosan-collagen films ........................................................... 58

4.2.7 Coating validation ................................................................................................. 58

4.2.8 Keratinocyte attachment on chitosan-only films .................................................. 59

4.2.9 H2O2 treatment on keratinocytes on the chitosan-collagen films ......................... 59

4.2.10 EarlyToxTM Cell Integrity assay ........................................................................... 59

4.2.11 Western blotting .................................................................................................... 60

4.2.12 Migration assay ..................................................................................................... 60

4.2.13 Immunostaining .................................................................................................... 61

4.2.14 Animals, wound model, and treatment ................................................................. 61

4.2.15 Histology analysis ................................................................................................. 62

4.2.16 Microvessel analysis algorithm ............................................................................. 63

4.2.17 Statistical analysis ................................................................................................. 63

4.3 Results ............................................................................................................................... 63

4.3.1 QHREDGS peptide prevents H2O2-induced apoptosis in human primary

keratinocytes and upregulates Akt and MAPKp42/44 signaling .............................. 63

4.3.2 Immobilized QHREDGS peptide promotes human primary keratinocytes

attachment, survival and migration in vitro .......................................................... 66

Page 15: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xv

4.3.3 Immobilized QHREDGS peptide promotes diabetic human primary

keratinocytes attachment, survival and migration in vitro .................................... 71

4.3.4 QHREDGS-immobilized hydrogel promotes wound healing in db/db diabetic

mice ....................................................................................................................... 74

4.3.5 Accelerated QHREDGS-induced diabetic wound healing does not involve

changes in the extent of angiogenesis of the granulation tissue ........................... 79

4.4 Discussion ......................................................................................................................... 82

4.5 Conclusion ........................................................................................................................ 86

4.6 Acknowledgments ............................................................................................................. 87

Chapter 5 ....................................................................................................................................... 88

5 Microfabricated perfusable cardiac biowire: a platform that mimics native cardiac bundle ... 88

5.1 Introduction ....................................................................................................................... 88

5.2 Materials and methods ...................................................................................................... 90

5.2.1 Biowire bioreactor design and fabrication ............................................................ 90

5.2.2 Perfusion system design and fabrication ............................................................... 90

5.2.3 Cell culture ............................................................................................................ 90

5.2.4 Generation of cardiac biowires ............................................................................. 91

5.2.5 Quantification of compaction rate ........................................................................ 92

5.2.6 Immunostaining and Fluorescent Microscopy ...................................................... 92

5.2.7 Quantification of nuclei elongation and alignment ............................................... 92

5.2.8 Characterization of perfusable biowires ............................................................... 92

5.2.9 Quantification of NO perfusion ............................................................................ 93

5.2.10 NO treatment of human cardiac biowires ............................................................. 93

5.2.11 Electrical stimulation ............................................................................................ 94

5.2.12 Atomic force microscopy (AFM) ......................................................................... 95

5.2.13 Statistical analysis ................................................................................................. 95

Page 16: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xvi

5.3 Results ............................................................................................................................... 96

5.3.1 Generation and characterization of cardiac biowires ............................................ 96

5.3.2 Generation and characterization of perfusable cardiac biowires ........................ 100

5.3.3 NO treatment of human cardiac biowires by perfusion ...................................... 101

5.3.4 Electrical stimulation of cardiac biowires ........................................................... 103

5.4 Discussion ....................................................................................................................... 105

5.5 Conclusion ...................................................................................................................... 109

5.6 Acknowledgments ........................................................................................................... 109

Chapter 6 ..................................................................................................................................... 110

6 Discussion and conclusions .................................................................................................... 110

6.1 Discussion ....................................................................................................................... 110

6.2 Significant contributions ................................................................................................. 114

6.3 Conclusion ...................................................................................................................... 116

Chapter 7 ..................................................................................................................................... 117

7 Recommendations for future work ......................................................................................... 117

7.1 Investigate cardiac regeneration by collagen patch immobilized with QHREDGS

peptide ............................................................................................................................. 117

7.2 Determine the mechanism of accelerated keratinocyte collective migration promoted

by QHREDGS peptide .................................................................................................... 117

7.3 Improve the perfusable biowire for drug candidates with high molecular weight ......... 118

7.4 Investigate the synergy between biochemical cues and topographical cues ................... 118

References ................................................................................................................................... 120

Appendices List of publications and contributions ..................................................................... 161

Copyright Acknowledgements .................................................................................................... 165

Page 17: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xvii

List of Tables

Table 2-1 Principal tissue distribution and cells of origin for different collagen types in human

body ................................................................................................................................................. 8

Table 2-2 Principal properties of chitosan in relation to its use in biomedical applications ........ 10

Page 18: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xviii

List of Figures

Figure 2-1 Chemical structure of chitosan, comprising N-acetyl-D-glucosamine (right) and D-

glucosamine (left) units. .................................................................................................................. 9

Figure 2-2 Three classic stages of wound healing. ....................................................................... 25

Figure 2-3 Different biochemical cues provided by matrix to regulate the native cells. .............. 29

Figure 2-4 Engineering heart tissue for replacement therapeutics and in vitro models by physical

and electrical control of cells and microenvironment. .................................................................. 38

Figure 2-5 Strategies for generating 2D and 3D cardiac tissue in vitro. ....................................... 41

Figure 3-1 Reaction diagram for immobilization of growth factors or peptides on collagen

sponges using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide HCl (EDC) and N-

hydroxysulfosuccinimide (Sulfo-NHS). ....................................................................................... 44

Figure 3-2 Characterization of scaffolds. ...................................................................................... 51

Figure 3-3 Characterisation of peptide immobilization. ............................................................... 53

Figure 4-1 Soluble QHREDGS peptide prevents H2O2-induced cell death in human primary

keratinocytes with up-regulation of Akt and MAPK phosphorylation. ........................................ 65

Figure 4-2 The presence of soluble QHREDGS peptide does not accelerate HEKs migration on

collagen coated surfaces. .............................................................................................................. 66

Figure 4-3 Immobilized QHREDGS peptide in chitosan-collagen films promotes human neonatal

primary keratinocytes survival and migration. ............................................................................. 68

Figure 4-4 The presence of the immobilized QHREDGS peptide promotes HEK attachment on

chitosan-only films. ....................................................................................................................... 69

Figure 4-5 HEKs form calcium-induced adherens junctions during migration and the accelerated

migration is not associated with a difference in cell density. ....................................................... 70

Page 19: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xix

Figure 4-6 Immobilized QHREDGS peptide in chitosan-collagen films promotes diabetic adult

human primary keratinocyte survival and migration. ................................................................... 72

Figure 4-7 The presence of immobilized QHREDGS peptide promotes DHEK attachment on

chitosan-only films. ....................................................................................................................... 73

Figure 4-8 DHEKs form adherens junctions during migration and the accelerated migration is not

associated with a difference in cell density. .................................................................................. 74

Figure 4-9 QHREDGS-immobilized hydrogel promotes wound healing in db/db diabetic mice. 77

Figure 4-10 Thickness of the unwounded epidermis. ................................................................... 78

Figure 4-11 An example of wound re-epithelialized after two weeks with a single treatment of

QHREDGS peptide in the chitosan-collagen hydrogel. ............................................................... 78

Figure 4-12 The improvements in the diabetic wound healing process induced by the QHREDGS

peptide are not associated with increased angiogenesis within the granulation tissue. ................ 79

Figure 4-13 QHREDGS peptide does not affect microvessel number and size within granulation

tissue. ............................................................................................................................................ 81

Figure 5-1 Cardiac bundles in native myocardium. ...................................................................... 96

Figure 5-2 Generation of cardiac biowires with microfabricated bioreactor. ............................... 97

Figure 5-3 The suture template provides topographical cues in the biowires for the

cardiomyocytes to elongate and align. .......................................................................................... 99

Figure 5-4 Generation of perfusable cardiac biowires. ............................................................... 100

Figure 5-5 Nitric oxide (NO) treatment on human tubing-templated biowires. ......................... 102

Figure 5-6 Electrical stimulation and perfusion of cardiac biowires. ......................................... 104

Page 20: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xx

List of Abbreviations

2D Two-dimensional

3D Three-dimensional

ABTS 2,2'-Azinobis [3-ethylbenzothiazoline-6-sulfonic acid]-diammonium salt

AFM Atomic Force Microscopy

Ang1 Angiopoietin-1

bFGF Basic fibroblast growth factor

BMP-2 Bone morphogenetic protein-2

BrdU Bromodeoxyuridine

BSA Bovine serum albumin

CABG Coronary artery bypass grafting

cDNA Complimentary DNA

CPCs Cardiac progenitor cells

cTnT Cardiac troponin T

DAPI 4',6-diamidino-2-phenylindole

DHEKs Diabetic human adult epithelial keratinocytes

DTMRI Diffusion tensor magnetic resonance imaging

EB Embryoid body

ECM Extracellular matrix

Page 21: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xxi

EDC 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide HCl

EDGS EpiLife Defined Growth Supplement

ELISA Enzyme-linked immunosorbent assay

eNOS Endothelial nitric oxide synthase

EPCs Endothelial progenitor cells

ESCs Embryonic stem cells

ET Excitation threshold

FDA Food and Drug Administration

GAG Glycosaminoglycan

HBOT Hyperbaric oxygen therapies

HEKs Human epithelial keratinocytes

HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid)

HGF Hepatocyte growth factor

HIF Hypoxia-inducible factor

ID Inner diameter

IL-1β Interleukin 1β

iNOS Induced nitric oxide synthase

iPSCs Induced pluripotent stem cells

KGF Keratinocyte growth factors

Page 22: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xxii

LDH Lactate dehydrogenase

LQT3 Long QT syndrome type 3

MAPK Mitogen-activated protein kinase

MCR Maximum capture rate

MEA Microelectrode arrays

MEFs Mouse embryonic fibroblasts

MES 2-(N-morpholino)ethanesulfonic acid

MI Myocardial infarction

miRNA Micro RNA

MMP Matrix metalloproteinase

mRNA Messenger RNA

MSCs Mesenchymal stromal cells

nNOS Neuronal nitric oxide synthase

NOS Nitric oxide synthase

OD Outer diameter

PBAE Poly(β-amino esters)

PBS Phosphate buffered saline

PCL Polycaprolactone

PDGF Platelet derived growth factor

Page 23: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xxiii

PDMS Poly(dimethysiloxane)

PEG Poly(ethylene glycol)

PGA Polyglycolic acid

PGS Poly(glycerol sebacate)

PHD2 Prolyl hydroxylase domain 2

pHEMA Poly(2-hydroxyethyl methacrylate)

PLLA Poly(L-lactide)

PNIPAAm Poly-N-isopropylacrylamide

POC Poly(1,8-octanediol-co-citric acid)

PTFE Polytetrafluoroethylene

PU Polyurethane

ROS Reactive oxygen species

SDF Stromal-derived factor

siRNA Small interfering RNA

SMA Smooth muscle actin

SNP Sodium nitroprusside

Sulfo-NHS N-hydroxysulfosuccinimide

SVR Surgical ventricular restoration

TCP Tissue culture polystyrene

Page 24: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

xxiv

TGF-β1 Transforming growth factor-β1

TOT Topical oxygen therapy

VEGF Vascular endothelial growth factor

α-SMA α-smooth muscle actin

Page 25: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

1

Chapter 1

1 Introduction

1.1 Overview

Tissue engineering holds the promise of restoring or regenerating functional tissues by

combining knowledge from cell biology, material science, engineering, and medicine [1]. The

focus of tissue engineering studies has evolved from replacing damaged tissues or organs with

functional tissues generated in vitro, into creating an instructive microenvironment to regenerate

the impaired tissue in situ [2]. Meanwhile, empowered by recent advances in microfabrication

technologies, a variety of functional micro-tissue constructs were generated in vitro and have

been proposed to be cogent platforms for pre-clinical drug screening studies with better

physiological relevance compared with animal models. Not surprisingly, both tissue regeneration

in vivo and micro-tissue generation in vitro can be facilitated by the biochemical and biophysical

cues in the microenvironment. In this thesis, we aimed to create instructive microenvironments to

facilitate cell-matrix interactions in tissue engineering applications with a particular focus on

cardiac tissue engineering and wound healing applications.

Native cardiac tissue has limited regeneration capacity and can be assisted by delivery of

external cells [3]. Mesenchymal stromal cells (MSCs) is the leading candidate for cell therapies

but MSCs from aged patients exhibit limited regeneration potential compared with MSCs from

young patient [4]. We propose that collagen scaffolds immobilized with angiogenic growth

factors (vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF))

can rejuvenate MSCs from aged donors and improve their cardiac regeneration potential for

surgical ventricular restoration (SVR).

Cutaneous wounds are generally capable of regeneration by themselves through a series of well-

orchestrated events [5]. However, diabetic patients suffer from chronic wounds due to disruption

of the well-coordinated cellular events involved in wound healing [6]. Impaired angiogenesis has

been the main target of current therapeutic interventions in diabetic chronic wounds yet their

clinical outcomes have been limited. A short peptide sequence derived from angiopoietin-1

(ang1), QHREDGS, has been reported to activate pro-survival pathways through integrin

Page 26: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

2

interaction in various cell types [7-13]. Therefore, it is of our interest to investigate the pro-

survival effect of QHREDGS peptide on human keratinocytes and its efficacy in promoting

diabetic wound healing in an angiogenesis-independent manner.

Cardiovascular related toxicity is one of the leading causes of marketed drug withdrawal and

current pre-clinical cellular drug screening platforms poorly recapitulate the three-dimensional

(3D) structure of native myocardium. Emerging microfabrication techniques offer the generation

of micro-tissues with precise control and greater complexity that recapitulates native tissue [14].

We proposed that perfusable cardiac micro-tissues can be generated under guidance of a

suspended template in microfabricated bioreactors and would better recapitulate the native

myocardium with a lumen in the center mimicking the capillary. This platform would be more

physiologically relevant for drug screening applications as the pharmaceutical agents are

circulated in the capillaries in vivo.

1.2 Hypothesis

The overarching hypothesis of this thesis is that topographical and biochemical cues provided by

the matrix can facilitate cell-matrix interaction and thus regulate cell assembly, cell functions,

and tissue morphogenesis. Specifically, we hypothesized that:

1. The QHREDGS peptide can be covalently immobilized onto collagen scaffold by 1-ethyl-

3-(3-dimethylaminopropyl) carbodiimide HCl (EDC) chemistry.

2. Collagen scaffolds immobilized with angiogenic growth factors (VEGF and bFGF) will

rejuvenate MSCs from aged donors and improve their regeneration potential for cardiac

remodeling.

3. The QHREDGS peptide will promote keratinocyte survival in vitro with up-regulated Akt

and MAPK phosphorylation and improve diabetic wound closure in vivo by accelerating

re-epithelialization.

4. A suspended template will guide tissue remodeling of cardiac microtissues generated in

microfabricated bioreactors and the microtissues can be perfused with a micro-tubing

template.

Page 27: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

3

1.3 Specific aims

Our overall goal is to design matrix systems that provide appropriate biophysical and/or

biochemical cues to assist tissue remodeling and regeneration in vivo or generate micro-tissues

with higher complexity that recapitulate the native tissue in vitro. The specific aims of the work

include:

1. Immobilize QHREDGS peptide or angiogenic growth factors onto collagen scaffold.

a) Covalently immobilize VEGF and bFGF onto collagen scaffold and characterize their

immobilization efficiency and release profile.

b) Covalently immobilize QHREDGS peptide onto collagen scaffold and characterize its

immobilization efficiency and release profile.

c) Characterize the physical properties of the collagen scaffold before and after growth

factor and peptide immobilization.

2. Investigate the pro-survival effect of QHREDGS peptide on human keratinocytes in vitro

and evaluate its efficacy in promoting diabetic wound healing in vivo.

a) Using normal human primary keratinocytes, investigate the effect of soluble

QHREDGS peptide on keratinocyte survival against H2O2 stress and collective

migration.

b) Using normal human primary keratinocytes, investigate the effect of immobilized

QHREDGS peptide on keratinocyte attachment, survival against H2O2 stress and

collective migration.

c) Using diabetic human primary keratinocytes, investigate the effect of immobilized

QHREDGS peptide on keratinocyte attachment, survival against H2O2 stress and

collective migration.

d) Evaluate the efficacy and the potential mechanism of immobilized QHREDGS peptide

in promoting wound healing in db/db diabetic mice.

Page 28: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

4

3. Develop perfusable cardiac micro-tissues recapitulating the 3D structure of native

myocardial fibers.

a) Design a bioreactor with suspended template to guide the self-remodeling of collagen-

based hydrogel seeded with cardiac cells.

b) Characterize the cardiac cell alignment under topographical guidance from the

template.

c) Develop a perfusable biowire system with micro-tubing in the center of the cardiac

micro-tissues.

d) Evaluate the efficacy of the perfusable biowires by perfusing nitric-oxide-releasing

reagent and investigating its effect on electrophysiological properties of the cardiac

micro-tissues.

Page 29: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

5

Chapter 2

2 Literature review1, 2, 3, 4

2.1 Instructive biomaterials for tissue engineering

2.1.1 Motivation for instructive biomaterials

Tissue engineering is defined as “an interdisciplinary field that applies the principles of

engineering and life sciences toward the development of biological substitutes that restore,

maintain, or improve tissue function.” [1] The classical tissue engineering approach to develop

biological substitutes involves incorporation of living cells into a scaffold, and cultivation of the

construct in a bioreactor. As potential alternative candidates for organ transplantations, tissue

constructs created in vitro have been used in clinical studies as biological substitutes for tissues

including skin [15], cartilage [16], bone [17], blood vessels [18, 19], heart valve [20], nerve [21],

bladder [22], trachea [23], urethra [24], and vaginal organs [25]. Moreover, modern tissue

engineering approaches have demonstrated great promises for treating and curing debilitating

health conditions including myocardial infarction [26], spinal cord injury [27, 28], osteoarthritis

[29], osteoporosis [30], diabetes [31], liver cirrhosis [32] and retinopathy [33].

1 Copyright © 2013 BioMed Central. Contents of this chapter have been published in Stem Cell Res Ther:

Thavandiran N, Nunes SS, Xiao Y, Radisic M. Topological and electrical control of cardiac differentiation and

assembly. Stem Cell Res Ther. 2013;4:14. Reuse with permission from BioMed Central. A link to the published

paper can be found at: http://www.stemcellres.com/content/4/1/14

2 Copyright © 2015 Elsevier. Contents of this chapter have been published in Methods: Xiao Y, Reis LA, Zhao Y,

Radisic M. Modifications of collagen-based biomaterials with immobilized growth factors or peptides. Methods.

2015;84:44–52. Reuse with permission from Elsevier. A link to the published paper can be found at:

http://www.sciencedirect.com/science/article/pii/S1046202315001723

3 Copyright © 2014 Cambridge University Press. Contents of this chapter have been published in: Chiu LLY, Zhang

B, Xiao Y, Radisic M. Cardiac tissue regeneration in bioreactors. Biomaterials and Regenerative Medicine,

Cambridge University Press; 2014, p. 640-668. Reuse with permission from Cambridge University Press. A link to

the published chapter can be found at:

http://ebooks.cambridge.org/chapter.jsf?bid=CBO9780511997839&cid=CBO9780511997839A046

4 Copyright © 2015 IOP Publishing. Contents of this chapter have been published in Biomedical Materials:

Davenport Huyer L, Montgomery M, Zhao Y, Xiao Y, Conant G, Korolj A, Radisic M. Biomedical Materials.

2015;10:034004. Reuse with permission from IOP Publishing. A link to the published chapter can be found at:

http://iopscience.iop.org/article/10.1088/1748-6041/10/3/034004

Page 30: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

6

Although the health benefits are obvious, the tissue engineering industry finds itself on a “roller

coaster ride” [34-39]. After its emergence and soaring development with enthusiasm from

business community in the 1990s, tissue engineering industry entered a dark period in the early

2000s, with the capital value of publicly traded tissue engineering companies reduced by 90%

between 2000 and 2002 [38]. Organogenesis (Canton, MA) and Advanced Tissue Sciences (ATS;

La Jolla, CA), two leading companies that brought the first commercially-produced tissue

engineering products to the market, engineered skin substitutes, declared bankruptcy in 2002

[40]. The dismal performance resulted in the reassessment of tissue engineering products, which

are typically limited by their long preparation time, challenging quality control, complex

distribution chains, and short shelf-life.

After the devastating years, tissue engineering industry has rebounded alongside with recent

advances in pluripotent stem cells, including embryonic stem cells (ESCs) and induced

pluripotent stem cells (iPSCs) [41]. Meanwhile, the market of tissue engineering products

expanded significantly. The capital value for publicly traded tissue engineering companies

increased over 10-fold from 2003 to 2008 with new products entering the market [39]. The sale

of products exceeded $1.3 billion in 2007, and half of this was contributed by the Medtronic

Infuse®, a recombinant bone morphogenic protein product that is acellular [42]. During this time,

the focus of tissue engineering approach has undergone considerable evolution from replacement

to regeneration in situ because it was recognized that instead of recreating the complexity of

living substitutes for transplantation [43], we should aim to develop instructive materials that

harness the body’s innate powers of self-repair [2] as the latter products may have a faster route

to the market. In these scenarios, the matrix not only serves as a scaffold that provides

mechanical support and defines the shape of tissue constructs, but also provides a multitude of

complex stimuli to support a range of cell functions and promote tissue remodeling.

Advances in biomaterials science combined with increasing knowledge of cell biology and cell-

extracellular matrix (ECM) interactions have led to the development of biomaterials tailored to

provide appropriate biological and mechanical guidance for tissue regeneration in vivo. More

importantly, the complexities in matrix design and fabrication were achieved by innovative

technologies with precise control and improved reproducibility [44]. Here, we discuss key

aspects of designing instructive biomaterials for tissue engineering applications, under the

Page 31: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

7

principle of facilitating the cell-ECM interactions with biochemical and mechanical cues

reminiscent of native ECM to support or induce tissue regeneration.

2.1.2 Naturally derived biomaterials

Biomaterials for tissue engineering applications can be categorized into naturally derived and

synthetic biomaterials with unique advantages of each group. Naturally derived biomaterials,

including polypeptides (e.g. collagen) and polysaccharides (e.g. chitosan), are found in many

products approved by the Food and Drug Administration (FDA). Their extensive use leads to

thorough characterization and small likelihood of side effects in new applications [45]. A key

advantage associated with naturally derived biomaterials is their general capacity to support cell

attachment, proliferation, and differentiation [46]. The inherent composition and structure

properties of naturally derived biomaterials enable biological recognition, including presentation

of receptor-binding ligands and susceptibility to cell-triggered matrix degradation and

remodeling [47]. On the other hand, synthetic biomaterials provide attractive alternatives with

greater control over material properties (e.g. biochemical cues, mechanical properties,

topography, structure etc.), simplified purification process, and reduced possibilities of

immunogenicity and pathogen transmission [47].

In the scope of this thesis, instructive biomaterials were designed based on naturally derived

biomaterials because of the orientation towards clinical transition. Specifically, biomaterials

based on collagen and chitosan have been approved by the FDA and widely used in clinical

tissue engineering applications such as wound dressings. It is our major interest to investigate the

modification of the biomaterials with immobilized biochemical cues such as angiogenic growth

factors and QHREDGS peptide. However, we think our results from naturally derived

biomaterials can be translated and will contribute to our general knowledge of instructive

biomaterials design including synthetic biomaterials as well.

Page 32: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

8

2.1.2.1 Collagen

Table 2-1 Principal tissue distribution and cells of origin for different collagen types in human body

(Reproduced from reference [48])

Collagen Type Principal Tissue Distribution Cells of Origin

I

Loose and dense connective tissue;

collagen fibers

Fibroblasts and reticular cells; smooth

muscle cells

Fibrocartilage

Bone Osteoblasts

Dentin Odontoblasts

II Hyaline and elastic cartilage Chondrocytes

Vitreous body of eye Retinal cells

III

Loose connective tissue; reticular fibers Fibroblasts and reticular cells

Papillary layer of dermis Smooth muscle cells; endothelial cells

Blood vessels

IV Basement membranes Epithelial and endothelial cells

Lens capsule of eye Lens fibers

V

Fetal membranes; placenta Fibroblasts

Basement membranes

Bone

Smooth muscle Smooth muscle cells

VI Connective tissue Fibroblasts

VII Epithelial basement membranes Fibroblasts

Anchoring fibrils Keratinocytes

VIII Cornea Corneal fibroblasts

IX Cartilage

X Hypertrophic cartilage

XI Cartilage

XII Papillary dermis Fibroblasts

XIV (undulin) Reticular dermis Fibroblasts

XVII P170 bullous pemphigoid antigen Keratinocytes

Collagen is the most abundant component of mammalian ECM and exists in tissues including

skin, bone, cartilage, cornea and blood vessels (Table 2-1) [48]. Native collagen has unique

Page 33: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

9

triple helix fibril structure and type I, II, III, V and XI collagen are known to form collagen fibers

[49]. Three α chains, each of which are based on the sequence -Gly-X-Y-, assemble into collagen

molecule. While 29 distinct collagen types have been characterized, type I collagen is most

widely used for tissue engineering applications [49]. Different forms of type I collagen products,

including freeze-dried sheets, pastes, pads, powder and hydrogels, have been derived using

techniques such as direct decellularization of ECM and chemical extraction in acid solutions [50],

neutral salt solutions [51], or proteolytic solutions [52]. For tissue engineering applications,

physical, chemical, and enzymatic crosslinking techniques have been developed to improve

mechanical properties and enzymatic resistance of collagen biomaterials [49]. Collagen scaffolds

have been applied as skin grafts (e.g. INTEGRA™ Matrix Wound Dressing), vascular implants

(e.g. Artegraft®), orthopaedic filings (e.g. Foundation®) and nerve guides (e.g. NeuraGen®).

Furthermore, micro-tissue analogues have been generated in vitro based on self-assembly of

collagen-based hydrogels encapsulating cells and serve as platforms for research in cell biology

and pharmaceutical development [53].

2.1.2.2 Chitosan

Figure 2-1 Chemical structure of chitosan, comprising N-acetyl-D-glucosamine (right) and D-

glucosamine (left) units.

Chitosan is a cationic polysaccharide derived from chitin, the second most abundant natural

polymer, by replacing at least 60% acetyl groups along chitin chain with amino groups (which

corresponds to a deacetylation degree of 60) (Figure 2-1) [54]. Due to these amino groups,

chitosan can be protonated in acidic solutions and become soluble. Indeed, chitosan is the only

positively charged naturally occurring polysaccharide [55]. Besides, the amino groups may form

complex with metals thus chitosan is used for waste water treatment [54]. More importantly, the

Page 34: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

10

amino groups may be quaternized or reacted with aldehyde groups under mild conditions through

reductive amination [54]. Therefore, various chitosan derivatives have been developed for a

multitude of applications in agriculture, waste treatment, food industry, cosmetics, and

biopharmaceutics [54].

For tissue engineering applications, chitosan possesses unique advantages such as antimicrobial

activity [56], mucoadhesion [57], analgesic effects [58], haemostatic properties [59], and

stronger mechanical properties compared to other naturally derived biopolymers [60]. Moreover,

chitosan can be biodegraded into non-toxic residues mainly by lysozymes [61]. Thus, chitosan

and its derivatives have been applied extensively in biomedical applications such as wound

dressings [62], drug delivery [63], bone grafts [64], and medical coatings [65] (Table 2-2).

Table 2-2 Principal properties of chitosan in relation to its use in biomedical applications

(Reproduced with permission from Elsevier, Rinaudo M. 2006 [54])

Potential biomedical applications Principle characteristics

Surgical sutures Biocompatible

Dental implants Biodegradable

Artificial skin Renewable

Rebuilding of bone Film forming

Corneal contact lenses Hydrating agent

Time release drugs for animals and human Nontoxic, biological tolerance

Encapsulating material Hydrolyzed by lysozyme

Wound healing properties

Efficient against bacteria, viruses, fungi

2.1.3 Instructive biochemical cues provided by biomaterials

The bioactivity of biomaterials is primarily conferred by the molecular information from the

basic scaffold material together with any embedded macromolecules, such as growth factors [2].

Growth factors are potent regulators that facilitate a multitude of cell activities including

migration, proliferation, differentiation, and survival. Since their mass production became

available through recombinant protein technology [66], growth factors have been extensively

applied to regulate different stages of tissue regeneration process in preclinical and clinical

studies.

Page 35: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

11

For example, basic fibroblast growth factor (bFGF) was administrated in a canine myocardial

infarct model to enhance neovascularization and improve left ventricular function [67]. Another

potent angiogenic growth factor, vascular endothelial growth factors (VEGF), was administrated

both intra-arterially and intra-muscularly in a rabbit model of chronic hindlimb ischemia and

significantly augmented revascularization [68, 69]. Platelet derived growth factor (PDGF) was

the first growth factor approved by the FDA (Regranex approved in 1997) as an adjunct to

proper ulcer care in the treatment of lower extremity diabetic neuropathic ulcers [70]. Bone

morphogenetic protein-2 (BMP-2) is another growth factor that has been approved by the FDA

for clinical applications (Infuse® bone graft) to promote interbody spinal fusion (approved in

2002) [71], tibial fracture recovery (approved in 2004) [72], and sinus augmentation (approved in

2007) [73, 74].

Peptides are short functional amino acid sequences that are derived from primary receptor-

domains of specific proteins. Different peptides have been identified in numerous primary

proteins and some of them were designed synthetically to enable novel properties such as stimuli

responsiveness [75] and self-assembly [76]. The most well-characterized peptide is the integrin-

binding Arg-Gly-Asp (RGD) sequence, which can be found in the sequence of many ECM

proteins, including fibronectin, collagen type IV, and laminin [77-79]. RGD sequence has been

extensively applied in modifications of synthetic biomaterials to provide cell-adhesion sites on

bio-inert materials [47]. Other peptide sequences have been derived from various growth factors

[80], bringing the advantage of cost-effectiveness in scalable chemical synthesis compared to

recombinant growth factors productions. While retaining comparable bioactivities, peptides are

more stable than native growth factors, which may simplify the preparation and distribution of

biomaterials [2]. Moreover, the bioactivity of peptide may be more specific compared to the

native growth factors, which are potent regulators of a multitude of cellular activities and

sometimes associated with increased risks of systematic side effects, such as carcinogenesis [81].

To overcome the short half-life and enzymatic degradation of free growth factors in solution,

controlled-release strategies are frequently adopted [2]. Dynamic release of different growth

factors with independent release profile might provide more leverage over cell behavior than

indiscriminate delivery, recapitulating the dynamic regulation in developmental pathways [82,

83]. Moreover, localized and sustained delivery of growth factors would decrease the dosage

Page 36: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

12

required while supraphysiological dosing may lead to systemic side effects such as hypotension

(VEGF) and nephrotoxicity (bFGF) [84]. Synergistic signaling of growth factors and ECM

proteins has been described in recent studies [85], which indicates necessities of sequestering

growth factors within the matrix, preventing their degradation and presenting them to cell-

surface receptors. Various growth factor immobilization methods have been used when

designing instructive biomaterials with growth factors to localize, enhance and sustain their

bioactivities [85-88].

2.1.4 Biomechanical instructions provided by biomaterials

There is growing recognition of the biomechanical cues provided by biomaterials that regulate

cell activities and tissue morphogenesis, with our expanding knowledge of mechanobiology and

emergence of technologies such as microfabrication and 3D printing. Incorporation of

topographical cues brings another dimension in biomaterials design, providing unique

opportunities in building tissue analogues involving multiple cell types, and could potentially

simplify biomaterials preparation.

Coupled by focal adhesions between the cytoskeleton and ECM, cells can pull on the matrix not

only to generate the traction forces, but also to sense the mechanical properties and structure of

the ECM, potentially through the bonding dynamics of integrin adhesion receptors [89]. The

importance of mechanical properties of ECM is first demonstrated on stem cell differentiation

with the first study showing different lineage and phenotype commitment of mesenchymal stem

cells seeded on substrates with varied rigidity [90]. Specifically, stiff substrates were myogenic,

soft substrates were neurogenic, and comparatively rigid substrates were osteogenic [90]. Recent

studies showed that stem cell fate can also be influenced by past mechanical environments,

suggesting a “mechanical memory” on stem cell differentiation [91]. Moreover, nano- and

micro-scale topographic features have been recently controlled to regulate stem cell renewal [92,

93]. Taken together, translation of biomechanical regulation from in vitro to in vivo becomes

more addressable with the wide range of emerging materials and analytical technologies and

future instructive biomaterial design may require synergistically providing both biomechanical

and biochemical factors [94].

Page 37: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

13

Cell alignment is critical for tissue-scale functionality in muscular tissues and can be regulated

by topographical guidance from substrates as well. Heart tissue possesses complex structural

organization on multiple scales. On macro-scale, native myocardium contains elongated

myofibers aligned in parallel; the structure enables coordinated contraction of the ventricle and

expulsion of blood. On micro-scale, adult cardiomyocytes are rod shaped and contain registries

of sarcomeres that enable cell contraction in response to electrical signals. On nano-scale, each

sarcomere contains precisely organized sarcomeric proteins (e.g. sarcomeric α-actin/α-actinin

and myosin heavy chain) that enable coordinated contractions of sarcomeres. Cardiomyocytes

alignment was first achieved by confining cell attachment on bio-inert substrates patterned with

cell-adhesive sites, such as fibronectin [95, 96]. Larger scale patterning aided with high-

resolution diffusion tensor magnetic resonance imaging (DTMRI) has created cardiomyocytes

monolayer that recapitulates cross-sections of native cardiac tissue [97]. Micro-and nano-scale

patterned grooves were designed to guide cardiomyocyte alignment as well and the tissue

constructs displayed anisotropic action potential propagation and contractility characteristic of

native heart tissue [98].

3D cardiac micro-tissues have been created by microfabrication technology and demonstrated 3D

alignment of heart cells more closely resemble native myocardium [99]. Constructs containing

both parallel channels and micro-pores were designed to guide multicellular organization with

cardiomyocytes predominantly occupying the parallel channels and grouping into bundles and

micro-pores remained acellular for mass transfer [100]. Other templates have been integrated to

guide the formation of 3D micro-tissues formed by hydrogel compaction driven by contractile

cells [44]. The anisotropic cell organization within 3D micro-tissues is critical for

electromechanical properties and better recapitulates the native heart tissue.

2.1.5 Mesenchymal stromal cells

In some tissues, such as the heart and brain, the native tissue has intrinsically limited

regeneration potential while the other tissues age with a declining progenitor population, thus

requiring the aid from external cell sources to regenerate [2]. The primary criteria for choosing

the external cells include easy access to autologous source, large population available after short

ex vivo expansion, capability of replenishing native cells or regenerating the native tissue. MSCs,

currently defined by their fibroblast-like morphology, adherence to plastic, expression of a

Page 38: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

14

specific set of surface antigens (CD105+, CD90+, CD73+), and capacity for osteogenic,

chondrogenic, and adipogenic lineages in vitro [101], are the leading candidate for cell therapy

and have demonstrated considerable promise in tissue regeneration in pre-clinical and clinical

studies [102].

A significant advantage of using MSCs in clinical application is that they can be readily obtained

from a variety of tissues including bone marrow [103], adipose tissue [104], placenta [105], skin

[106], umbilical cord blood [107], umbilical cord perivascular cells [108], umbilical cord

Wharton’s jelly [109], dental pulp [110], amniotic fluid [111], synovial membrane [112], and

breast milk [113]. Moreover, MSCs proliferate rapidly in vitro with up to 18-fold increase after 2

weeks of culture [114]. Therefore, a large number of MSCs may be available readily to meet the

dose requirement for clinical trials (up to millions of cells/kg body weight).

Although the exact mechanism of how MSCs exert their regenerative benefits remains to be fully

defined, several potential mechanisms have been suggested by recent studies. (1) MSCs may

transdifferentiate into specific cell type and replenish the damaged tissue, which has been

reported in clinical trials to treat bone and cartilage defects [115, 116]. (2) Fusion of MSCs with

endogenous cells may improve their regeneration capacity, which has been mainly described in

cardiac tissue [117, 118]. (3) MSCs modulate both adaptive and innate immuno systems by

suppressing T cells and modulating inflammatory cytokines expression [119, 120]. (4) The

secretome from MSCs, the spectrum of regulatory and trophic factors secreted by MSCs,

including growth factors, cytokines, and chemokines, exhibit paracrine effects that stimulate

regeneration capacity of endogenous cells [121, 122].

Clear understanding of the mechanism is important to optimize the design of MSCs clinical

studies to maximize the therapeutic benefits. For example, acellular approach would be more

cost-effective than MSCs transplantation if the therapeutic benefit of their secretome is well-

defined and could be further enhanced by targeted preconditioning and genetic manipulation

[121]. For MSCs transplantation, poor retention at target location necessitates engraftment within

delivery vehicle and various biomaterials have been used for different tissue engineering

applications [123-126]. With a growing body of literature reporting conflicting results, the

interactions between MSCs and tumor cells requires special attention for the safety concerns

related to MSCs clinical applications [127]. Also, limited regeneration potential associated with

Page 39: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

15

MSCs from aged patients compared to the ones from younger patients raises reevaluation

between autologous and allogenic cell sources for aged patients [128].

2.2 Cardiac tissue engineering

2.2.1 Motivation for cardiac tissue engineering

Myocardial infarction (MI) leads to the death of cardiomyocytes, and the infarct area becomes

replaced by a fibroblastic scar tissue that has no contractile function. This reduces the pumping

ability of the heart and the cardiac output. In addition, the scar tissue thins due to the lack of

vasculature to provide oxygen and nutrients to the infarct site, thus leading to high wall stress

and cardiac dilatation, which may ultimately lead to heart failure. Heart failure triggered by MI is

a leading cause of death globally [129].

The adult heart has a limited regenerative capacity. The shortage of donor organs further

suggests a need to develop new treatment strategies for cardiovascular diseases. Cardiac tissue

regeneration can be achieved through several strategies, including (1) gene therapy, (2) cell

transplantation, and (3) implantation or injection of biomaterials or engineered cardiac tissues.

The goal of these cardiac tissue regeneration strategies is to repair the damaged myocardium

through supporting vascularization and cell survival, in turn reducing wall thinning and

preventing dilatation and heart failure.

Since the damaged myocardium has limited capacity to regenerate, cell transplantation can

replace the damaged and lost cells, thus attenuating pathological remodeling. However, cell

transplantation is limited by the washout of cells from the injection site and the inability of

injected cells to integrate with the native tissue. Solutions include delivering cells within a

scaffold or hydrogel to support cell engraftment, and growing functional cardiac tissues that can

be implanted and would integrate with the native tissue.

2.2.2 Cell sources

Generally, cells for cardiac tissue engineering should be 1) expandable to achieve high numbers

of cells; 2) compatible with the host without causing immune reactions; and 3) able to survive

and maintain function in vitro and in vivo [130]. Cells for cardiac tissue engineering can be

Page 40: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

16

categorized into primary cardiac cells, ESCs, iPSCs, resident cardiac progenitor cells, and adult

progenitor cells from other native tissues.

2.2.2.1 Fetal and neonatal cardiac cells

Adult cardiac cells are the target cell source in cardiac regeneration since they are differentiated

cardiomyocytes with developed contractile apparatus and can integrate with host cardiomyocytes

through gap junctions and intercalated disks. However, they are present in low numbers and have

little proliferative and developmental potential. The advantages of using fetal and neonatal

cardiac cells include their greater proliferative and developmental potential than that of adult

cardiac cells. These cells also showed efficacy in cardiac regeneration, since the injection of fetal

or neonatal cardiomyocytes attenuated pathological ventricular remodeling by forming viable

grafts, increasing the ventricle thickness, and improving left ventricular function [131, 132]. By

contrast, injected adult cardiomyocytes were unable to survive both in acutely cryoinjured

myocardium and in granulation tissue. Contractile cardiac tissues have also been engineered

using fetal or neonatal rat cardiomyocyte-enriched cell populations [133-135]. However, fetal or

neonatal cardiac cells are not available in large numbers to provide the millions of

cardiomyocytes required for cardiac tissue regeneration, thus motivating the search for

alternative cell sources. Moreover, while fetal or neonatal cardiac cells from animals can readily

be used, a direct translation to human studies will not be possible due to obvious ethical

considerations involving the use of human cells.

2.2.2.2 Embryonic stem cells

ESCs have been investigated intensively due to their ability to undergo indefinite self-renewal

without losing the capacity to differentiate into all cell types. ESCs are commonly cultivated and

differentiated in 3D aggregates known as embryoid bodies (EBs). Quantification of “beating”

EBs is commonly applied to assess cardio-genicity in ESCs. However, despite robust cardiomyo-

genicity in EBs, only 1%–5% of their total cell number consists of cardiomyocytes under

standard culture conditions (i.e. plated on 0.1% gelatin-coated culture dishes and cultured in 80%

knockout Dulbecco’s Modified Eagle’s Medium with 20% fetal bovine serum, 1 mM l-glutamine,

0.1 mM mercaptoethanol, and 1% non-essential amino acid stock [136]). The most efficient and

reproducible protocols to date for differentiation of cardiomyocytes from pluripotent stem cells

Page 41: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

17

are those that have replicated the signaling pathways that regulate lineage commitment in the

early embryo. With this approach, the earliest stages of cardiovascular development in ESC

differentiation cultures were mapped, identifying a multipotent cardiovascular progenitor that

displays the capacity to generate cardiac and vascular progeny [137, 138]. In mouse Flk1 and in

humans KDR expression can be used to enrich for cardiac-specified mesoderm [139]. When

isolated from the differentiated EBs and cultured as a monolayer, these progenitors generate

contracting cardiomyocytes [137, 138]. As these progenitors differentiate, they progress through

the developmental stages thought to be involved in the establishment of the cardiovascular

lineages in vivo, for which specific cytokines are required. The combination of activin A and

BMP4 on days 1–4 of EB differentiation induces a primitive-streak-like population and

mesoderm development. Subsequent application of WNT inhibitor DKK1 and KDR ligand

VEGF165 significantly enhances the differentiation of KDR+ progenitors into cardiomyocytes

[137, 138], while bFGF is added to support continued expansion of cardiovascular lineages.

ESC-derived cardiomyocytes have been used to engineer cardiac tissues. Guo et al. [140] seeded

mouse ESC-derived cardiomyocytes into circular molds with collagen type I and Matrigel to

produce engineered heart tissues. Stevens et al. [141, 142] tricultured human ESC-derived

cardiomyocytes with mouse embryonic fibroblasts and ESC-derived endothelial cells or human

umbilical vein endothelial cells to generate scaffold-free cardiac patches.

2.2.2.3 Induced pluripotent stem cells

The recent advent of iPSCs has been considered as a solution for the ethical issues and

immunological concerns associated with the usage of ESCs. Briefly, fully differentiated cells can

be reprogrammed to have pluripotency like ESCs by introducing a set of transcription factors

including Oct3/4, Sox2, c-Myc, and Klf4 [143]. Although the efficiency of current protocols for

derivation of iPSCs is still low, iPSCs can serve as a novel cell source for cardiac tissue

engineering. Previously, Mauritz et al. [144] generated functional cardiomyocytes from mouse

iPSCs. Importantly, human iPSCs, which were derived from reprogramming of adult fibroblasts,

were differentiated into cardiomyocytes [145, 146]. The derivation of cardiomyocytes from

iPSCs allows the generation of autologous human cardiomyocytes necessary for cardiac tissue

regeneration [146]. Human embryonic stem cell and human iPSC-derived cardiomyocytes were

previously cultured in collagen matrices using uniaxial mechanical stress conditioning and the

Page 42: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

18

addition of endothelial cells and stromal supporting cells [147]. The engineered human

myocardium was transplanted onto hearts of athymic rats. The grafts resembled host

myocardium and contained microvessels that were perfused by the host coronary circulation.

Fibroblasts, with their demonstrated ability to be transdifferentiated into skeletal muscle cells

[148], have been considered as an attractive cell source for cardiac tissue engineering. Efe et al.

[149] reprogrammed mouse embryonic fibroblasts (MEFs) directly into cardiomyocytes through

over-expression of Oct4, Sox2, Klf4 and c-Myc. Reprogramming fibroblasts into cardiomyocytes

is a very appealing approach since “on-site” therapy would be possible if the reprogramming

could be done directly in the infarct area.

However, it should also be noted that clinical applications of iPSCs are still under development

and are currently challenged by the possibility of tumor development after transplantation [150,

151]. Specifically, carcinogenesis can be caused by the integrated oncogenes used in

reprogramming, c-Myc in particular [152], by insertional mutagenesis from viral vectors [153],

by disruption of tumor suppressor genes [154], and by genetic and epigenetic abnormalities in

the reprogrammed cells [155].

2.2.2.4 Cardiac progenitor cells

Recent studies have revealed evidence for DNA synthesis and an increase in the number of

cardiomyocytes in diseased human hearts [156, 157]. Although it occurs at a very slow rate, the

regeneration of cardiomyocytes in the heart seems to be an appealing solution for myocardial

infarction. Endogenous regeneration of the myocardium may be due to cardiomyocytes re-

entering the cell cycle and dividing, or the proliferation and differentiation of resident

populations of cardiac progenitor cells (CPCs) [158]. Genetic manipulation or application of

bioactive molecules can be used to alter the cell cycle control in adult cardiomyocytes in order to

promote cell proliferation.

Urbanek et al. [159] reported that the increase in cardiac mass after human aortic stenosis is a

result of combined myocyte hypertrophy and hyperplasia. The number of cells expressing stem

cell markers and telomerase was increased in aortic stenosis. The cell clusters containing these

stem cells made the transition to cardiogenic and myocyte precursors. There was also evidence of

primitive myocytes turning into terminally differentiated myocytes. In a separate study, Urbanek

Page 43: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

19

et al. [160] found a cardiac stem cell pool in the human heart that is capable of promoting

myocardial regeneration after infarction. The number of cardiac stem cells increased both in

acute and in chronic infarcts. These cells were telomerase-competent dividing stem cells that

were committed to the myocyte, smooth muscle, and endothelial cell lineages. This suggests the

activation of cardiac stem cells in response to ischemic injury. However, chronic infarcts had

fewer functionally competent cardiac stem cells in the viable myocardium than were present in

acute cases, as indicated by the higher expression of markers of cellular senescence (e.g. p16 and

p53), shorter telomeres, and greater apoptosis in cardiac stem cells of chronic infarcts. This

underlies the progressive cardiac deterioration that leads to terminal cardiac failure in chronic

infarcts. It was later reported that the myocardium has interstitial structures with stem cell niches

that contain resident cardiac stem cells and lineage-committed cells [161]. These cells are

connected through gap and adherens junctions to myocytes and fibroblasts, which act as

supporting cells. The cardiac stem cells divide both symmetrically and asymmetric- ally, with a

dominance of asymmetric division in which the cells give rise to one daughter cardiac stem cell

and one daughter committed cell. This preserves primitive cells while generating myocyte

progenitors, endothelial cells, and smooth muscle cells.

Laugwitz et al. [162] reported the identification of isl1+ cardiac progenitors in post-natal rat,

mouse, and human myocardium. When co-cultured with neonatal myocytes, these isl1+ cells

underwent highly efficient conversion into the mature cardiac phenotype. Subsequent studies

have identified CPCs in post-natal hearts with the expression of surface markers such as c-kit and

Sca-1, and physiological properties such as the ability to efflux fluorescent dye and to form

multicellular spheroids [163, 164]. However, these cell subpopulations are insufficient for

cardiac repair without appropriate signaling. Bioactive molecules and biomaterials can be used to

recruit these cells to the infarct region for in situ cardiac repair. In addition, these cells can be

isolated and then expanded in vitro for further use in cardiac tissue regeneration.

Different resident stem cell populations have been used for cardiac repair. The injection of lin−,

c-kit+ cells into the ischemic heart improved the regeneration of the myocardium [165]. In

addition, cardiomyocytes were derived from the isl1+ cardiac progenitor population [166]. These

cells were then used to generate beating thin films [159]. Adult cardiac progenitors can also be

isolated from explant cultures of human endomyocardial biopsies, and can be expanded as

Page 44: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

20

cardiospheres in vitro [167, 168]. These cardiosphere-derived cells improved cardiac

regeneration in mice through direct differentiation and the secretion of angiogenic growth factor

to improve cell survival [169]. However, it remains a challenge to generate high numbers of

cardiac progenitor cells for cardiac tissue engineering.

2.2.2.5 Bone marrow cells

Bone-marrow-derived stem cells have elicited considerable interest in cardiac tissue engineering

because there were reports of hematopoietic stem cells regenerating into cardiomyocytes [170].

Adult bone marrow cells have been used to regenerate and improve the functional properties of

the myocardium [171-174]. Marrow-derived stromal cells, also referred to as mesenchymal stem

cells, have been applied in clinical trials, and this resulted in a significant improvement in

ejection fraction [175]. However, subsequent studies showed that the improvement brought about

by bone marrow cells was more likely due to their secretion of soluble factors and to their

vasculogenic potential after transplantation rather than their differentiation into cardiomyocytes.

2.2.2.6 Adipose-derived cells

Besides bone marrow cells and cardiac progenitor cells, the adipose tissue is another source of

autologous adult stem cells. A significantly higher density of MSCs is present in the adipose

tissue than in the bone marrow (5% compared with 0.01%) [176]. Adipose-derived cells have

been shown to improve cardiac tissue regeneration by direct differentiation and secretion of

paracrine factors [177-179].

2.2.3 Biomaterials

The cells described above hold great promise in cardiac regeneration yet face the challenge from

poor engraftment at the target site. Delivery within a supporting matrix can improve the

engraftment of cells by preventing cell loss during delivery, and providing a protective

environment afterwards. Moreover, the matrix provides mechanical support to alleviate the

increased wall stress caused by functional cardiomyocytes loss. The biomaterials applied can be

categorized into porous scaffolds and hydrogels and each of them have advantages over the other.

Porous scaffolds provide stronger mechanical support during cell delivery and thereafter while

hydrogel systems can be delivered in a minimally invasive manner.

Page 45: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

21

Many natural and synthetic biomaterials have been fabricated into scaffolds for cardiac tissue

engineering by techniques such as freeze-drying, salt casting, and electro-spinning. Scaffolds can

be made of naturally derived biomaterials such as collagen [180, 181], gelatin [135], and alginate

[133]. Synthetic biomaterials have also been extensively investigated such as polyglycolic acid

(PGA) [182], polycaprolactone (PCL) [183], poly(L-lactide) (PLLA) [134, 184], polyurethane

(PU) [185], poly(1,8-octanediol-co-citric acid) (POC) [186], and poly(glycerol sebacate) (PGS)

[99]. More recently, decellularization technique provides an attractive method of generating

naturally derived scaffolds (mainly composed of collagen) with proper cardiac architecture [187].

Different hydrogel systems have been developed to deliver cell therapies for cardiac regeneration.

Naturally derived hydrogels have been investigated such as collagen [188, 189], fibrin [190, 191],

chitosan [192, 193], alginate [194, 195], and Matrigel [196, 197]. Hydrogels based on synthetic

polymers including poly(ethylene glycol) (PEG) [198], poly-N-isopropylacrylamide (PNIPAAm)

[199, 200], and poly(2-hydroxyethyl methacrylate) (pHEMA) [199, 201] have been developed to

deliver cells for cardiac regeneration as well.

In cardiac tissue engineering, biomaterials serve predominately as scaffolds for tissue formation

and vehicles for the delivery of engineered tissues [202-207]. Scaffolds for cardiac tissue

engineering require a number of criteria to be carefully considered to allow for optimal tissue

function including: physical properties of the polymer (e.g. strength and elasticity), degradation

rates, and host response [204]. Furthermore, these properties help to dictate the body's elicited

immune response.

To satisfy the dynamic nature of heart function and myocardial remodelling post-MI, the ideal

cardiac biomaterial should account for several design parameters. Matching the mechanical

properties of the myocardium is an important cardiac biomaterial property [208]. The Young's

modulus of the adult human myocardium ranges non-linearly from 10–20 kPa (start of diastole)

to 200–500 kPa (end of diastole) [209-212]. A rigid and inelastic patch placed on the heart will

impede contraction. A scaffold should not be constructed too soft as pathological cardiac dilation

can be reduced by mechanically reinforcing the myocardium [213]. In addition, materials

capable of achieving tissue-like compliance (e.g. hydrogels) must allow for easy

handling/suturing. An ideal biomaterial should also comply with the changes in strain

experienced by the myocardium of approximately ±15% [214-216]. The anisotropic

Page 46: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

22

(directionally dependent) stiffness of the heart tissue [216-220] is an important design parameter

that the scaffold should replicate. Mathematical modelling and in vivo experiments on dogs

demonstrated that the patches with anisotropic mechanical properties (Dacron with slits), placed

onto an infarcted myocardium resulted in improved functionality compared to isotropic patches

[221, 222]. The cyclic contraction of the myocardium necessitates a biomaterial that is

elastomeric. The patch should biodegrade over the desired period, matching the remodelling

process of the heart post MI, that usually takes 6–8 weeks, to avoid fibrous capsule formation

and a chronic inflammatory response in the myocardium. Ensuring that the material degrades at

the same rate as the heart heals is crucial for success [223].

Comprehending the immune response to an implanted biomaterial is required to determine the

clinical relevance. The immediate response of the host to implantation of a foreign material is

known as the tissue response [224-227]. Upon implantation, host proteins adhere to the

implanted biomaterial surface. These proteins signal monocytes and other leukocytes to begin the

foreign body cascade [227-229]. Macrophage recruitment and differentiation dictates the

formation of foreign body giant cells and fibrous capsule formation [227-229]. The way a

biomaterial influences macrophage polarization has been found to influence the host immune

response to implanted devices significantly. Specifically, M1 macrophages induce an

inflammatory response in the body, while M2 macrophages encourage tissue repair [227-229].

Material design looks to minimize the inflammatory and immune response as well as the fibrous

capsule formation. Formation of a fibrous capsule inhibits the efficacy of a tissue scaffold, as

cells are not able to integrate and interact with the surrounding cells, and the apparent stiffness of

the material is increased [230]. Furthermore, this limits vascularization, which is important as it

facilitates growth through delivery of blood, as well as ensures the viability of exogenously

applied cells [231]. Many designs of cardiac biomaterials aim to encourage the polarization of

macrophages to the M2 phenotype, in order to minimize fibrous capsule formation and promote

incorporation into host tissue [227, 229, 232]. Minimization of fibrous capsule formation, as well

as reduction in the foreign body reaction and chronic inflammation, are considered benchmarks

to biocompatibility assessment [203, 226, 233, 234].

Page 47: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

23

2.3 Tissue engineering for wound healing

2.3.1 Wound healing process

Wound healing is a complex process involving a highly regulated cascade of biochemical and

cellular events coordinated to achieve restoration of tissue integrity following tissue damage or

loss [5, 235]. The most extensively studied wound healing process is cutaneous wound healing as

skin is the largest organ of the human body and the outermost barrier. The skin is composed of

two main layers, the actual barrier, epidermis, and the underneath supporting layer, dermis. The

epidermis is mainly composed of hard, cornified keratinocytes, which maintain skin integrity and

serve as the physical barrier against external environment. The dermis is the residence of various

cell types, including dermal fibroblasts, macrophages, lymphocytes, and mast cells. The cells

within dermis, together with the fibrous and amorphous ECM between them, provide tensile

strength, support, and moisture retention to the skin. Moreover, the dermis is where blood vessels

are located, which provides oxygen and blood to the skin.

Clinically, cutaneous wound healing process can be characterized as repair by first, second and

third intention [236]. Primary intention healing occurs when tissue is incised in a clean manner

and re-approximated (i.e. after surgical procedures) and generally results in less scarring because

the incisional defect re-epithelizes rapidly and is sealed by matrix deposition. Secondary

intention healing occurs in open wounds where the wound edges are not approximated and the

defect heals by formation of granulation tissue, wound contraction and migration of epithelial

cells. Third intention healing (also called delayed primary intention healing) occurs when a

wound is allowed to heal open for a few days (usually due to gross contamination), and then

closed as in primary healing.

Cutaneous wound healing consists of a symphony of well-orchestrated events that can be divided

into three overlapping phases: inflammation, proliferation and remodeling. Right after injury, a

fibrin clot (a meshwork of mainly fibrin with platelets embedded) forms to stop bleeding

(hemostasis) and protect the wound. Meanwhile, platelets release a wide range of growth factors

such as PDGF to recruit inflammatory cells, among which neutrophils are the first cell type

coming from the bloodstream to the wound. At this time, the wound is hypoxic due to the

damage of the blood vessels immediately after injury. This seemingly deleterious situation

Page 48: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

24

actually has beneficial effects as it increases keratinocytes migration, early angiogenesis,

fibroblasts proliferation, and the transcription and synthesis of crucial growth factors and

cytokines, including PDGF, VEGF, and transforming growth factor-β1 (TGF-β1) (Figure 2-2a).

Page 49: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

25

Figure 2-2 Three classic stages of wound healing. (a) Inflammation. The wound is characterized by a

hypoxic (ischemic) environment in which a fibrin clot has formed. Bacteria, neutrophils and platelets are

abundant in the wound. (b) New tissue formation. An eschar (scab) has formed on the surface of the

wound. Most cells from the previous stage of repair have migrated from the wound, and new blood

vessels form (angiogenesis). The migration of keratinocytes can be observed under the eschar. (c)

Remodelling. Disorganized collagen has been laid down by fibroblasts in the wound. Re-epithelialization

and contraction contributed to the final closure of the wound. (Reproduced with permission from Nature

Publishing Group, Gurtner G. et al. 2008 [5])

The second phase is proliferation and is marked by the appearance of granulation tissue, a robust,

dark pink, granular-appearing tissue that consists of fibroblasts, macrophages, newly forming

ECM, and developing blood vessels. Fibroblasts are recruited to the wound by PDGF (secreted

by platelets and macrophages) and deposit new matrix, including collagen, elastin, reticulin,

proteoglycans, and glycosaminoglycans (GAGs). Monocytes from circulation take up residence

at the wound site as tissue macrophages, which serve a dual role in wound healing process:

between inflammatory and proliferation phase, macrophages shift from a proinflammatory

secreting phagocyte that clears debris to an anti-inflammatory initiator of repair. Macrophages

also serve a dual role in angiogenesis: they first respond to hypoxia by promoting angiogenesis,

and later, they respond to interferons by inhibiting angiogenesis. Some fibroblasts differentiate

into a tension-generating phenotype known as myofibroblasts induced by TGFβ1.

Myofibroblasts express α-smooth muscle actin (α-SMA) and participate in wound contraction

through their extensive cell-matrix contacts (supermature focal adhesions) which are linked to

their intracellular stress fibers. Keratinocyte migration is mainly mediated by keratinocyte

growth factors (KGF-1 and KGF-2), which are produced by fibroblasts. Importantly,

keratinocytes do not bind to the fibrin clot, but to the dermal matrix, therefore providing an

efficient debridement of eschar from the dermal layer while re-epithelializing. Coverage by

keratinocytes over the wound site marks the final step of the repair phase as these cornified

epithelial cells provide the outermost cellular barrier to the environment. (Figure 2-2b)

The last phase of wound healing is remodeling, which mainly involves collagen maturation and

reorganization. Over time, the immature dermal matrix is reorganized to a stronger, more mature

scar mainly due to realignment of the collagen fibers from a random orientation to specific

structure according to function. The remodeling is usually accompanied by regression of blood

vessels and degradation of excess ECM synthesized at earlier stages. (Figure 2-2c)

Page 50: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

26

2.3.2 Diabetic wound healing

Optimum cutaneous wound healing process requires a well-orchestrated integration of the

complex biological and molecular events of cell migration and proliferation, and of ECM

deposition and remodelling. This complicated process can be described as several overlapping

phases: inflammation, new tissue formation (angiogenesis and re-epithelialization), and

remodeling. The transition between phases is mediated by appropriate and precise regulation

from inflammatory mediators, growth factors, cytokines, and mechanical forces [5, 235].

However, this orderly healing process can be impaired by systemic complications such as

diabetes mellitus, and stalled in a specific phase resulting in chronic wounds that fail to heal [6].

Chronic wounds are pervasive worldwide, affecting more than 70 million people, with senior

population especially vulnerable [237]. Many face harsh medical and social consequences.

Specifically, diabetic foot ulcer affects 15% of people with diabetes and is the leading cause of

nontraumatic amputation [238].

Denervation of local sympathetic nerve system is a feature of diabetic neuropathy and results in

sensory deficits, so that the diabetic patients do not respond to external insults including pressure

and heat [239]. Absence of protective symptoms against the persistent insults leads to further

deformities and increased infection rates [6]. Meanwhile, diabetic wound healing is challenged

by vasculopathy including impairments in both vasculogenesis and angiogenesis. Vasculogenesis

is impaired in diabetic wounds due to insufficient mobilization of circulating endothelial

progenitor cells (EPCs) from the bone marrow [240] and impaired homing to the wound site with

diminished expression of the chemokine stromal derived factor-1α (SDF-1α) [241]. Impaired

endothelium function also involves a reduction of nitric oxide synthesis and faster nitric oxide

degradation due to the presence of excess reactive oxygen species (ROS) [242]. Moreover,

excessive glycosylation of matrix proteins induced by hyperglycemia leads to crosslinks between

molecules in the basement membrane of ECM and impairs angiogenesis [243]. Excessive

activation of some matrix metalloproteinases (MMPs) such as MMP9 can impair cell migration

and lead to breakdown of some critical matrix proteins and growth factors [244].

On the cellular level, the resident cells in diabetic chronic wounds have been reported to be

associated with abnormalities compared to those from normal wounds. Fibroblasts isolated from

diabetic chronic ulcers are senescent with decreased responses to various growth factors

Page 51: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

27

including transforming growth factor-β1 (TGF-β1), platelet derived growth factor (PDGF),

epidermal growth factor (EGF), insulin-like growth factor-1 (IGF-1), basic fibroblast growth

factor (bFGF) [245-248]. Macrophages in diabetic wounds show a decrease in release of

cytokines, including tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), and vascular

endothelial growth factor (VEGF) [249]. Keratinocytes attachment and migration, the critical

events in re-epithelialization, are impaired in diabetic chronic wounds due to altered ECM

composition and an enhanced ECM degradation rate [250, 251]. Meanwhile, all the resident cells

in the wound site are stressed by excessive production of reactive oxygen species (ROS) from

macrophages and neutrophils due to consistent inflammation, coupled with an impaired

antioxidant defense capability in response to hyperglycemia [252-254].

2.3.3 Current tissue engineering products for topical wounds

Wound dressing was the first category of FDA-approved tissue engineering products and has

seen numerous innovations since then. In general, wound dressings can be categorized into

acellular dressing and dressings with cells. Based on how acellular dressings interact with the

native wound environment, they can be further grouped into bio-inert dressings and bioactive

dressings.

Bio-inert dressings mainly serve as a physical barrier and keep the wound environment moist.

Winter et al. first described that moisture-retaining dressings speed epithelialization of acute,

superficial wounds in pigs compared with air-exposed wound [255] and similar results were

observed in humans later [256]. Moreover, some conformable bio-inert dressings can absorb

exudate in the draining wounds [257].

A group of bioactive acellular wound dressings were developed to deliver bioactive agents to the

local wound environment. Antibiotic drugs have been delivered in wound dressings that include

streptomycin [258], minocycline [259], vancomycin [260], neomycin [261] and ciprofloxacin

[262, 263]. Antiseptic agents have been extensively applied such as iodine-releasing agents (e.g.

Hyiodine®), silver-releasing agents (e.g. SilverSeal®), iodine (e.g. Iodozyme™), chlorhexidine

(e.g. BACTIGRAS◊), and nitric oxide [264]. Having been applied in cutaneous wound treatment

for decades [265], natural honey received a lot of attention recently as the underlying

Page 52: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

28

mechanisms of honey-promoted wound healing became better understood, which mainly

involves antimicrobial and anti-inflammatory effects [266].

Another important function of bioactive wound dressing is providing the matrix for wound

regeneration, which is critical for angiogenesis and re-epithelialization. Type I collagen is used

most often for this purpose due to its close similarity to the native ECM [267]. Scaffolds based

on chitosan [62], cellulose [268], fibrin [269], gelatin [270, 271], silk [270], and alginate [272]

have been investigated as well. More recently, a bioelectric dressing that generates physiologic

levels of micro-current (2-10 μA) (Procellera®) has been reported to accelerate wound healing by

promoting re-epithelialization [273].

Living skin substitutes consist of bioactive dressing hosting dermal cells and/or epidermal cells.

With current techniques, epidermal grafts capable of covering the entire surface area of the body

can be generated from a 3-cm2 biopsy from autologous tissue [274]. However, these living skin

substitutes are still limited by their cost in terms of time and money needed for preparation, short

shelf-life and difficulties in storage [15]. More recently, MSCs from various sources have also

been reported to promote both normal and diabetic wound healing and many clinical trials are

under way [275].

2.3.4 Instructive biochemical cues for wound healing

The importance of biochemical cues on cellular behavior and tissue morphogenesis has been well

recognized and utilized in tissue engineering applications. When designing biomaterials,

biochemical property is often the first to consider as the bioactivity of biomaterials is primarily

conferred by the molecular information from the basic scaffold material together with any

embedded macromolecules, such as growth factors [2]. For wound healing applications, growth

factors and derivatives, bioactive matrix materials (e.g. collagen and chitosan), small bioactive

molecules (e.g. oxygen and nitric oxide), and genetic regulators (e.g. cDNA, siRNA, and miRNA)

have been implemented in instructive matrix designed for regulating wound healing (Figure 2-3).

Page 53: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

29

Figure 2-3 Different biochemical cues provided by matrix to regulate the native cells. Growth factors

and derivatives can interact with native cells through their specific receptors. The composition of ECM

proteins are usually recognized by integrins. Small bioactive molecules including oxygen and nitric oxide

can diffuse into the cells and mainly affect mitochondria activities. Genetic regulators including

complimentary DNA (cDNA), small interfering RNA (siRNA) and micro RNA (miRNA) can be

delivered by non-viral vehicles and facilitate gene transcription and translation.

2.3.4.1 Growth factors and derivatives

Various growth factors have been delivered within wound dressings, including epidermal growth

factor (EGF) [276], VEGF [277], bFGF [278], transforming growth factor-β (TGF-β) [279] and

the FDA-approved PDGF [70]. Conventional delivery methods often apply the growth factors as

soluble form which result in burst release that can be affiliated with severe systematic side

effects such as hypotension (VEGF) and nephrotoxicity (bFGF) [84]. Growth factor variants

have been developed to render desirable properties including solubility [280], improved retention

[281], ECM affinity [85]. Moreover, short peptide sequences have been developed to recapitulate

the bioactivity of target growth factors by chemical synthesis with better cost-effectiveness.

Recently, novel delivery strategies other than providing growth factors as soluble supplements

have been explored. Synergistic signaling between growth factor receptors and integrin ligands

Page 54: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

30

has been proposed, which motivates immobilizing the growth factors in close affinity to ECM

proteins [282]. Hubbell et al. demonstrated that the growth factors delivered by this method can

be effective at lower dosage, which is important for both eliminating potential side effects by

supraphysiological dosage and improving cost effectiveness of growth factors therapies [282].

2.3.4.2 Small bioactive molecules

2.3.4.2.1 Oxygen

It is with no doubt that among all the chemicals, oxygen is the most vital to the human body due

to its involvement in cellular respiration and energy production. Upon injury, the wound site

immediately suffers from ischemia due to the disruption of vasculature and the tissue oxygen

tension in chronic wounds has been transcutaneously measured to be lower than normal tissues

[283]. Historically, hyperbaric oxygen therapies (HBOT) have been applied exposing the body

intermittently to pure oxygen under pressure in a stationary pressure chamber [284]. HBOT

delivers oxygen through systematic circulation therefore its efficacy is limited in tissues with

poor circulation. Since the early 1960s, topical oxygen therapy (TOT) has been developed, which

typically involves applying pure oxygen with sealing around wound tissue for a mean of 90 min,

once a day at an absolute pressure slightly above atmospheric pressure [285]. More recently,

Oxygen-releasing wound dressings (e.g. Oxyzyme™) have been developed to promote wound

healing by addressing cellular hypoxia after tissue damage [284]. Instead of attaching a bag filled

with pure oxygen to the wound, Oxyzyme™ system generates oxygen by chemical reaction.

The importance of oxygen in wound healing is well documented as increased energy is

demanded in the granulation tissue for cellular activities throughout wound healing including

bacterial defense, cell proliferation, and cell migration. Once recruited to the wound site, the

bactericidal activity of leukocytes is positively correlated to local oxygen concentration [286].

HBOT has been recently shown to reverse the diabetic defect in endothelial progenitor cell

mobilization from bone marrow, which is critical for angiogenesis [287]. During remodeling

phase, fibroblast proliferation and collagen synthesis are oxygen dependent [288, 289]. Other

than directly supplying energy for cellular metabolism, oxygen has been found critical for

growth factor signaling as well [290].

Page 55: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

31

The metabolically more active cells in the wound consume large amount of oxygen and this,

together with interrupted blood supply, contributes generation of a hypoxia gradient in the

wound tissue. Recent discovery of hypoxia induced factor α (HIF 1-α) highlighted the

importance of hypoxia gradient in wound healing process [291]. This hypoxia gradient is

important and sometimes prerequisite for effective wound healing and stabilized HIF 1-α

expression is critical to improved diabetic wound healing [292]. However, it should be noted that

the hypoxia gradient only benefits wound healing process with transient presence while chronic

hypoxia would result in wounds that fail to heal.

2.3.4.2.2 Nitric oxide

Ever since establishing its important role in physiological regulation as the endothelial relaxing

factor [293], nitric oxide (NO) has been intensively studied and its extensive physiological

impact has been revealed in virtually all organ and tissue systems under both normal and

pathological conditions [294]. Specifically, NO has been reported to be involved throughout the

three phases of cutaneous wound healing and function as an important diffusible, gaseous

regulator of angiogenesis, inflammatory response, and collagen deposition [295]. Over the past

two decades, many NO delivery devices and vehicles have been developed to transit the

therapeutic potential of NO to improve cutaneous wound healing.

NO is generated by the enzyme nitric oxide synthase (NOS), which catalyzes the conversion of

amino acid L-arginine to L-citrulline. Three NOS isoforms have been identified: two

constitutively expressed isoforms (endothelial NOS (eNOS) and neuronal NOS (nNOS)) and one

inducible isoform (iNOS) [296-298]. The expression of iNOS is induced by a variety of

cytokines, growth factors, and inflammatory stimuli on target cells which lead to high expression

levels and NO output compared with eNOS and nNOS [299]. All three NOS isoforms have been

found in skin tissue: nNOS has been observed in keratinocytes and melanocytes [300, 301];

eNOS have be detected in keratinocytes of the basal epidermal layer, dermal fibroblasts,

endothelial capillaries, and eccrine glands [302, 303]; and iNOS have be induced in keratinocytes

[304, 305], dermal fibroblasts [302], and endothelial cells [306].

The implication of NO in wound healing process has received a lot of attention since studies

showing correlation between cutaneous wound healing process and increased levels of NO

Page 56: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

32

metabolites (e.g. nitrite and nitrate) [307], mRNA and protein expression of all three NOS

isoforms [308-310]. Moreover, inhibition of NOS by competitive inhibitors decreases collagen

deposition and breaking strength of wounds and impairs healing [307]. The critical role of NO in

wound healing is further proved by both eNOS and iNOS knockout mice, which showed serious

deficit in cutaneous wound healing [311, 312]. Importantly, there are also strong correlations

between impaired wound healing in diabetic animals and decreased NOS expression and activity,

and/or NO levels [309, 313, 314].

It is evident that NO participates throughout the three phases of wound healing. First of all, NO

has been applied as an antimicrobial agent [264]. During the early inflammatory phase, NO

regulates infiltration of monocytes and neutrophils by activating pro-inflammatory cytokines (e.g.

IL-8 and TGF-β1) and also serving as chemoattractant by itself [315-317]. NO is involved in

proliferative phase as it has been reported to promote angiogenesis and keratinocytes migration.

Importantly, NO is vital to the activity of VEGF as blockade of NOS prevents VEGF-induced

endothelial cell proliferation and mitogen-activated protein (MAP) kinase [318]. NO also

mediates angiogenesis directly by promoting ECs proliferation and migration [319]. Moreover

NO has been found to promote proliferation of keratinocytes and inhibits their apoptosis [320,

321]. At the final phase of cutaneous wound healing, NO predominantly regulates collagen

synthesis in fibroblasts. Treatments with NO donors, dietary L-arginine, or iNOS overexpression

all enhance collagen deposition in the wound [322-324].

The wide-ranging functionalities of NO in the wound healing process motivated numerous

attempts to implement NO as therapeutic agent to improved wound healing outcomes. However,

as a gas molecule NO is extremely difficult to handle owing to its instability and potential to be

oxidized into toxic nitrogen dioxide molecule. Therefore, NO delivery devices vehicles including

S-nitrosothiols [325, 326], diazeniumdiolates (NONOates) [327, 328], and nano particles[324,

329, 330] have been designed and demonstrated improved wound healing outcomes.

2.3.4.3 Genetic regulators

Genetic therapies have been developed originally using viral vectors and successfully applied in

promoting wound healing [331]. The general principle is to deliver nucleic acids that are

developed either for blocking harmful genes or for restoring the activity of defective genes.

Page 57: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

33

However, the use of viral vectors in humans faces safety challenges such as immune reactions.

Therefore genetic regulators including naked DNA [332] and RNA interference [333] delivered

by non-viral vectors have been developed. Efficient delivery and targeted release are the main

challenges for non-viral based genetic therapies and recent advances such as electroporation have

significantly improved the efficiency [334]. Here we review recent advances in developing

genetic regulators delivered by non-viral vectors with a specific focus on wound healing

applications.

2.3.4.3.1 Complementary DNA (cDNA)

Complementary DNA (cDNA) is a DNA copy synthesized from the target messenger RNA

(mRNA) that encodes specific protein via reverse transcription. Non-viral vectors including

cationic polymers, cationic liposomes, and naked plasmids have been designed to deliver cDNAs

encoding for peptides (e.g. LL-37 [335], secretoneurin [336]) and growth factors (e.g. VEGF

[337], KGF [338]) to regulate different wound healing phases and promote skin regeneration.

Sonic hedgehog is a prototypical morphogen that plays essential roles during embryonic

development and it has been shown to regulate postnatal tissue remodeling and regeneration by

promoting angiogenesis [339]. Asai et al. demonstrated that topical sonic hedgehog gene therapy

delivered by plasmid vector accelerated wound healing in type 2 diabetic mice by promoting

microvascular remodeling [340]. Park et al. delivered sonic hedgehog DNA intradermally as

polyplexes formed with biodegradable cationic poly(β-amino esters) (PBAE) and reported

similar angiogenic benefits and improved wound healing [341].

The safety of delivering cDNA using naked plasmids has been demonstrated in a number of

recent clinical studies using intramuscular injections of hepatocyte growth factor (HGF) DNA

plasmids to treat critical limb ischemia [342-345]. Importantly, local delivery of HGF plasmids

did not cause peripheral edema and did not increase systemic HGF protein level [343]. Moreover,

therapeutic benefits including reduced pain, increased ankle-brachial index and improved wound

healing have been demonstrated in these studies and some ischemic ulcers healed completely

[342-345].

cDNA transfection usually results in transient expression of exogenous genes and methods have

been developed to prolong the gene expression. Kulkarni et al. encapsulated lipoplexes carrying

Page 58: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

34

eNOS encoding gene in fibrin microspheres and formed a “fibrin in fibrin” temporal release

system [346]. Electrospun fibers have been immobilized with plasmids to deliver growth factors

such as bFGF [347] and EGF [348].

2.3.4.3.2 Small interfering RNA (siRNA)

While cDNA transfection enables production of a functional protein, small interfering RNAs

(siRNAs) can be delivered to silence deleterious genes by complimentary binding to the mRNA

sequences of the corresponding target genes. Therefore, cDNA and siRNA can serve as controls

for each other in recovery experiments designed for specific pathways [349, 350]. More

importantly, siRNA-based post-transcriptional modification methods have been applied in

wound healing applications to achieve transient local functional ablation of malignant genes at

different phases of wound healing including angiogenesis and ECM remodeling. Overview of

siRNA-based therapies for other diseases such as cancer can be found in other excellent review

papers [351, 352].

Chen et al. identified INT6/eIF3e as regulator of hypoxia-inducible factor 2α (HIF2α) activity

and they reported that a single siRNA-Int6 application promoted neoangiogenesis by

accumulating HIF2α and downstream transcription of angiogenic factors in a hypoxia-

independent manner [353]. Using a previously developed agarose matrix topical delivery system,

Nguyen et al. achieved near-complete local knockdown of p53, a cell cycle regulator, and

accelerated wound healing in diabetic mice with increased vasculogenic cytokines including

VEGF and SDF-1 [354]. Wetterau et al. delivered prolyl hydroxylase domain 2 (PHD2) siRNA

with agarose matrix in diabetic mice and suppression of PHD2 increased the expression of

HIF1α and subsequent angiogenic regulators [355].

Remodeling phase is another therapeutic target of wound healing as dysregulated remodeling

could result in hypertrophic scars that are unaesthetic. Lee et al. first delivered Smad3 siRNA to

inhibit skin fibrosis induced by radiation [356]. Similarly, Wang et al. inhibited activation of

TGF-β/Smad signaling cascade by applying TGF-β type I receptor siRNA and reported reduced

hypertrophic scarring [357]. To sustain the release of these TGF-β/Smad targeting siRNAs,

delivery matrices have been designed including trimetyl chitosan [358] and a pressure-sensitive

Page 59: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

35

hydrogel [359]. MMP-responsive nanofibrous matrix was developed for diabetic wound

remodeling to control release MMP-2 siRNA and restored the wound recovery rates [360].

More recently, Charafeddine et al. developed nanoparticles encapsulating siRNA to deplete

Fidgetin-like 2 in vivo and found accelerated cell migration potentially caused by increased

directional cell motility modulated by microtubule growth [361]. In another study, Randeria et al.

demonstrated that delivery of siRNA that suppress ganglioside-monosialic acid 3 synthase, a

critical mediator of insulin resistance, in spherical nucleic acid gold nanoparticle conjugates was

able to reverse impaired diabetic wound healing [362].

2.3.4.3.3 MicroRNA (miRNA)

Besides the genetic information that is transacted by proteins, recent evidence shows that the

majority of the genome is actually transcribed into noncoding RNAs. MicroRNAs (miRNAs) are

a major group of these noncoding RNAs, and are important regulators of the coding genes by

posttranscriptional gene regulation. They are small (approximately 22-nuleotide long)

endogenously formed repressors that usually bind to the 3’-untranslated region of the target

mRNAs. Importantly, their interactions are non-complimentary and a single microRNA can

regulate multiple mRNAs simultaneously and a single mRNA can be regulated by various

microRNAs [363]. Recent studies suggest that miRNAs play important roles in dermal wound

healing including regulating angiogenesis, re-epithelialization and wound remodeling [364, 365].

Angiogenesis is an important step in wound healing and microRNAs have been shown to be

important regulators [366]. Bonauer et al. first discovered that miR-92a targets mRNAs

corresponding to several important angiogenic proteins and systematic inhibition of miR-92a

improves angiogenesis during myocardial tissue recovery [367]. In diabetic wounds, Chan et al.

showed that downregulating miR-200b supports angiogenesis and accelerates healing by

desilencing GATA binding protein 2 and VEGF receptor 2 [368]. In another study, they

described that downregulation of miR-199a-5p promotes angiogenesis both in vitro and in vivo

by inducing Ets-1 and MMP-1 expression [369]. Other studies have demonstrated the promise of

pretreatment of angiogenic cells (e.g. MSCs) with microRNAs to improve wound healing

outcomes after cell implantation [370].

Page 60: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

36

Re-epithelialization is another essential step of wound healing and has been a target for

microRNA interventions. Biswas et al. found that hypoxia induces miR-210 expression and

down-regulate the cell-cycle regulatory protein E2F3, resulting in impaired wound closure with

limited keratinocyte proliferation [371]. miR-21 has been reported to be induced by TGF-β [372]

and promote keratinocytes migration in vitro and re-epithelialization in vivo [373, 374]. However,

Pastar et al. reported contradicting results showing that miR-21 inhibits wound healing with

suppressed leptin receptor (LepR) signaling [375]. miR-203 has been discovered as an important

regulator of mRNAs responsible for both keratinocyte proliferation and migration, including

RAN and RAPH1 [376]. Sundaram1 et al. identified miR-198 as an important regulatory switch

in controlling multiple gene expressions to facilitate re-epithelialization [377]. Li et al. identified

miR-31 as another key regulator to promote keratinocyte proliferation and migration with

epithelial membrane protein 1 as its direct target [378]. In another study, they identified miR-132

as a regulator to facilitate transition from inflammation to proliferation phase and its implication

in chronic wounds, which are stalled in the inflammation phase with impaired re-epithelialization

[379].

Another important phase of wound healing is remodeling and it is essentially related to scar

formation that is usually caused by dysregulated collagen production and remodeling [380]. Kato

et al. first identified miR-192 as a regulator of TGF-β-induced collagen expression in diabetic

kidney glomeruli [381]. miR-29 has been identified as a key regulator of collagen expression in

systemic sclerosis [382] and collagen deposit in skin fibroblasts [383]. Yang et al. reported that

downregulation of miR-155 at wound sites does not accelerate wound closure but leads to a

reduced fibrosis with less collagen and α-SMA expression [384].

2.4 Cardiac tissue engineering in vitro

2.4.1 Motivation for generating cardiac micro-tissues

Cardiac tissue engineering in vitro aims to manipulate the microenvironment cells interact within

in order to facilitate cell assembly and build functional tissue with the goal of providing

replacements for diseased or damaged native tissues. Additionally, engineered heart tissue may

serve as increasingly accurate in vitro model for studies in normal and diseased heart physiology,

as well as drug discovery, validation, and toxicology [385-387]. With the advent of serum-free

Page 61: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

37

cardiac differentiation protocols [388-392] comes the ability to generate large quantities of

cardiomyocytes derived from human pluripotent stem cell sources for engineered heart tissue.

Additionally, cardiomyocyte-specific surface markers have been identified and microfluidic cell

separation methods have been advanced which can be used to purify heterogeneous populations

[393-395].

The adult mammalian heart is composed of a complex and well-integrated mosaic of anatomical

modules. The contractile muscle (atria, and ventricles) positioned between the supporting epi-

and endocardium, the conduction system (pacemaker nodes, and Purkinje fiber network), and the

highly dense vasculature (endothelial and smooth muscle cells) constitute the key elements of the

cardiac system, which is the engine for the larger cardiovascular system. In the heart, the many

cell types form specific integrated structures which contribute to their individual cell and overall

organ function. To engineer these cells in the appropriate positions and to temporally give them

the correct biochemical, physical, and electrical cues is the overarching goal (Figure 2-4).

Page 62: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

38

Figure 2-4 Engineering heart tissue for replacement therapeutics and in vitro models by physical

and electrical control of cells and microenvironment. Depiction of current methods used to

manipulate heart cells to develop, mature, and assemble into functional heart tissue. Tuning the cell

microenvironment by means of geometry and electrical control exhibits upstream effects on adhesion,

cell-cell and cell-extracellular matrix interactions, growth and differentiation, cellular and tissue

alignment via cytoskeletal organization, and electrical and contractile apparatus. The small dark arrows in

the flow diagrams indicate the sequence by which the specific method of microenvironmental control

effectively manifests downstream. These end changes in the cardiac cells include changes in gene/protein

expression, electrical properties, and mechanical properties. Top: during development pluripotent stem

cells differentiate into mesodermal progenitors, then cardiovascular progenitors that give rise to various

cell types in the heart (cardiomyocytes, fibroblasts, endothelial and smooth muscle cells). Cell

differentiation and assembly into a highly organized structure is governed by biochemical, mechanical

and electrical stimuli in vivo. Tissue engineering aims to recapitulate some of these environmental factors

in vitro. Middle: control of substrate topography and stiffness affects cell orientation and, as a result,

functional properties. Bottom: control of electrical properties is achieved by use of conductive

biomaterials, electrical stimulation bioreactors or changes in gene expression of key ion channels. The

large green arrows (middle and bottom) depict the span of current techniques used in the field and link

them to the regimes of cardiac differentiation and assembly where they have been applied (top). CM,

cardiomyocyte; CVP, cardiovascular progenitor; E-C, excitation-contraction; EC, endothelial cell; ECM,

extracellular matrix; ET, excitation threshold; FB, fibroblast; MCR, maximum capture rate; PSC,

pluripotent stem cell; SMC, smooth muscle cell.

(Reproduced with permission from BioMed Central, Thavandiran N. et al. 2013 [44])

Page 63: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

39

2.4.2 Cardiac micro-tissues as research platform

Serving as models to discover novel therapeutics or drug toxicities in vitro, single

cardiomyocytes cultured in vitro have been well-characterized by various methods, such as

immunostaining and patch-clamping [396-398]. However, single cardiomyocytes are not

necessarily an appropriate model for all toxicology studies, and do not lend themselves to in vivo

applications. Therefore, two-dimensional (2D) and 3D tissue constructs have been developed to

better recapitulate the higher functions of tissues containing a multitude of cardiomyocytes.

Studies that have focused on the development of different tissue constructs are outlined below.

Cardiomyocytes cultured as monolayers enable cell-cell coupling by gap junctions, which plays

an important role in the action potential propagation in cardiac tissue. Moreover, the alignment

and cell-cell interaction in the monolayer could be controlled by micro-patterning methods. Karp

et al. patterned photocrosslinkable chitosan on glass and tissue culture polystyrene to create cell-

repellent regions and cardiomyocytes on the cell-adhesive regions were able to beat

spontaneously after one week [399]. Nima et al. cultured cardiomyocytes following micro- and

macroscopic guidance of fibronectin coating and recapitulated the directions of native cardiac

fibres characterized by high-resolution diffusion tensor magnetic resonance imaging [97].

Microelectrode arrays (MEA) have been developed to culture cardiomyocyte monolayers and

provide insight on cardiac field potentials by impedance measurements [400-402]. Natarajan et

al. created cardiomyocyte monolayers aligned with MEAs by patterning fibronectin to guide

cellular attachment thus controlling action potential propagation [403] (Figure 2-5A). Creating

topographical cues on culture substrates guides the alignment of cardiomyocytes, which results

in the desired characteristics of anisotropy and improved physiological properties [404, 405].

Moving from 2D culture of cardiomyocyte monolayers, researchers from Parker’s group

developed contractile cardiomyocyte thin films on elastomers to measure contractile forces

combined with a quantification of action potential propagation (Figure 2-5B) [406]. This high-

throughput platform has been used to provide new insights into the pathophysiology underlying

the cardiomyopathy of Barth syndrome [407].

Native cardiac tissue has a 3D structure where it works to generate force against a load. This

structure is replete with extracellular matrix and cellular interactions on all geometric faces of

Page 64: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

40

cardiomyocytes, as well as a topography and stiffness which 2D substrates have a difficult time

replicating. 3D scaffolds made of natural or synthetic polymers can create an environment on

which dense cardiac tissue can be seeded that has properties comparable to the human

myocardium [387]. More recently, technologies including stereolithography and femtosecond

laser scanning have been applied to precisely control the geometry and structure of scaffolds. Ma

et al. seeded cardiomyocytes derived from long QT syndrome type 3 (LQT3) IPS-CMs onto

synthetic filamentous matrices fabricated by femtosecond laser and studied the cardiac

contractility in response to a panel of drugs [408].

Gel compaction has been a widely-applied method to generate cardiac tissue constructs as the

self-assembled constructs produce increased force of contraction due to the higher cell density

after the gel compaction [409]. During cultivation, the cells in the hydrogel contract and generate

pulling forces that lead to hydrogel compaction and this process highly depends on the cell

population and the hydrogel stiffness [410]. Importantly, placing microfabricated constraints to

guide the gel compaction process can control the geometries of the final cardiac microtissue.

Moreover, during cultivation, the cells developed cell-cell and cell-matrix interactions to

compact the cell-gel mixture and cell alignment was then mediated by the stress due to resistance

of constraints. Hansen et al. developed a technique to construct a large series of fibrin-based mini

engineered heart tissues that developed high yield and reproducibility (Figure 2-5C) [411].

Boudou et al. generated arrays of cardiac microtissues using small cell number (~5000 cardiac

cells) per construct with potential in high-throughput drug screening (Figure 2-5D) [412].

Thavandiran et al. showed that appropriate population percentage (25%) of nonmyocytes played

critical roles in the tissue remodeling dynamics and enhanced final structure and function

properties of cardiac microtissues [410]. The biowire platform (Figure 2-5E) used suspended

suture templates to create longitudinal microtissues that remained stable for weeks and generated

more matured cardiac tissues with electrical stimulation after the gel compaction stage [413].

Xiao et al. further developed this platform by replacing the suture template with micro-tubing to

mimic the capillaries in cardiac bundles. This created a novel method to introduce

pharmaceutical agents to cardiac tissue by perfusion and the negative inotropic effect of nitric

oxide has been demonstrated as an example [414].

Page 65: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

41

Figure 2-5 Strategies for generating 2D and 3D cardiac tissue in vitro. (A) Cardiomyocyte

monolayers cultured on patterned MEA for guided action potential propagation. (B) Cardiomyocyte

monolayers cultured on flexible elastomer films for contractile force measurement. (C) High-throughput

platform for monitoring contractile activities of an array of fibrin-based engineered heart tissues. (D)

Cardiac micro-tissues around micro-cantilevers to measure contractile properties. (E) Cardiac biowires

set-up with a suspended template to guide tissue formation and cell alignment. The phenotype of the

cardiomyocytes were matured under external electrical stimulation and the drug candidates were applied

by perfusion through micro-tubing within the biowire, providing improved physiological relevance.

(Reproduced with permission from IOP Publishing, Davenport Huyer L. et al. 2015 [53])

Page 66: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

42

Chapter 3

3 Collagen patches immobilized with growth factors or peptides for cardiac regeneration5, 6

3.1 Introduction

Tissue engineering aims at generating tissues and organs to regenerate and restore the structure

and function of their pathologically altered native counterparts [1]. Biomaterials for tissue

engineering do not merely serve as a mechanical support, but rather provide an instructive

microenvironment that facilitates cellular behaviors and tissue regeneration. General principle of

designing these biomaterials is to mimic the biological and physical properties of native ECM.

Bioactive agents such as growth factors and peptides have been incorporated in biomaterials to

promote or enhance the tissue regeneration process [415].

Growth factors are potent regulators that modulate many cellular functions including migration,

proliferation, differentiation, and survival. Various growth factors have been applied to facilitate

different stages of specific tissue regeneration process. For example, basic fibroblast growth

factor (bFGF) was administrated in a canine myocardial infarct model to enhance

neovascularization and improved left ventricular function [67]. Another potent angiogenic

growth factor, VEGF, was administrated both intra-arterially and intramuscularly in a rabbit

model of chronic hindlimb ischemia and significantly augmented revascularization [68, 69].

PDGF was the first growth factor approved by US FDA (Regranex approved in 1997) as an

adjunct to proper ulcer care in the treatment of lower extremity diabetic neuropathic ulcers [70].

However, clinical translations of growth factors have been limited, partly because of

5 Copyright © 2015 Elsevier. Contents of this chapter have been published in Methods: Xiao Y, Reis LA, Zhao Y,

Radisic M. Modifications of collagen-based biomaterials with immobilized growth factors or peptides. Methods.

2015;84:44–52. Reuse with permission from Elsevier. A link to the published paper can be found at:

http://www.sciencedirect.com/science/article/pii/S1046202315001723

6 Copyright © 2012 Elsevier. Contents of this chapter have been published in J Am Coll Cardiol: Kang K, Sun L,

Xiao Y, Li SH, Wu J, Yao TM, Weisel RD, Radisic M, Li RK. Aged human cells rejuvenated by cytokine

enhancement of biomaterials for surgical ventricular restoration. J Am Coll Cardiol. 2012;60:2237–2249. Reuse

with permission from Elsevier. A link to the published paper can be found at:

http://www.sciencedirect.com/science/article/pii/S0735109712043677

Page 67: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

43

supraphysiological uses of them risk systemic side effects such as hypotension (VEGF) and

nephrotoxicity (bFGF) [84]. In 2008, Regranex received boxed warning from FDA as patients

treated with three or more tubes of it had increased rate of mortality secondary to malignancy

[416]. Recently, we and others have demonstrated that controlled release of growth factors by

immobilization onto biomaterials localizes, enhances and sustains their bioactivities [85-87]. As

a result, the growth factors could be delivered locally and remain effective at a lower total dose,

addressing the issues for their clinical transitions.

Peptides are short amino acid sequences that are derived from primary receptor-domains of

specific proteins. The adhesion-promoting peptide derived from fibronectin, RGD, is the most

intensively-investigated peptide and has been used to promote cell attachment on different

surfaces and materials [77]. Our group has recently described a novel ang1fibrinogen-like

domain based peptide, QHREDGS [7]. It has demonstrated pro-survival benefits on cardiac cells

[7-9], endothelial cells [10], osteoblasts [12], and human iPSCs [11]. The advantages of using

small peptides is that they are more stable than their growth factor counterparts and also less

susceptible to conformational changes during immobilization or encapsulation processes [2].

Moreover, peptide sequences can be synthesized and purified in more cost-effective manner

compared to recombinant human proteins.

Traditional biomaterials for tissue engineering applications could be categorized into porous

scaffolds and hydrogels and both of them have merits that are exclusive of the other. Porous

scaffolds provide strong mechanical support and guidance for cell growth with the overall

architecture and porous structures tunable by different fabrication methods [417]. Hydrogel

systems can be delivered in a minimally invasive manner with or without encapsulating cells to

promote tissue regeneration [418]. With recent advances in microfabrication, hydrogel systems

have also been applied to generate micro-tissues in vitro for drug screening applications [410,

413, 419, 420]. Here, we discuss modification and characterization of collagen-based

biomaterials in the forms of both porous sponge and hydrogel systems with growth factors or

peptides by covalent immobilization using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide

(EDC) chemistry.

Page 68: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

44

3.2 Materials and methods

3.2.1 Materials

Avitene® Ultrafoam™ (2 mm thick, 8 cm × 12.5 cm) was purchased from Davol Inc. Ultrapure

PROTASAN™ chitosan salt (UP G 213, viscosity = 20-200 mPa·s, Mw = 200-600 kDa, 75-90%

DA) was purchased from NovaMatrix®. 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide

hydrochloride (EDC, Cat# 22980) and N-hydroxysulfosuccinimide (Sulfo-NHS, Cat#24510)

were purchased from Thermo Fisher Scientific Inc.

3.2.2 Covalent immobilization of growth factors and peptides on collagen scaffolds

The carboxyl groups (-COOH) in collagen sponge react with N-hydroxysulfosuccinimide (Sulfo-

NHS) in the presence of EDC, resulting in a semi-stable Sulfo-NHS ester intermediate, which

may then be reacted with primary amine groups (-NH2) in growth factors or peptides to form

stable amide crosslinks (Figure 3-1). The semi-stable intermediate enables us to perform step

immobilization, which has shown enhanced immobilization efficiency compared to bulk

immobilization [87]. We use phosphate buffered saline (PBS) as a buffer rather than distilled

water or 2-(N-morpholino)ethanesulfonic acid (MES) according to previous studies [87].

Figure 3-1 Reaction diagram for immobilization of growth factors or peptides on collagen sponges using 1-

ethyl-3-(3-dimethylaminopropyl) carbodiimide HCl (EDC) and N-hydroxysulfosuccinimide (Sulfo-NHS).

1. Using sterile tools, cut the 2 mm thick collagen sponge sheet into desired shape (2 cm × 2

cm).

2. For each collagen sponge (2 cm × 2 cm), prepare 1 mL EDC reaction solution composed of

Page 69: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

45

24 mg/mL EDC and 60 mg/mL Sulfo-NHS in PBS (Lonza).

3. Filter-sterilize the EDC reaction solution using sterile 0.2 μm syringe filters (VWR) and use

it promptly.

4. Immerse the collagen sponge into the EDC reaction solution in a 6-well plate and allow the

activation to proceed for 20 minutes at room temperature.

5. Prepare 600 μL growth factor solution composed of 1 µg/mL VEGF (PeproTech) and 1

µg/mL bFGF (PeproTech) or peptide solution composed of 1 µg/mL QHREDGS (Biomatik)

in PBS in another 6-well plate.

6. Remove the activated collagen sponge from EDC reaction solution and drain excess liquid

by dabbing to the wall.

Note: The NHS-activated collagen sponge should be proceeded immediately to step 7.

7. Immerse the activated collagen sponge into prepared growth factor solution or peptide

solution and allow the reaction to proceed for 2 hours at room temperature.

8. Wash the sponge with fresh PBS for 8 times of at least 5 minutes each time, to wash away

the uncrosslinked EDC, sulfo-NHS, and growth factors or peptides.

9. Keep the sponge in PBS at 4°C and use within 24 hours after preparation.

3.2.3 Quantification of growth factor immobilization efficiency

In order to investigate the amount of growth factors immobilized onto collagen sponge, modified

sponges were prepared freshly (immobilization group). Sponges were also prepared following

the protocol without adding the EDC reaction solution (physical adsorption group) or without

adding the growth factor solution (control group). All the washes in step 8 were collected

(labelled as supernatant). After the washes, all the collagen sponges were digested by 0.276

mg/mL collagenase type IA (Sigma Aldrich) for 1.5 hours at 37 °C and the digestion solution

was collected. All the samples were stored at -80 °C if not used immediately.

Enzyme-linked immunosorbent assay (ELISA) utilizes antigen-antibody interaction and color

developing enzyme to analyze amount of specific proteins in solution. Here we used ELISA

Development Kit (PeproTech) to quantify the amount of immobilized VEGF and bFGF on

collagen sponges and the unbounded amount in the supernatant in all three groups

(immobilization, physical adsorption, and control). To obtain accurate results, all samples and

standards were arranged in the same ELISA plate.

Page 70: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

46

1. Reconstitute all the components from the kit, aliquot and store at recommended temperature.

2. Dilute capture antibody with PBS to 0.5 μg/mL and add 100 μL to each ELISA plate well.

Seal the plate with parafilm and incubate overnight at room temperature. Aspirate and wash

the plate 4 times.

Note: After each wash, it is easier to remove liquid by blotting on clean paper towel.

3. Add 300 μL blocking buffer to each well and incubate for 2 hours at room temperature.

Aspirate and wash plate 4 times.

4. Prepare standards (from the ELISA kit) and samples on ice (dilute with PBS when necessary)

and add 100 μL to each well in triplicate. Incubate at room temperature for at least 2 hours.

Aspirate and wash the plate 4 times.

5. Dilute detection antibody in 0.05% Tween-20, 0.1% BSA to the final concentration of 0.25

μg/mL and add 100 μL to each well. Incubate at room temperature for 2 hours. Aspirate and

wash plate 4 times.

6. Dilute avidin-HRP conjugate 1:2000 in 0.05% Tween-20, 0.1% bovine serum albumin (BSA)

and add 100 μL to each well. Incubate 30 minutes at room temperature. Aspirate and wash

the plate 4 times.

7. 2,2'-Azinobis [3-ethylbenzothiazoline-6-sulfonic acid]-diammonium salt (ABTS) liquid

substrate should be equilibrated at room temperature. Add 100 μL of substrate solution to

each well. Shake the plate gently and monitor color development with SpectraMax i3

(Molecular Device) at 405 nm at 5-minute intervals for 30 minutes.

8. The built-in software (SoftMax Pro 6.4) calculates the concentration of target growth factor

in sample solutions automatically by comparing with standard solutions based on absorbance

readings.

3.2.4 Quantification of QHREDGS peptide immobilization efficiency

Fluorescently labelled peptide, FITC-QHREDGS was used to assess the conjugation efficiency

and true final concentration of peptide attached to chitosan post dialysis. This then determined

the final concentration of peptide in the hydrogels. FITC-QHREDGS (Biomatik) was substituted

for regular peptide in the protocol above with the critical addendum that all steps in the method

were protected from light to prevent photobleaching of the fluorophore. As the molecular weight

cut-off of the dialysis membrane is at most a tenth that of chitosan it can be safely assumed that

Page 71: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

47

all of the chitosan is retained and recovered, and the peptide present is only that which was

successfully attached to the chitosan.

1. Standards of the FITC-QHREDGS in PBS should be made ranging from 0.0005 to 0.01

mg/mL.

2. Both the standards and the reaction solutions recovered post dialysis should have their pH

adjusted to 7 using 0.1N NaOH and 0.1N HCl, as fluorescence is greatly affected by pH

[421].

3. Test samples (reaction solutions recovered post dialysis) should be diluted 1:10 and 1:100.

4. Pipette 100μL of sample or standards, in triplicate, into wells of a 96-well black plate.

5. Run the plates through a fluorometer (Spectra Max Geminin EM, Molecular Devices) at an

excitation wavelength of 490 nm and emission of 520 nm.

6. The true final concentrations of the peptide and conjugation efficiency are calculated by

comparing the fluorescence of the samples to the standards, correcting for the dilution factor

and for the volume recovered post dialysis.

3.2.5 Characterization of release profile

It is important to investigate the release profile of the growth factors or peptides immobilized on

the modified biomaterials to confirm that they are being released gradually over extended time

(e.g. 2-3 weeks). We immersed freshly-prepared sponges in 1 mL PBS in 24-well plates and

incubated at 37 °C for 28 days. The supernatant was collected on day 1, 3, 7, 14, 21, and 28 and

stored at -80 °C. The collagen sponge was digested after collecting the supernatant on day 28 by

methods described in 3.2.2. All the samples (all the supernatant samples and digestion samples)

were characterized for VEGF and bFGF amount by ELISA as described before. Similarly, in the

case of peptide immobilization, the supernatant was collected and analyzed as described in 3.2.3.

3.2.6 Scanning electron microscopy

Porous scaffolds provide mechanical support and guidance for the cell infiltration and it is

necessary to confirm that our modification does not change their porous structures (e.g. clotting

the pores). We used environmental scanning electron microscopy (Hitachi S-3400 N) to examine

both collagen sponges and collagen-chitosan hydrogel samples. A filter paper was used to gently

remove the excess water from the samples. The chamber was closed and the temperature of the

Page 72: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

48

chamber decreased to -20°C. The samples were imaged under variable pressure mode at 70 Pa

and15 kV.

3.2.7 Tensile testing of porous collagen scaffolds

The collection of human samples was approved by the Research Ethics Board of University

Health Network. Each patient provided informed consent. Bone marrow aspirates were obtained

from the sternum of patients undergoing coronary artery bypass grafting (CABG) at Toronto

General Hospital. “Young” hMSCs were isolated from patients age ≤57 years (50.0 ± 8.0 years,

N = 4); “old” hMSCs were isolated from patients age ≥66 years (74.5 ± 7.4 years, N = 4). Four

groups of scaffolds were tested with and without growth factors: fresh (scaffolds right after

preparation), blank (scaffolds incubated in media for 3 days), young (scaffolds seeded with

young hMSCs and incubated in media for 3 days), and old (scaffolds seeded with old hMSCs and

incubated in media for 3 days).

We used a protocol modified based on ASTM D412-06a to perform the tensile testing of

modified collagen sponges using ElectroForce 5200 BioDynamic Test Instrument (Bose) with a

22 N load sensor. Please note that “displacement” in following protocol is defined by the

ElectroForce program as the distance between the two grips.

1. After modification (Step 2.2), carefully cut the collagen sponge into straight specimen (2 cm

× 1 cm) and briefly dry with Kimwipes (50 ± 5% moisture).

2. Start the machine and set the displacement to be 2 mm.

3. Place the specimen in both grips to the same depth, carefully adjust the grips to align the

specimen with the direction of pull. Make sure the specimen is not stretched initially and

reset the force reading to 0.

4. Set the displacement rate at 1 mm/min and displacement to 3 mm (approximately 50%

strain). Start recording both displacement and force reading.

5. Start pulling and record the distance L0 where force reading starts increasing. Wait until

displacement stops at 3 mm.

6. Set the displacement to 2 mm (original distance between the two grips) and start again. The

force reading should decrease to 0 at displacement L0, indicating that the sponge behaves

like elastomer within 50% strain range.

Page 73: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

49

Note: We aim for 50% strain range based on final application for heart implantation. The elastic

behavior would be better verified by plotting the stress-strain curve.

7. Set the displacement to 10 mm and start. Release the specimen after rupture.

Calculations:

1. Calculate the strain as the elongation of specimen in percentage:

ε = the strain of the specimen,

L = observed displacement,

L0 = initial displacement when the specimen is not extended.

2. Calculate the tensile stress at any specified strain as follows:

T(x) = tensile stress at (x) % elongation,

F(x) = force at specified elongation,

A = cross-sectional area of unstrained specimen.

3. Calculate the ultimate tensile strength as follows:

UTS = ultimate tensile strength, the stress at rupture,

Ft = the force at rupture,

A = cross-sectional area of unstrained specimen.

4. Young’s modulus should be calculated from the elastic region of the stress-strain curve as

follows:

E = Young’s modulus

Tx = tensile stress at (x) % elongation,

εx = the strain on the specimen at (x) % elongation.

Page 74: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

50

3.3 Results and discussion

It is important to characterize the mechanical properties of the modified porous scaffolds to

ensure it will withstand tensile force for adequate performance in certain tissue engineering

application. For example, scaffolds for myocardium implantation should have Young’s modulus

close to the native myocardium (10-150 kPa in the physiological regime [422]) and remain

elastic within the deformation of myocardial segments during beating (about 20% in longitudinal

direction and up to 60% in radial direction [423, 424]). We characterized the scaffolds to ensure

that cytokine immobilization did not adversely affect the stiffness, strength, or porosity of the

scaffolds and, therefore, their usefulness for surgical ventricular restoration (SVR). Stress–strain

curves demonstrated that cytokine-free and -enhanced scaffolds exhibited similar behaviors

during the breaking process (Figure 3-2A). Young’s modulus, a measure of stiffness, did not

differ with the addition of growth factors to the scaffold (Figure 3-2B). Ultimate tensile strength,

the maximum stress a material can withstand while being stretched, was lower in cytokine-

enhanced scaffolds than in cytokine-free scaffolds, with the exception of the Old group (Figure

3-2C). However, the ultimate tensile strength was calculated at the final breaking point, at

approximately 300% strain. Because this strain is not applicable in vivo, the Young’s modulus is

more suitable for characterizing the mechanical property of the scaffolds for our application.

Scanning electron microscopy showed no significant differences in the porosity or pore

structures of the scaffolds with or without cytokines (Figure 3-2D).

To ensure covalent immobilization of the growth factors to the collagen scaffolds (and not just

physical attachment), we used ELISA to determine the immobilization efficiencies for VEGF

and bFGF, which were 42% and 24%, respectively, and the physical bonding efficiencies, which

were 2% and 5% (Figure 3-2E), demonstrating that both cytokines were chemically immobilized

to the scaffolds. The scaffolds were prepared to provide a slow release of cytokines over time

after SVR. The release study, based on ELISA, demonstrated that the amount of cytokine

released from the scaffolds over 4 weeks was small, but the release rates were similar for VEGF

and bFGF (Figure 3-2F). A significant amount of each cytokine remained in the scaffolds after

28 days (Figure 3-2G).

Page 75: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

51

Figure 3-2 Characterization of scaffolds. (A) Stress-strain curves for cytokine-free scaffolds (solid lines) and

cytokine-enhanced scaffolds (dashed lines) showed similar tensile strength with or without cells. (B) Young’s

modulus was not significantly different between cytokine-free and –enhanced scaffolds. (C) Ultimate tensile

Page 76: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

52

strength was significantly lower in cytokine-enhanced scaffolds in all groups but the Old group (**p < 0.01).

However, this measure was obtained outside of the physiological range of strain. Young’s modulus provides a better

estimate of the physical characteristics of the patch in vivo. (D) Scanning electron microscopy at 100× (scale bar =

500 μm) and 500× (scale bar = 100 μm) magnification revealed similar porosity in scaffolds with and without

growth factors. (E) Enzyme-linked immunosorbent assay (ELISA) demonstrated that both vascular endothelial

growth factor (VEGF) and basic fibroblast growth factor (bFGF) (150 ng of each added to the scaffolds) were

immobilized on the scaffolds, with little in the supernatant. Scaffolds without 1-ethyl-3-(3-dimethylaminopropyl)

carbodiimide HCl/N-hydroxysulfosuccinimide treatment served to show physical bonding of the cytokines to the

scaffolds (not covalent immobilization). (F) ELISA demonstrated that little VEGF and bFGF was released from the

scaffolds over 28 days. (G) After day 28, ELISA showed that substantial amounts of VEGF and bFGF remained in

the scaffolds. n = 3/group. bFGF = basic fibroblast growth factor; EDC = 1-ethyl-3-(3-dimethylaminopropyl)

carbodiimide HCl; GF = growth factor; PBS = phosphate-buffered saline; VEGF = vascular endothelial growth

factor.

The described covalent immobilization strategy using EDC chemistry is effective in

immobilizing growth factors or peptides to both porous scaffold and hydrogels. We characterized

collagen sponge modified with FITC-QHREDGS with the critical addendum that all steps in the

method were protected from light to prevent photobleaching of the fluorophore (Figure 3-3).

The presence of fluorescently labelled immobilized molecule was visible using even simple

fluorescence microscopy in comparison to the blank scaffolds and the scaffolds that relied on

physical adsorption alone (Figure 3-3A).

The immobilization efficiency was characterized using sample preparation described in 3.2.1 and

fluorescence-based assay described in 3.2.3, demonstrating up to eight-fold higher amount of the

immobilized QHREDGS peptide in the EDC treated scaffold compared to the physical

adsorption alone (Figure 3-3B). Importantly, as the scaffold degrades using cell culture or

incubation with PBS or culture media, the peptide will slowly be released into the environment.

This obviates the need for continuous application of the biomolecule with culture media changes,

thus it decreases the totally applied dose. Importantly, the pore structure and porosity was not

appreciably changed with EDC based covalent immobilization of biomolecules (Figure 3-3C).

Page 77: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

53

Figure 3-3 Characterisation of peptide immobilization. (A) FITC-QHREDGS modified collagen sponges under

the microscope (all images acquired at same exposure time). Left: control sponge modified with EDC reaction

solution without adding FITC-QHREDGS; Middle: physical adsorption sponge modified with FTIC-QHREDGS

without adding EDC reaction solution; Right: immobilization sponge modified with EDC reaction solution with

presence of FITC-QHREDGS. Scale bar = 200 μm. (B) Quantification of the amount of FITC-QHREDGS peptide in

freshly prepared collagen sponges. The signals in the control group come from collagen auto-fluorescence. (C) SEM

images of the collagen sponges with (right) or without (left) EDC modification.

Using the immobilization protocol described in 3.2.1, the immobilization efficiency should be

above 20% as we described previously [181]. The amount of growth factors or peptides released

over 4 weeks in aqueous media (e.g. PBS) should be small with significant amount remaining in

Page 78: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

54

the scaffolds after 4 weeks [181]. The mechanical properties of collagen sponges will change as

we showed an increased Young’s modulus due to crosslinking of collagen molecules to

themselves [425]. The collagen sponge modified with VEGF and bFGF has been shown to

promote proliferation and down-regulate aging-related gene expression of human MSCs from old

donors, which improves the cell function to restore cardiac function after SVR [181].

Using the conjugation protocol described in 2.3.1, the conjugation efficiency of the peptide to

chitosan should be above 50% as we described previously [9]. Characterized by rheology, the

modified 1:1 chitosan-collagen hydrogel should be mechanically stable at 37°C based on the

values of its storage modulus [9]. As characterized by live/dead staining and lactate

dehydrogenase (LDH) assay, the presence of QHREDGS peptide should improve cardiomyocyte

viability and metabolic activity in vitro as we showed previously [9]. Compared to unmodified

hydrogel, the peptide-conjugated chitosan-collagen hydrogel also improves the morphology of

cardiomyocytes in vitro [9]. Moreover, the modified hydrogel inhibits paclitaxel-induced

apoptosis of endothelial cells and stimulates tube-like structure formation in vitro [10].

3.4 Conclusion

Here we have described our protocols for preparation and characterization of collagen-based

biomaterials (both porous scaffolds and hydrogels) modified with growth factors or peptide by

EDC chemistry. As detailed above, quick handling during EDC crosslinking is critical for

effective immobilization. In addition, the physical and biological properties of the modified

biomaterials should be carefully characterized for different tissue engineering applications. The

bioactivity of immobilized growth factors or peptides should be tested on in vitro cell cultures

before in vivo application.

3.5 Acknowledgments

This work is funded by the Heart and Stoke Foundation GIA T6946, the Canadian Institutes of

Health Research (CIHR) Operating Grant (MOP-126027), NSERC-CIHR Collaborative Health

Research, Grant (CHRPJ 385981-10), NSERC Discovery Grant (RGPIN 326982-10), and

National Institutes of Health grant 2R01 HL076485. M.R. is supported by Canada Research

Chair (Tier 2) and Steacie Fellowship.

Page 79: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

55

Chapter 4

4 Diabetic wound regeneration using peptide-modified hydrogel targeting the epithelium

4.1 Introduction

Chronic ulcers are considered a major healthcare challenge as they affect 6.5 million people in

the United States [426]. Non-healing wounds, including chronic ulcers, can be caused by a

number of common diseases and medications, such as vascular insufficiency, diabetes mellitus,

and local-pressure effects, which disrupt the well-orchestrated cellular and molecular interactions

during the wound healing process [235]. Specifically, diabetic foot ulcers affect 15% of people

with diabetes and are a leading cause of amputation [427]. The mechanism underlying diabetic

chronic wounds remains elusive and new interventions for diabetes-impaired wound healing are

needed. After almost two decades without new chemical entities approved by the Food and Drug

Administration (FDA) (Regranex® was approved in 1997), it has been recognized that an

optimal wound healing outcome requires a multifaceted approach that addresses different issues

(e.g. persistent inflammation, insufficient angiogenesis, and impaired re-epithelialization) at once

[235].

Keratinocytes are the major cell type in the epidermis, the outermost layer of skin. Upon injury,

keratinocytes migrate from the wound edge into the wound to re-epithelialize the damaged tissue

and restore the epidermal barrier. The hallmark of non-healing human wounds is non-migratory

and hyper-proliferative keratinocytes, resulting in epidermis thickening at the wound edge and an

absence of wound closure [428]. Scarless embryonic wound healing and complete healing in

animals with a high regenerative potential such as newts critically depend on rapid re-

epithelialization [429, 430].

Additionally, non-healing diabetic wounds are trapped in a state of prolonged inflammation,

characterized by supra-physiological oxidative stress that can induce keratinocyte injury,

dysfunction and apoptosis [431-433]. It results from the excess production of reactive oxygen

species (ROS) by macrophages and neutrophils, coupled with an impaired antioxidant defense

capability in response to hyperglycemia [253]. Moreover, altered extracellular matrix (ECM)

Page 80: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

56

composition in non-healing wounds and an enhanced ECM degradation rate due to elevated

matrix metalloproteinase levels can impair keratinocyte attachment, leading to aberrant cell

signaling and impaired migration [250, 251, 434].

To address these challenges, we sought to develop a novel wound healing approach that could

recapitulate key aspects of scarless embryonic wound healing by 1) promoting effective

keratinocyte migration, 2) protecting the wound-bed cells against oxidative stress and 3)

providing a new matrix for cell attachment.

Our group has recently described a novel angiopoietin-1derived peptide, QHREDGS, which

interacts with integrins, receptors that function in cell-adhesion and ECM-binding. The

QHREDGS peptide was shown to enhance endothelial cell metabolism, tube formation kinetics,

and survival in response to apoptotic stimuli [10]. QHREDGS was also shown to promote

neonatal rat cardiomyocyte attachment and survival [7], to inhibit human induced pluripotent

stem cell (hiPSC) apoptosis during cells expansion [11], to induce osteoblast matrix deposition

and mineralization [12], and to have cardiac protective effects in a chitosan-collagen hydrogel

both in vitro and in vivo [9, 13].

We therefore hypothesized that the QHREDGS peptide could promote keratinocyte survival and

migration and thereby accelerate diabetic wound healing. We investigated the effect of the

QHREDGS peptide as a soluble supplement on the survival of normal human keratinocytes upon

oxidative stress. The effect on attachment, survival upon oxidative stress, and collective

migration of both normal and diabetic keratinocytes was assessed by immobilizing the

QHREDGS peptide within a chitosan-collagen film coating. We further investigated the ability

of the QHREDGS peptide to promote diabetic wound repair in vivo using a full-thickness

excision wound model in db/db diabetic mice.

4.2 Materials and methods

4.2.1 Primary human keratinocytes cell culture

Primary neonatal human epithelial keratinocytes (HEKs) were purchased (Cascade Biologics)

and cultured in EpiLife medium supplemented with EpiLife Defined Growth Supplement (EDGS)

as recommended by the manufacturer (Cascade Biologics; referred to as complete medium).

Page 81: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

57

HEKs were cultured on surfaces coated with a coating matrix kit (Cascade Biologics) and

passaged using 0.025% trypsin/EDTA (Cascade Biologics) until 70-80% confluence was reached.

Third and fourth passage HEKs were used in our experiments.

Diabetic human adult epithelial keratinocytes (DHEKs) from a patient (72 years old female) with

type II diabetes were purchased from Lonza and cultured in KGM-GoldTM BulletKitTM

medium as recommended by the manufacturer (Lonza). DHEKs were passaged using

ReagentPackTM Subculture Reagents (CC-5034, Lonza) until 70-80% confluent. Second and

third passage DHEKs were used in our experiments.

4.2.2 Evaluation of soluble QHREDGS in vitro

The effect of the QHREDGS peptide as in the soluble form was assessed on HEKs in vitro. 24-

or 96-well plates were coated with 0.05 mg/mL type I collagen (BD Biosciences) in 0.02 N

acetic acid overnight and then rinsed once with phosphate buffered saline (PBS) (Lonza). HEKs

were seeded in complete EpiLife medium and attached for at least 2 h. 100 µM or 650 µM

QHREDGS was supplemented to the EpiLife medium as Low or High dosage, respectively.

4.2.3 Proliferation assay

HEKs were seeded at a density of 1×104 cells/cm2 in collagen-coated 96-well plates. After 4 h,

media was removed and replenished with EpiLife media supplemented with 20 µM

bromodeoxyuridine (BrdU) (Sigma Aldrich) in the presence or absence of QHREDGS peptide.

After incubating for 8 h, proliferating HEKs were identified by BrdU staining. Briefly, HEKs

were fixed with 4% paraformaldehyde (PFA), permeablized with 0.25% triton X, treated with

DNase for 30 min at 37 °C, and blocked with 5% bovine serum albumin (BSA). The HEKs were

then incubated with a rat anti-BrdU antibody (AbD Serotec) overnight at 4 °C followed by an

anti-rat TRITC-labelled secondary antibody (Jackson Immuno Research). BrdU-positive cells

were counted in 5 randomly chosen fields under 20× magnification and then normalized to the

total number of cells labelled by 4',6-diamidino-2-phenylindole (DAPI) counterstaining.

4.2.4 H2O2 treatment on HEKs with soluble QHREDGS peptide

HEKs were treated by H2O2 following the regimen shown in Fig 4-1B. HEKs were seeded at a

sub-confluent density in a 96-well plate for the cell integrity assay or a 24-well plate for Western

blot analysis, and serum starved overnight. The HEKs were then pretreated with soluble

Page 82: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

58

QHREDGS (100 µM for Low and 650 µM for High) for 2 h. The cells were then exposed to

fresh EpiLife basal medium supplemented with 500 µM H2O2 in the presence or absence of

QHREDGS peptide. For the cell integrity assay, the cells were stained using the EarlyToxTM Cell

Integrity kit (Molecular Devices) after 2 h of H2O2 treatment. For the Western analysis, another

set of wells were seeded in parallel for the Ctrl, Low and High conditions and protein samples

were collected after 0, 15 min and 2 h of H2O2 treatment.

4.2.5 Conjugation of QHREDGS to chitosan

The QHREDGS peptide was conjugated to chitosan using 1-ethyl-3-(3-dimethylaminopropyl)

carbodiimide (EDC) chemistry as previously described [88]. Briefly, chitosan (UP G 113,

Novamatrix) was dissolved at 20 mg/ml in 0.9% normal saline and the QHREDGS peptide at 10

mg/ml in PBS. These were then mixed with EDC and N-hydroxysulfosuccinimide (S-NHS),

dissolved in PBS, to a final solution concentration of 5mg/ml chitosan and 0.5 mg/ml of

QHREDGS peptide (Low peptide group) or 3 mg/ml of QHREDGS peptide (High peptide

group). In the reaction solution, the mass ratio of [EDC]/ [peptide] and [S-NHS]/ [EDC] were

kept constant at 0.8 and 2.75, respectively. The reaction solution was left on a vortex mixer

(VWR) at 650 rpm for 3 h, diluted 4× with PBS and dialyzed against distilled water for 48 h

(Spectra/POR MWCO 3500, Spectrum Labs). The dialyzed solution was then filter sterilized,

lyophilized for 48 h and stored at -20 °C until use.

4.2.6 Solvent casting of chitosan-collagen films

The chitosan samples were reconstituted at 2 mg/ml in 0.5 N acetic acid and mixed with 2 mg/ml

type I collagen (BD Biosciences) to obtain a film coating solution composed of 1 mg/ml each of

chitosan and collagen. 24-well plates were coated with 250 µL film coating solution per well and

96 well plates with 50 µL per well. The film coating solution was fully evaporated in a biosafety

hood and the chitosan-collagen films were cast in the wells. The coated plates were rinsed three

times with ample PBS before use.

4.2.7 Coating validation

Fluorescently labeled peptide, FITC-QHREDGS (Biomatik), was used to validate peptide

concentrations in the film. FITC-QHREDGS was substituted for regular peptide in the protocol

above and all steps were protected from light. Standards of the FITC-QHREDGS in PBS were

Page 83: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

59

made ranging from 0.025 pg/ml to 0.5 pg/ml. After rinsing with PBS three times, the coated 96-

wells were filled with 200 µl PBS and then run, with the standards in the same plate, through a

fluorometer (SpectraMax i3, Molecular Devices) at excitation and emission wavelengths of 490

nm and 520 nm, respectively. The final amounts of peptide were quantified using SoftMax Pro

6.4 software.

4.2.8 Keratinocyte attachment on chitosan-only films

Chitosan-only films were prepared by casting 1 mg/ml chitosan with the presence or absence of

conjugated QHREDGS peptide in 0.5 N acetic acid in the wells of a 96-well plate and drying in

the a biosafety hood overnight. The coated plates were rinsed three times with ample PBS before

use. HEKs or DHEKs were seeded in supplemented EpiLife or KGM medium and allowed to

attach on the chitosan-only films. After 2 h, unattached cells were carefully rinsed off using PBS

and cells were then fixed with 4% paraformaldehyde. The number of attached cells were

quantified using DAPI counterstaining and the cell numbers on chitosan-only films in the

presence or absence of conjugated QHREDGS peptide were normalized to the number of cells

attached to regular tissue culture polystyrene (TCP).

4.2.9 H2O2 treatment on keratinocytes on the chitosan-collagen films

HEKs or DHEKs were seeded in complete EpiLife or KGM medium and allowed to attach for 4

h. HEKs were then changed to basal EpiLife medium supplemented with 500 µM H2O2 and

DHEKs were changed to basal KBM medium supplemented with 2 mM H2O2. After 2 h, cells

were changed to a complete medium with EarlyTox™ Cell Integrity staining reagent and

subjected to cell integrity assay. Cells treated by H2O2 for 15 min were used for Western blotting

together with non-treated controls.

4.2.10 EarlyToxTM Cell Integrity assay

The EarlyTox™ Cell Integrity Kit (Molecular Devices, R8213) is based on two nuclear dyes: live

red dye is cell permeant and marks both live and dead cells (Excitation: 622 nm/Emission: 645

nm); dead green dye is cell impermeant and stains only cells with damaged outer membranes

(Excitation: 503 nm/Emission: 526 nm). To avoid cell detachment, half of the medium in each 96

well was removed by micropipette and the equal volume of the staining solution with double

concentration of the dyes was added into the well carefully, such that the final concentration in

Page 84: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

60

the well was the one suggested by the manufacturer. The plate was then incubated at 37 °C for

30 min and imaged using a SpectraMax i3 plate reader (Molecular Devices). The percentage of

viable cells after H2O2 treatment on chitosan-collagen films in the presence or absence of

QHREDGS peptide was normalized to the viability percentage of non-treated controls on the

same coating film condition.

4.2.11 Western blotting

Protein was isolated from keratinocytes in the 24-well plate after H2O2 treatment using 60-80 µL

Lysis Buffer per well (10× Cell Lysis Buffer, Cell Signaling Technology; complete Mini,

EDTA-free protease inhibitor cocktail tablet, Roche; in ddH2O). Proteins were separated by

electrophoresis in Novex Tris-Glycine gels (Life technologies) and transferred using the iBlot

(Life technologies) to a PVDF iBlot Transfer Stack (Life technologies). Membranes were probed

for phospho-p44/42 MAPK (pMAPKp42/44), p44/42 MAPK (MAPKp42/44), phospho-Akt, Akt, or

GAPDH as a loading control (Millipore). All primary antibodies were purchased from Cell

Signaling unless stated otherwise. HRP conjugated goat anti—mouse or goat anti—rabbit

secondary antibodies were used (DAKO). Membranes were developed with Amersham ECL or

Amersham ECL Prime Western Blotting Detection Reagent (GE Healthcare) and exposed to the

films. The films were scanned and densitometry was performed using ImageJ or Image Studio™

Lite (LI-COR Biosciences).

4.2.12 Migration assay

Culture-inserts were purchased from ibidiⓇand carefully placed on the chitosan-collagen films in

24-wells. HEKs were seeded in complete EpiLife medium at 0.3-0.4×106 cells/mL with 70-100

µL per chamber and allowed to attach for 2 h. The culture-inserts were then carefully lifted and

the unattached HEKs were immediately removed by rinsing twice using warm PBS. Basal

EpiLife medium was then added to the wells and the wells were imaged every 2 h for 8 h and the

next day using the SpectraMax i3 plate reader. After 24 h, HEKs were fixed with 4%

paraformaldehyde.

The Ca2+ level in the EpiLife medium was increased from 0.06 mM to 0.12 mM to ensure HEKs

migrated collectively. After initiating the HEK migration by lifting the ibidiⓇ culture inserts,

HEKs were maintained in EpiLife basal medium supplemented with CaCl2 at a final Ca2+

Page 85: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

61

concentration of 0.12 mM. HEKs were fixed with 4% paraformaldehyde after exposure to

increased Ca2+ concentration for the indicated period of time.

Similarly, DHEKs were seeded in complete KGM medium with CellMask Green (1000× dilution)

at a density of 0.15-0.2×106 cells/mL in ibidi migration chambers and allowed to attach for 2 h.

Migrations were initiated by lifting the culture-inserts as mentioned above and the wells were

imaged every 2 h for 6 h using the SpectraMax i3 plate reader. After 6 h, DHEKs were fixed

with 4% paraformaldehyde.

4.2.13 Immunostaining

At the end of migration, HEKs and DHEKs were fixed and stored in PBS at 4 °C. Cell

monolayers were permeablized with 0.25% triton X, blocked with 5% bovine serum albumin

(BSA), and then incubated with mouse anti-E-cadherin primary antibody (BD Biosciences; 1:200)

overnight at 4 °C followed by goat anti-mouse Alexa 488 secondary antibody (Jackson Immuno

Research; 1:400). Cell nuclei were counterstained with 4',6-diamidino-2-phenylindole (DAPI)

(Biotium; 1:100). The 24-well plates were imaged at 20× magnification using a fluorescence

microscope (Olympus IX81).

4.2.14 Animals, wound model, and treatment

The Animal Care Committee of the University of Toronto approved all described animal studies.

8 weeks old, genetically diabetic, maleBKS.Cg-Dock7m +/+ Leprdb/J mice (db/db) (Stock 000642)

were ordered from Jackson Laboratories (Bar Harbor, USA). Mice were acclimatized for one

week and their blood glucose levels were tested with a glucometer (Accu-ChekⓇ Aviva) to

confirm plasma glucose levels were over 300 mg/dL the day before surgery.

The chitosan-collagen hydrogel was prepared in a similar manner as previously described.[88]

The final hydrogel consisted of 2.5 mg/ml chitosan (with or without conjugated QHREDGS

peptide) and 2.5 mg/ml type I collagen neutralized by 1 N NaOH and 10× PBS. The final

hydrogel solution was mixed thoroughly and kept on ice until use. For in vivo application, only

the Low-peptide chitosan-collagen was applied and the pre-gel solution was warmed for about 10

min at 37 °C to initiate the gelling process and applied to the wound site with a 23½ G needle.

Page 86: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

62

Mice were anaesthetized with inhaled isoflurane (5%) and the dorsal surface of the mouse was

shaved with an electric shaver, followed by treatment with a hair removal cream (VeetⓇ).

Betadine and 70% ethanol were applied in series to the surgical site. 8 mm Biopsy punches

(VWR) were used to create mid-dorsal full-thickness wounds by excising the epidermis and

dermis, including the panniculus carnosus. Either 50 µL control hydrogel without conjugated

QHREDGS peptide or 50 µL hydrogel with Low peptide (containing a total of 2.2nmol peptide)

was applied topically to the wound beds, or the wounds were left untreated as blank. The wound

beds were then covered by Tegaderm™ film (Fig. 4-1A). Buprenorphine (0.03 mg/kg) was given

subcutaneously before and right after the surgery as an analgesic. Thereafter, the mice were

housed individually and observed every other day. Digital photographs of wounds were taken at

the same distance by a camera (Canon) with a calibration scale on the side every two days. Mice

were sacrificed using CO2 asphyxiation, followed by cervical dislocation, on day 14.

4.2.15 Histology analysis

Following euthanasia, the wound tissue was excised together with surrounding tissue and fixed

in 10% formalin (Sigma). Tissue samples were embedded in paraffin blocks and then sliced into

5 µm-thick sections. Sections were processed and stained with hematoxylin and eosin (H&E),

Masson’s trichrome, or immunostained using anti-CD31 and anti-smooth muscle actin (SMA)

antibodies at the University Health Network Pathology Research Program laboratory. Stained

slides were scanned (20×) using the Aperio ScanScope XT (Aperio Technologies, USA) at the

Advanced Optical Microscopy Facility (AOMF, Toronto, Canada). The images of scanned slides

were analyzed using the Aperio ImageScope (Version 11).

In order to characterize the healing of the wounds, Masson’s trichrome stained slides were

scanned with a ScanScope XT whole slide scanner and measured using Aperio ImageScope (v11,

Aperio Technologies). The wound edge was defined as the panniculus carnosus muscle gap, the

epithelial gap as the distance between the epithelial tongues, and the re-epithelization percentage

as the ratio of epithelial gap over wound edge. The size of the granulation tissue was defined by

the highly cellular tissue between epidermis and the fat/muscle tissue. The epidermal thickness

was defined as the average thickness of the leading epithelial tongue (300 µm) from both ends.

Page 87: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

63

In order to characterize angiogenesis, six views (300 µm × 300 µm) within the granulation tissue

were randomly selected for each scanned CD31 slide and then the number of CD31 positive

vessel-like structures was counted and normalized to the area of granulation tissue in the view to

determine the vessel density. Similarly, the SMA positive area percentage was determined from

six random views within the granulation tissue. An automated algorithm built-in with the Aperio

ImageScope (Microvessel Analysis Algorithm) was used to quantify the lumen area, vascular

area, vessel area, vessel perimeter, and vessel wall thickness of each CD31 positive vessel-like

structure over the entire granulation tissue.

4.2.16 Microvessel analysis algorithm

Scanned images of the histology slides stained with anti-CD31 antibody were analyzed using the

Microvessel Analysis Algorithm with Aperio ImageScope (Version 11). The entire granulation

tissue was selected using a drawing tool and CD31 positive staining was thresholded by Color

Deconvolution. Microvessels were thresholded using a Region Joining Parameter of 8 μm, a

Vessel Completion Parameter of 10 μm, and a Vessel Area between 1 and 20000 μm2. The

automated algorithm calculated the Lumen Area, Vascular Area, Vessel Area, Vessel Perimeter,

and Vessel Wall Thickness for each vessel within the selected granulation tissue area and output

the results in histograms.

4.2.17 Statistical analysis

All results are presented as mean ± SD. Statistical analysis was performed using SigmaPlot 11.0.

Differences between experimental groups were analyzed using one-way or two-way ANOVA

followed by a Tukey post hoc test for pairwise comparison. A value of P < 0.05 was considered

as statistically significant.

4.3 Results

4.3.1 QHREDGS peptide prevents H2O2-induced apoptosis in human primary keratinocytes and upregulates Akt and MAPKp42/44 signaling

To evaluate the effect of the QHREDGS peptide on keratinocytes, we first focused on normal

neonatal human epidermal keratinocytes (HEKs) cultured with the soluble QHREDGS peptide at

doses previously reported to be effective for endothelial cell survival (Low: 100 µM; High: 650

Page 88: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

64

µM) [10]. The percentage of proliferating HEKs in the population was not affected by the

presence of the QHREDGS peptide at either concentration as quantified by bromodeoxyuridine

(BrdU) incorporation (Fig. 4-1A). No significant difference in the HEK migration rate was

observed with the soluble QHREDGS peptide at either concentration (Fig. 4-2).

To investigate the effect of the soluble QHREDGS peptide on keratinocyte survival under

oxidative stress, we pre-conditioned HEKs by incubating the cells with or without the

QHREDGS peptide and then exposed the HEKs to 500 µM H2O2 for 2 h (Fig. 4-1B). An

endpoint cell integrity assay showed a significant dose-dependent increase in the percentage of

viable HEKs in the presence of supplemented QHREDGS (Fig. 4-1C).

Given that the full-length protein from which the QHREDGS peptide was derived, angiopoietin-

1, is known to protect skin cells from oxidative damage and to increase the activation of the

prosurvival Akt and MAPKp42/44 pathway[435], we investigated whether improved survival upon

H2O2 stress in the presence of the QHREDGS peptide was associated with the upregulation of

Akt and MAPKp42/44 phosphorylation. HEKs treated with 500 µM H2O2 showed transient

phosphorylation of both Akt (Fig. 4-1D) and MAPKp42/44 (Fig. 4-1E) at 15 min by Western blot

analysis. Indeed, the presence of the soluble QHREDGS peptide during pre-conditioning and

H2O2 treatment increased the phosphorylation of Akt and MAPKp42/44and the increase was dose-

dependent (Fig. 4-1D and E).

Page 89: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

65

Figure 4-1 Soluble QHREDGS peptide prevents H2O2-induced cell death in human primary keratinocytes

with up-regulation of Akt and MAPK phosphorylation. (A) Kct-positive HEKs cultured in the presence or

absence of different concentrations of soluble QHREDGS peptide did not incorporate significantly different

amounts of BrdU, indicating similar proliferation rates in all three condition (Low: 100 μM; high: 650 μM). Scale

bar = 50 μm. n=3-4. (B) Hydrogen peroxide treatment regimen for HEKs in the presence or absence of QHREDGS

peptide. HEKs were allowed to attach for 2 h and serum-starved overnight. For the peptide groups, HEKs were pre-

conditioned with QHREDGS peptide for 2 h and then treated with 500 μM hydrogen peroxide in the presence or

absence of the peptide. (C) HEK survival after hydrogen peroxide treatment was determined by the EarlyTox Cell

Integrity assay. The QHREDGS peptide protected HEKs against H2O2-induced cell death in a dose-dependent

manner (scale bar = 200 μm). One representative experiment is shown of n=3 independent experiments with 9

replicates for each condition in one experiment. (D and E) Immunoblotting with phosphorylated Akt or MAPKp42/44

Page 90: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

66

and Akt or MAPK p42/44 antibodies showed transient activation of Akt and MAPK p42/44 pathways signaling under

H2O2 stress. GAPDH was used to ensure even loading. The presence of QHREDGS peptide in the culture medium

up-regulated Akt and MAPK p42/44 phosphorylation. n=4 independent experiments and each experiment performed in

duplicates or triplicates. Data presented as mean ± SD. * indicates P < 0.05.

Figure 4-2 The presence of soluble QHREDGS peptide does not accelerate HEKs migration on collagen

coated surfaces. (A) Representative images of HEKs on collagen-coated substrates in EpiLife basal medium in the

presence or absence of soluble QHREDGS peptide at the different times indicated. Scale bar = 200 μm. (B) Image

analysis showed no difference in HEK migration over 24 h among the three groups. One representative experiment

is shown of n=3 independent experiments with at least four replicates for each condition in one experiment. Data

presented as mean ± SD.

4.3.2 Immobilized QHREDGS peptide promotes human primary

keratinocytes attachment, survival and migration in vitro

While we observed increased survival without excessive proliferation in the presence of soluble

QHREDGS, we did not observe enhanced keratinocyte migration (Fig. 4-2). This motivated our

further optimization of the method by which we presented the peptide to the cells. Given that the

Page 91: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

67

QHREDGS peptide is reported to primarily function through integrin interactions [10, 11] and

there is an ever-growing body of literature showing the increased efficacy of integrin ligands

when immobilized to a matrix [436, 437], we covalently immobilized the QHREDGS peptide to

a chitosan-collagen hydrogel. Chitosan and collagen interact through a combination of thermal

and ionic mechanisms, stabilized by polyanion (collagen) and polycation (chitosan) electrostatic

interactions.[438] Conjugation of the QHREDGS peptide to chitosan was achieved using

previously described methods [88] and chitosan-collagen films with or without immobilized

QHREDGS peptide were cast in the wells of 24-well or 96-well plates. Quantification using

fluorescently labelled peptide, FITC-QHREDGS, demonstrated effective immobilization in both

Low (4.7 ± 0.1 nmol/cm2) and High (13.8 ± 1.4 nmol/cm2) peptide concentrations and absence of

the peptide in the Ctrl condition (Fig. 4-3A). Normalized to the mass of chitosan in the films, the

amount of immobilized QHREDGS peptide was 14.9 ± 0.3 nmol/mg in the Low condition and

44.1 ± 4.6 nmol/mg in the High condition.

There was no significant difference in the attachment of HEKs to the various chitosan-collagen

films (Fig. 4-3B). However, in the settings of chitosan-only films wherein adhesion was poor,

the QHREDGS peptide clearly promoted HEK attachment in a dose-dependent manner (Fig. 4-4).

This indicates that while the QHREDGS peptide can promote HEK attachment, the presence of

collagen adhesion sites in the setting of the chitosan-collagen film masks this effect. Furthermore,

Western blot analysis showed the increased activation of Akt and MAPKp42/44 during 2 h

attachment on chitosan-collagen films in the presence of immobilized QHREDGS peptide (Fig.

4-3C).

We then investigated the effect of the immobilized QHREDGS peptide on HEK survival

following 500 µM H2O2 treatment. HEKs were allowed to attach to the chitosan-collagen films

for 4 h, then treated with H2O2 for 2 h. Subsequent cell integrity assessment showed an increased

percentage of viable HEKs in the presence of the immobilized QHREDGS peptide (Fig. 4-3D).

HEKs treated with 500 µM H2O2 and non-treated controls were also compared using Western

blot analysis, wherein phosphorylation of MAPKp42/44 was increased in the presence of

immobilized QHREDGS peptide relative to the control (Fig. 4-3E).

Page 92: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

68

Figure 4-3 Immobilized QHREDGS peptide in chitosan-collagen films promotes human neonatal primary

keratinocytes survival and migration. (A) Quantification of the amount of QHREDGS peptide immobilized

within chitosan-collagen films. n=3. (B) HEK attachment on the chitosan-collagen films in the presence or absence

Page 93: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

69

of conjugated QHREDGS peptide. Image analysis showed no difference in the number of attached HEKs (stained

with DAPI) among the three groups (scale bar = 200 μm). n=3 independent experiments and each experiment

performed in triplicates. (C) Immunoblotting with anti-phosphorylated Akt or MAPK p42/44 and anti-Akt or

MAPKp42/44 showed up-regulation of Akt and MAPK p42/44 activation during HEK attachment. GAPDH was used to

ensure even loading. n=3 independent experiments and each experiment performed in duplicates. (D) HEK survival

after hydrogen peroxide treatment was determined by the EarlyTox Cell Integrity assay. QHREDGS peptide in the

chitosan-collagen film protected HEKs against H2O2-induced cell death in a dose-dependent manner. One

representative experiment is shown of n=3 independent experiments with four replicates for each condition in one

experiment. (E) Immunoblotting with phosphorylated MAPK p42/44 and MAPK p42/44 antibodies showed up-regulation

of the MAPK p42/44 activation in HEKs under H2O2 stress at 15 min. GAPDH was used to ensure even loading. n=3

and each experiment performed in duplicates. (F) Representative examples of HEK wounding experiments on

chitosan-collagen films in the presence or absence of conjugated QHREDGS peptide (scale bar = 200 μm).

Confluent HEK monolayers were wounded (time 0) and maintained for 24 h in EpiLife basal medium with 0.12 mM

Ca2+. The wounds were outlined and the area at the various time points was normalized to the initial wound size

(time 0). HEK migration on the QHREDGS-immobilized films was accelerated compared to the control peptide-free

films. One representative experiment is shown of n=3 independent experiments with six replicates for each condition

in one experiment. Data presented in mean ± SD. * indicates P < 0.05.

Figure 4-4 The presence of the immobilized QHREDGS peptide promotes HEK attachment on chitosan-only

films. (A) Representative images of HEKs on chitosan-only films in the presence or absence of immobilized

QHREDGS peptide. Scale bar = 200 μm. Cell nuclei are shown in blue (DAPI). (B) Image analysis showed an

increased number of attached HEKs in the presence of the immobilized QHREDGS peptide in a dose-dependent

manner. The number of attached HEKs was normalized to the number that attached to tissue culture polystyrene

(TCP) in the same experiment. One representative experiment is shown of n=3 independent experiments with three

replicates for each condition in one experiment. Data presented as mean ± SD. * indicates P < 0.05.

Keratinocyte migration is essential for wound healing as a wound cannot heal in the absence of

re-epithelialization [439]. We therefore assessed the effect of the immobilized QHREDGS

peptide on HEK migration in 2D monolayers using an Ibidi migration assay system. Importantly,

the Ca2+ concentration in the culture medium was increased from 0.06 mM to 0.12 mM upon

initiation of the migration assay to ensure collective HEK migration (essential for wound

Page 94: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

70

healing), as demonstrated by the formation of E-cadherin mediated cell-cell junctions (Fig. 4-

5A). The presence of the immobilized QHREDGS peptide accelerated collective HEK migration

in a dose-dependent manner (Fig. 4-3F). The accelerated migration was not due to increased

proliferation as there was no difference in cell density among the three groups as characterized at

the migration endpoint (Fig. 4-5B).

Figure 4-5 HEKs form calcium-induced adherens junctions during migration and the accelerated migration is

not associated with a difference in cell density. (A) Representative images of HEKs on Ctrl substrates (chitosan-

collagen films without conjugated QHREDGS peptide) in EpiLife basal medium at different times as indicated,

following an increase in calcium from 0.06 mM to 0.12 mM. Adherens junctions (green = E-cadherin) were

established as early as 2 h. Scale bar = 50 μm. Cell nuclei are shown in blue (DAPI). (B) HEK cell density

characterized by DAPI counterstaining at the end of migration (24 h). There was no difference in the number of

HEKs on the chitosan-collagen films in the presence or absence of the immobilized QHREDGS peptide. n=3 and

each experiment performed in triplicates. Data presented as mean ± SD.

Page 95: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

71

4.3.3 Immobilized QHREDGS peptide promotes diabetic human primary

keratinocytes attachment, survival and migration in vitro

In diabetic chronic wounds, keratinocytes experience hyperglycemia and supra-physiological

oxidative stress, which challenges keratinocyte’s proliferation and survival [253, 440]. Therefore,

we examined the effect of the immobilized QHREDGS peptide on adult diabetic human

epidermal keratinocytes (DHEKs) by seeding DHEKs onto chitosan-collagen films in the

presence or absence of immobilized QHREDGS peptide. Similar to the results with normal HEK

cells, we found the presence of the immobilized QHREDGS peptide promoted DHEK

attachment to chitosan-only films (Fig. 4-7) but did not affect DHEK attachment to chitosan-

collagen films (Fig. 4-6A).Western blot analysis showed that the activation of Akt was increased

in DHEKs in the presence of the QHREDGS peptide during a 2 h attachment (Fig. 4-6B).

Because of prolonged inflammation, diabetic wounds experience a higher level of oxidative

stress compared to the normal wounds [440]. To mimic this scenario, we investigated the effect

of immobilized QHREDGS peptide on DHEK survival following a 2 h treatment with 2 mM

H2O2 (4-times higher exposure than used for the HEK survival assay), after allowing DHEKs to

attach to the chitosan-collagen films for 4h. Cell integrity assessment showed that DHEK

survival under H2O2 stress was improved in the presence of immobilized QHREDGS peptide

(Fig. 4-6C), despite the higher H2O2 concentration used. DHEKs treated with 2 mM H2O2 and

non-treated controls were also compared by Western blot analysis (Fig. 4-6D) and

phosphorylation of Akt and MAPKp42/44 upon H2O2 treatment was increased in the presence of

immobilized QHREDGS peptide in a dose-dependent manner (Fig. 4-6E).

We then assessed the effect of immobilized QHREDGS peptide on DHEK migration using the

Ibidi migration assay. Importantly, the Ca2+ concentration in KGM medium (0.1 mM) was

sufficient to ensure collective DHEK migration as demonstrated by the formation of E-cadherin

mediated cell-cell junctions without additionally elevating the Ca2+ concentration (Fig. 4-8A).

The presence of the immobilized QHREDGS peptide also accelerated DHEK collective

migration (Fig. 4-6F) and this was not due to cell density differences as characterized at the

migration endpoint (Fig. 4-8B).

Page 96: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

72

Figure 4-6 Immobilized QHREDGS peptide in chitosan-collagen films promotes diabetic adult human

primary keratinocyte survival and migration. (A) DHEK attachment on the chitosan-collagen films in the

presence or absence of immobilized QHREDGS peptide. Image analysis showed no difference in the number of

Page 97: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

73

attached DHEKs (stained with TO-PRO) among the three groups (scale bar = 200 μm). n=3 independent

experiments and each experiment performed in triplicate. (B) Immunoblotting with anti-phosphorylated Akt and

anti-Akt antibodies showed up-regulation of Akt activation during DHEK attachment. GAPDH was used to ensure

even loading. n=3 independent experiments and each experiment performed in duplicates. (C) DHEK survival

following hydrogen peroxide treatment was determined by the EarlyTox Cell Integrity assay. QHREDGS peptide in

the chitosan-collagen film protected DHEKs against H2O2-induced cell death in a dose-dependent manner. n=4-6

and each experiment performed with at least six replicates for each condition. (D) Representative immunoblots of

phosphorylated Akt or MAPKp42/44 and total Akt or MAPK p42/44. (E) Quantification of immunoblotting revealed up-

regulation of the Akt and MAPK p42/44 activation of DHEKs under H2O2 stress. GAPDH was used to ensure even

loading. n=3 independent experiments and each experiment performed in duplicates. (F) Representative examples of

DHEK wounding experiments on chitosan-collagen films with or without immobilized QHREDGS peptide (scale

bar = 200 μm). Confluent HEKs were stained with CellMask Green, wounded at time 0 and maintained for 6 h in

KBM basal medium. The wounds were outlined and the area at the indicated times were normalized to the initial

wound size (time 0). DHEK migration on the films with QHREDGS peptide was accelerated compared to the

peptide-free control. n=3 independent experiments and each experiment performed with at least four replicates for

each condition. Data presented as mean ± SD. * indicates P < 0.05.

Figure 4-7 The presence of immobilized QHREDGS peptide promotes DHEK attachment on chitosan-only

films. (A) Representative images of DHEKs on chitosan-collagen films in the presence or absence of immobilized

QHREDGS peptide. Scale bar = 200 μm. Cell nuclei are shown in blue (DAPI). (B) Image analysis showed an

increased number of attached DHEKs in the presence of immobilized QHREDGS peptide. The number of attached

DHEKs was normalized to the number that attached to tissue culture polystyrene (TCP) in the same experiment. One

representative experiment is shown of n=3 independent experiments with three replicates for each condition in one

experiment. Data presented as mean ± SD. * indicates P < 0.05.

Page 98: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

74

Figure 4-8 DHEKs form adherens junctions during migration and the accelerated migration is not associated

with a difference in cell density. (A) Representative images of DHEKs in KGM basal medium (0.1 mM Ca2+) on

chitosan-collagen films in the presence or absence of QHREDGS peptide at the end of migration (6h). Adherens

junctions (green = E-cadherin) were present in all three groups. Scale bar = 50 μm. Cell nuclei are shown in blue

(DAPI). (B) DHEK cell density characterized by DAPI counterstaining at the end of migration (6 h). There was no

difference in the number of DHEKs on chitosan-collagen films in the presence or absence of QHREDGS peptide.

n=3 and each experiment performed with four replicates. Data presented in mean ± SD.

4.3.4 QHREDGS-immobilized hydrogel promotes wound healing in

db/db diabetic mice

We investigated whether the QHREDGS peptide immobilized to the chitosan-collagen hydrogel

could accelerate wound healing in diabetic mice. This hydrogel system was chosen as a delivery

vehicle because of its rapid gelation under physiological conditions and its persistence for a

period of 3 weeks in vivo [13]. In vivo biocompatibility was also demonstrated in previous

myocardial infarction model studies [13]. Therefore, only one application of the hydrogel onto

the wounds was needed for the 2 week study. A full-thickness excision wound (Fig. 4-9A) was

created on eight weeks old, male BKS.Cg-Dock7m +/+ Leprdb/J mice (db/db). This model was

Page 99: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

75

selected because the animal is leptin receptor deficient and represents a type II diabetes model

characterized by hyperglycemia, obesity, hyperinsulinemia, and impaired wound healing.

Moreover, this strain heals wounds primarily by granulation tissue formation rather than by

contraction [441].

Quantification using fluorescently labelled peptide demonstrated that in reconstituted chitosan

solutions, the amount of conjugated QHREDGS peptide was 17.5 ± 2.2 nmol/mg(chitosan) in Low

conditions and 41.5 ± 1.4 nmol/mg(chitosan) in High conditions. This was converted to a peptide

concentration in the final chitosan-collagen hydrogel of 43.8 ± 4.4 μM in Low conditions and

103.8 ± 3.5 μM in High conditions. With a view to future clinical translation, we only tested the

Low condition in the in vivo studies to minimize the amount of peptide applied to the wound. A

single application of 50 μL Low chitosan-collagen hydrogel (2.2 nmol immobilized QHREDGS

peptide; Peptide) was applied to the wound. The chitosan-collagen hydrogel alone without the

peptide (Ctrl) and a no hydrogel/no peptide (Blank) were used as controls. A secondary dressing,

Tegaderm™ film, was applied on top of the wound with or without the hydrogel, to maintain a

moist environment. As shown in Fig 4-9B, the presence of immobilized QHREDGS in the

hydrogel resulted in significantly smaller wounds on day 14 compared to the controls. Image

analysis of the wound gross morphology performed by an investigator blinded to the study

groups demonstrated faster wound healing in the Peptide group starting on day 8. Administration

of the chitosan-collagen hydrogel without the immobilized QHREDGS peptide (Ctrl) had no

significant effect on the wound closure rate compared to the Blank controls.

We also examined the wound histology by Masson’s trichrome staining and confirmed the

location of the epithelial tongue using pan-keratin staining (Fig. 4-9C). The wound edge was

defined as the distance between the two boundaries of intact skin (thin musculature of the

panniculus carnosus).There was no significant difference amongst the three groups in the wound

edge distance, indicating no difference in wound contraction (Fig. 4-9D i). The epithelial gap

was defined as the distance between the two advancing epithelial tongues (Fig. 4-9C i-vi) and

the epithelial gap in the Peptide group was significantly smaller than in the Blank and Ctrl

groups (Fig. 4-9D ii, Fig. 4-11). The re-epithelialization percentage was defined as the ratio of

the distance that has been re-epithelialized over the wound edge distance, and the re-

epithelialization percentage was higher in the presence of the QHREDGS peptide compared to

Page 100: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

76

the controls (Fig. 4-9D iii, Fig. S6). The Peptide group also developed significantly more

granulation tissue (Fig. 4-9D iv) compared to the controls. Moreover, the epidermal thickness of

the advancing epithelial tongue was significantly smaller in the Peptide group than in the Blank

and Ctrl groups (Fig. 4-9D v), which indicates more effective epidermal cell migration. There

was no difference in the epithelial thickness of the skin remote from the wounds among the three

experimental groups (Fig. 4-10).

Page 101: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

77

Figure 4-9 QHREDGS-immobilized hydrogel promotes wound healing in db/db diabetic mice. (A)

Representative images of the 8-mm full-thickness dorsal wounds on db/db diabetic mice. (B) Representative gross

images of the initial wounds on day 0 (D0) and the wounds at 14 days (D14) after treatment with no hydrogel

Page 102: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

78

(blank), ctrl chitosan-collagen hydrogel (ctrl), and QHREDGS peptide conjugated chitosan-collagen hydrogel

(peptide). Quantification of the wound size as a percentage of the original wound area revealed faster wound closure

in the peptide-treated mice at day 8-14. n=4. (C) Representative images of Trichrome stained tissue sections of

wounds treated with blank, control chitosan-collagen hydrogel, and peptide conjugated chitosan-collagen hydrogel

on day 14. Black arrows indicate wound edges; red arrows indicate the tips of the healing epithelial tongue. The tips

of the healing epithelial tongue were confirmed by pan-keratin staining as shown in the insets. Inset scale bar = 50

μm. (D) Quantification of wound size from histological samples collected 14 days after treatment. (i) Image analysis

showed no significant difference among the three groups in the wound edge distance. (ii) The peptide treatment

significantly reduced the size of epithelial gap, indicating accelerated wound closure compared with the blank and

control groups. (iii) The peptide treatment significantly increased the re-epithelialization percentage at the end of

experiment compared with the blank and control groups. (iv) The peptide treatment significantly increased the size

of the granulation tissue compared to the blank and control groups. (v) Average thickness of the epidermis within

300 μm of the leading edge of the wound. The epidermal thickness in the peptide-treated group was lower than the

blank and control groups. n=4. Data presented as mean ± SD. * indicates P < 0.05.

Figure 4-10 Thickness of the unwounded epidermis. (A) Representative images of Trichrome stained tissue

sections of unwounded epidermis in Blank, Ctrl and Peptide groups. Scale bar = 50 μm. (B) Image analysis revealed

no significant difference in unwounded epidermal thickness among the three groups.

Figure 4-11 An example of wound re-epithelialized after two weeks with a single treatment of QHREDGS

peptide in the chitosan-collagen hydrogel. Black arrows indicate wound edges; red arrows indicate tips of the

healing epithelium tongue. Re-epithelialization was quantified at 92%.

Page 103: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

79

4.3.5 Accelerated QHREDGS-induced diabetic wound healing does not involve changes in the extent of angiogenesis of the granulation tissue

Figure 4-12 The improvements in the diabetic wound healing process induced by the QHREDGS peptide are

not associated with increased angiogenesis within the granulation tissue. (A) Representative images of CD31-

stained tissue sections of wounds treated with no hydrogel (blank), control chitosan-collagen hydrogel (ctrl), and

QHREDGS peptide conjugated chitosan-collagen hydrogel (peptide) on day 14. Scale bar = 300 μm. (B)

Representative images of smooth muscle actin (SMA)-stained tissue sections from blank, control, and peptide

groups on day 14. Scale bar = 300 μm. (C) Image analysis showed no significant difference among the three groups

in (i) vessel density, (ii) CD31-positive area percentage, and (iii) SMA-positive area percentage. n=4. Data presented

as mean ± SD. * indicates P < 0.05.

To further characterize the granulation tissue, we compared the density of microvessels and

contracting myofibroblasts in the three experimental groups by immunohistochemistry with

antibodies against CD31 (Fig. 4-12A) and α-smooth muscle actin (α-SMA) (Fig. 4-12B). There

was no difference in the vessel density and CD31 positive area percentage among the three

groups based on measurements obtained from six random locations within the granulation tissue

(Fig. 4-12C i and ii). This was further confirmed by an automated algorithm analysis of the

Page 104: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

80

entire granulation tissue that showed no significant difference amongst the three groups in terms

of vessel density, CD31 positive percentage, lumen area, vascular area, vessel area, vessel

perimeter, or vessel wall thickness (Fig. 4-13). There was also no significant difference in the

density of myofibroblasts amongst the three groups as determined by α-SMA staining (a

common, albeit non-specific, marker of myofibroblasts) (Fig. 4-12C iii). Myofibroblasts are

considered to be the main contributors to wound contraction [442] and the absence of a

difference in myofibroblast cell density is consistent with the histological results showing no

difference in the wound edge size amongst the groups (Fig. 4-9D i). Taken together, the

accelerated wound healing in the presence of the conjugated QHREDGS peptide cannot be

attributed to changes in granulation tissue blood vessel density or to myofibroblasts, although

there was more granulation tissue overall in the Peptide group (Fig. 4-9D iv).

Page 105: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

81

Figure 4-13 QHREDGS peptide does not affect microvessel number and size within granulation tissue. Micro-vessel analysis of (A) CD31 positive area percentage and microvessel density, (B) lumen area, (C) vascular

area, (D) vessel area, (E) vessel perimeter, and (F) vessel wall thickness within the entire granulation tissue. Data

presented as mean ± SD. n=4.

Page 106: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

82

4.4 Discussion

Angiogenesis has been a primary target of therapeutic interventions in wound healing by local

application of growth factors (e.g. epidermal growth factor [443, 444], fibroblast growth factor

[445, 446], vascular endothelial growth factor [447, 448], and angiopoietin [449]) or angiogenic

cells [450, 451]. However, diabetic patients are often reported to have dysfunctional endothelium,

which cannot respond efficiently to growth factor stimulation [452, 453]. Diabetics also suffer

from the insufficient recruitment of circulating endothelial progenitor cells (EPCs) critical for

wound repair [240, 241]. As a result, many of these approaches failed to meet the FDA-accepted

primary efficacy endpoint standard of “complete wound closure within 12 weeks for any dose”

[454].

We chose to take a different approach here, by considering the hallmarks of true regenerative

healing as observed during the closure of embryonic wounds and scarless healing in model

organisms such as flies, zebrafish and newts. In all of these models, rapid, coordinated and

collective migration of epidermal cells was critical for regenerative healing [429, 430]. Hence,

we focused on developing a hydrogel treatment that would promote the collective migration of

keratinocytes and enhanced granulation tissue formation. Different mechanisms have been

proposed to explain the cell-cell interactions in collective cell migration, including local tractions

pulling cooperatively towards unfilled space (termed “kenotaxis”) [455], mechanical exclusion

interactions between cells [456], and intercellular adhesion and tension [457]. In our in vitro

studies, calcium concentration was increased when necessary to ensure that both normal and

diabetic keratinocytes migrated collectively rather than individually so that the treatment effect

on collective migration could be accurately described.

Following its discovery [458], there has been a growing recognition that angiopoietin-1 is an

important regulator of cellular processes including vascular protection [459], cardiac remodeling

[460-462], inflammation [463-465], and wound healing [449]. Due to the insolubility of

angiopoietin-1, its derivatives have been developed to promote wound healing by angiogenesis

[466-468]. Here, we assessed the effect of a novel water-soluble peptide, QHREDGS, derived

from the fibrinogen-like domain of angiopoietin-1 and conserved amongst species [460], on

promoting normal and diabetic keratinocyte survival and collective migration. In this study,

soluble QHREDGS peptide supplemented in the culture medium protected human keratinocytes

Page 107: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

83

against H2O2 stress and induced up-regulation of Akt and MAPKp42/44 phosphorylation. These

findings are consistent with previous reports for angiopoietin-1 in human keratinocytes and

melanocytes [435]. Our previous studies have also reported that a scrambled peptide

(DGQESHR or DQSHGER) does not exert prosurvival effects similar to the QHREDGS peptide

in cardiac cells [7, 469], endothelial cells [10], and iPSCs [11] motivating the omission of the

scrambled peptide in the studies described here.

Importantly, the QHREDGS peptide functions through interactions with 1- containing integrins

that are involved in cell-matrix interactions, rather than Tie2 receptors that reside mainly on

endothelial cells [10, 11, 13]. This enables the QHREDGS peptide to act on various cell types

including keratinocytes, which express the 31 integrin implicated in their enhanced survival

and migration [470-472]. The integrin interaction also motivated the presentation of the

QHREDGS peptide as a matrix bound ligand. Furthermore, the short peptide sequence

QHREDGS can be modified using versatile chemical methods and does not require a specific

orientation or conformation to function, as does the full length protein [473]; and peptides are

less susceptible to degradation due to proteolysis or hydrolysis during modification and after

delivery to the native environment compared to full length proteins [2].

Here, we used EDC chemistry to conjugate the QHREDGS peptide to the backbone of chitosan,

which is a zero-length cross-linker that does not add any other moieties to the final product,

features important for further clinical translation. Peptide immobilization also provides the

advantages of localized action in the target tissue and a lower total dose requirement over

application in an encapsulated or soluble form. We confirmed not only that the prosurvival effect

of the QHREDGS peptide was preserved after conjugating and immobilizing it in the chitosan-

collagen films, but that the immobilized QHREDGS peptide was able to promote keratinocyte

collective migration whereas the soluble peptide did not. Notably, in the context of the chitosan-

collagen film system, the conjugated QHREDGS peptide is presented in close proximity to

collagen, thereby creating an environment reminiscent of the collagen-glycosaminoglycan

interaction in the native ECM [474].

Using DHEKs from an adult diabetic patient we determined that the prosurvival, promigratory

effects of immobilized QHREDGS peptide could be translated to diabetic keratinocytes. Diabetic

Page 108: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

84

chronic wounds often occur to elderly population (the source of the primary DHEKs in this study)

due to age-associated comorbidities [475]; and advanced protein glycosylation has been

associated with diabetes and aging that can alter the functional properties of important ECM

components such as collagen [243]. Despite these challenges, the immobilized QHREDGS

peptide was effective in the in vitro diabetic wound healing model, which suggested the

possibility of in vivo benefits to diabetic wound healing.

Although our previous in vitro study showed that QHREDGS peptide promoted endothelial cell

survival, metabolism, and tube formation mediated by integrin interactions [10], the accelerated

diabetic wound healing shown in the animal studies here was not associated with enhanced

angiogenesis per area of the granulation tissue. Since there was more granulation tissue in the

Peptide group, consequently higher total number of blood vessels was achieved in the wound

with the Peptide hydrogel treatment, although blood vessel density was the same amongst the

groups. This is consistent with our previous study in a rat myocardial infarction (MI) model,

which showed an increase in large vessels within the MI border zone but no difference in the

total vascularization [13]. Alternatively, the diabetic endothelium is associated with enhanced

degradation of nitric oxide, an important regulator of inflammation, angiogenesis, and re-

epithelialization, due to the presence of excessive ROS [242]; and in our previous study it was

shown that the QHREDGS peptide can induce enhanced endothelial cell nitric oxide production

[10]. Hence, it is possible the improved wound healing effects induced by the QHREDGS

peptide could be attributable in part to nitric oxide production.

Our previous study demonstrated the retention of the chitosan-collagen hydrogel over two weeks

post-injection in the infarcted rat heart, an extremely dynamic, inflammatory environment [13].

In this study, we applied the QHREDGS peptide in the same chitosan-collagen hydrogel used as

a single treatment and found it to be sufficient to accelerate wound healing in a clinically

relevant, genetically modified db/db diabetic mouse model at two weeks. The ability to induce an

effective wound healing response with a single application is an important clinical consideration

as it removes the need for frequent repeated applications that can disturb the healing process, it

reduces the pain and discomfort associated with frequent dressing changes, and it requires fewer

healthcare resources [475, 476]. We also selected the use of a single mouse model for the

analysis of therapeutic efficacy, which can be seen as a potential limitation of our study.

Page 109: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

85

However, the db/db diabetic mice are used most often as a model of human diabetic chronic

wound [477] and the genetically modified db/db diabetic mouse has been used previously to

support clinical trial development with other molecules [478].

To tease out the extent to which the QHREDGS peptide could induce wound healing, we

selected a larger 8-mm diameter initial wound size rather than the commonly used 6-mm wounds

[282, 362]. We also selected the chitosan-collagen hydrogel with the low concentration of

QHREDGS peptide for our in vivo study based on the concept that pharmaceutical agents should

be applied at a minimal effective dosage to avoid potential side effects. For example, an

increased rate of mortality secondary to malignancy was reported for patients treated with > 45 g

of Regranex® (180 nmol PDGF-BB) [479]. Here a single dose of 4.4 nmol peptide/cm2 of wound

was found to be an effective dose to promote granulation tissue formation and wound closure in

vivo, whereas daily application of 0.4 nmol PDGF-BB (Regranex®)/cm2 of wound was found to

promote granulation tissue formation but did not shorten the time to wound closure in a db/db

mouse model [480].

The QHREDGS peptide is available by cost-effective synthesis with a precisely defined

composition, offering an additional advantage to potential clinical applications. The cost of the

synthetic QHREDGS peptide used in our study was $1.6CAD/mg, which is approximately 20

000-times cheaper than the commercially available rhPDGF-BB, an active component of the

FDA approved Regranex®, which costs $33,000CAD/mg (Fisher Scientific). The choice of the

hydrogel components, collagen and chitosan, has also been motivated by clinical translation

considerations. Chitosan has been approved by the FDA for use in humans in topical applications

(e.g. HemCon® bandages). Similarly, collagen is a component of numerous wound dressings

currently on the market in different formulations such as freeze-dried sheet, pastes, pads,

powders, and gels (e.g. INTEGRA™ Matrix Wound Dressing, BIOSTEP Collagen Matrix, BGC

Matrix®, Stimulen™ Collagen).

Here, we have demonstrated that keratinocyte survival and collective migration represent

promising alternative therapeutic targets for diabetic chronic wounds that support re-

epithelialization, a hallmark of wound regeneration and closure. In a genetically modified

diabetic mouse, the immobilized QHREDGS peptide accelerated the wound healing process by

promoting re-epithelialization rate and granulation tissue formation, without significantly

Page 110: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

86

affecting angiogenesis. Moreover, the immobilized QHREDGS peptide did not increase the

number of -SMA positive cells, e.g. myofibroblasts, that can induce the antithetic wound

regeneration processes of wound contraction, collagen overproduction, and scar formation [481].

On the basis of our reported results, a number of questions arise. The efficacy of a potential

therapeutic intervention providing synergistic regulation of angiogenesis from angiogenic growth

factors (e.g. VEGF and bFGF) and re-epithelialization from QHREDGS peptide in diabetic

chronic wounds is yet unknown. Previously, we have successfully immobilized angiogenic

growth factors (VEGF and bFGF) on collagen scaffold and observed promoted angiogenesis in

vivo [88]. Notably, the chitosan-collagen hydrogel system is capable of delivering other

molecules at the same time as the chitosan backbone is amenable to various chemical

modification [482, 483].

In conclusion, the QHREDGS peptide promoted keratinocyte adhesion and collective migration

in vitro, as well as survival against H2O2 stress through Akt and MAPKp42/44 pathways. In vivo,

the QHREDGS peptide immobilized to a chitosan-collagen hydrogel accelerated diabetic wound

healing by enhanced re-epithelialization and granulation tissue formation. Together, our data on

both normal and diabetic human primary keratinocytes and in a db/db diabetic mouse model

demonstrate the translational relevance of the QHREDGS peptide in treating diabetic wounds.

We propose the QHREDGS peptide as a therapeutic candidate for promoting diabetic wound

healing.

4.5 Conclusion

In conclusion, the QHREDGS peptide promoted keratinocytes adhesion, survival against H2O2

stress, and collective migration in vitro involving up-regulation of Akt and MAPKp42/44 pathways.

When immobilized in a chitosan-collagen system, the QHREDGS peptide accelerated diabetic

wound healing in a manner not dependent on angiogenesis or wound contraction. Together, our

data on human primary keratinocytes, both normal and diabetic, and in a db/db diabetic mouse

model demonstrate clinical relevance and we propose QHREDGS peptide as a therapeutic

candidate for promoting diabetic wound healing.

Page 111: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

87

4.6 Acknowledgments

We thank L.A. Reis for training on the chitosan-collagen hydrogel preparation. We thank L.E.

Fitzpatrick and M.V. Sefton for input on animal study and useful discussion. This work is funded

by the NSERC Steacie Fellowship to M.R., Canadian Institutes of Health Research (CIHR)

Operating Grants (MOP-126027 and MOP-137107), NSERC Discovery Grant (RGPIN 326982-

10), and National Institutes of Health Grant 2R01 HL076485. The authors declare that they have

no competing interests. All data and materials are available.

Page 112: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

88

Chapter 5

5 Microfabricated perfusable cardiac biowire: a platform that mimics native cardiac bundle7

5.1 Introduction

Cardiovascular diseases are important targets for pharmacological therapy because they are

associated with high morbidity and mortality rates[484]. In vitro engineered models may serve as

cost-effective alternatives to animal models due to improved system control and higher

throughput. In recent years, tissue engineering methods have been significantly advanced to

generate functional three-dimensional (3D) cardiac tissues in vitro[485-487], which better

recapitulate the complexity and electro-mechanical function of native myocardium compared to

conventional in vitro systems of single cell suspensions or monolayers. Moreover, with the

opportunities brought about by human pluripotent stem cells (hPSC), tissue engineering methods

hold a great promise in developing patient-specific medical treatment[488].

In native myocardium, cardiomyocytes are highly anisotropic, usually in a length of 80-100 μm

and 20-30 μm in diameter[489]. Each cardiomyocyte adjoins neighboring cardiomyocytes by

specialized intracellular junctions, such as gap junctions and desmosomes, to form a complex 3D

network, or syncytium. On tissue level, native cardiomyocytes are organized into spatially well-

defined cardiac bundles with supporting vasculature. This highly organized architecture is

critical for electro-mechanical activation, propagation of electrical signals, and global cardiac

function[490]. Cardiac tissues generated by current tissue engineering methods often poorly

recapitulate this architecture.

Increased cardiovascular risk is one of the major unwanted side effects of new drug candidates,

which frequently leads to usage restriction or even withdrawal from the market[491]. Cardiac

7 Copyright © 2014 Royal Society of Chemistry. Contents of this chapter have been published in Lab Chip: Xiao Y,

Zhang B, Liu H, Miklas JW, Gagliardi M, Pahnke A, Thavandiran N, Sun Y, Simmons C, Keller G, Radisic M.

Microfabricated perfusable cardiac biowire: a platform that mimics native cardiac bundle. Lab Chip. 2014; 14:869–

82. Reuse with permission from Royal Society of Chemistry. A link to the published paper can be found at:

http://pubs.rsc.org/en/content/articlelanding/2014/lc/c3lc51123e

Page 113: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

89

toxicity was the main reason behind withdrawal of numerous drugs from the market, including

well known examples such as Vioxx or Avanida, accounting for up to 20% of all drug

withdrawals[492, 493]. Thus, it is essential to identify these risks at an early stage in drug

development process to define safety profile and avoid cost escalation. Despite the exceptional

progress in developing cardiac disease models with hPSC (Timothy[494], long QT[495],

LEOPARD syndrome[496] and dilated cardiomyopathy patients[497]), most studies still use

cardiac monolayers that do not capture architectural complexity of the native cardiac niche. After

pharmacologic agents are administrated into human body, they are circulated through the

vasculature and delivered to the myocardium by the blood in capillaries. Current in vitro drug

testing systems, however, expose the cardiac cells to the pharmacologic agents directly from the

culture media in conventional well plates[411, 498-500]. Thus, developing in vitro cardiac

systems that can recapitulate the perfusion scenario could provide improved physiological

relevance when assessing pharmacological effects on cardiac tissue in vitro.

We recently described a development of a human cardiac micro-tissue, termed biological wire,

which captures some of the architectural complexity of the native myocardium and enables

maturation of cardiomyocytes derived from human pluripotent stem cells with the application of

electrical stimulation[413]. However, these original biowires lacked perfusion, a critical aspect

for mimicking native physiology and mass transfer. We describe here technological

developments required to create a perfusable biowire conducive to electrical field stimulation

and we prove the feasibility of drug testing in this system.

Perfusable cardiac biowires were generated using a polytetrafluoroethylene (PTFE) tubing

template in microfabricated bioreactors, which provided contact guidance for cells to align and

elongate. To demonstrate the feasibility of this platform for drug testing, we supplied nitric oxide

(NO) in the cell culture channel to provide biochemical stimulation to cardiomyocytes within the

biowire. NO was released from perfused sodium nitroprusside (SNP) solution and passed

through the tubing wall to reach the tissue constructs with cardiomyocytes. This bioreactor was

also integrated with electrical stimulation to further improve phenotype of cardiomyocytes.

Page 114: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

90

5.2 Materials and methods

5.2.1 Biowire bioreactor design and fabrication

The biowire bioreactor consisted of two main units, the microfabricated platform made of

poly(dimethysiloxane) (PDMS) and the suspended template made of either silk 6-0 suture or

PTFE micro-tubing. To fabricate the PDMS platform, standard soft lithography technique was

used to make to a two-layer SU-8 (Microchem Corp., Newton, MA) master[501]. The first layer

included the template channel and the cell culture chamber, while second layer included only the

cell culture chamber. Then PDMS was cast onto the SU-8 master and baked for 2 hr at 70 °C. A

biowire template was then anchored to the two ends of the PDMS platform followed by the

bioreactor sterilization in 70% ethanol and overnight UV irradiation.

5.2.2 Perfusion system design and fabrication

In order to provide perfusion through the tubing template, two microfabricated modules, drug

reservoir and connecting channel, were added to the biowire bioreactor. Both modules were

fabricated by first molding PDMS with a single-layer SU-8 master (length × width × height =

10 × 1 × 0.3 mm). The drug reservoir was created by cutting through the PDMS using an 8 mm

biopsy punch (Sklar). The biowire bioreactor channel was connected to the drug reservoir and

connecting channel with the PTFE tubing (inner diameter (ID) = 0.002 inch, outer diameter (OD)

= 0.006 inch, Zeus). Tygon tubing (ID = 0.01 inch, OD = 0.03 inch, Thomas Scientific)

connected the perfusion system to external negative pressure generated by a peristaltic pump.

The perfusion rate was characterized by the liquid volume collected at the outlet from the

peristaltic pump (n = 3). All the connecting points were secured by epoxy glue and three

microfabricated modules were plasma bonded to a glass slide.

5.2.3 Cell culture

Neonatal rat cardiomyocytes were obtained from 2-day old neonatal Sprague-Dawley rats as

described previously[9] and according to a protocol approved by the University of Toronto

Committee on Animal Care. The culture media contained 10% (v/v) fetal bovine serum, 1% (4-

(2-hydroxyethyl)-1-piperazineethanesulfonic acid) (HEPES), 100 U/ml penicillin-streptomycin,

1% Glutamine, and the remainder Dulbecco`s modified Eagle`s medium.

Page 115: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

91

Cardiac differentiation in embryoid bodies (EBs) of HES-2 human embryonic stem cell (hESC)

line was performed as described previously[388, 502]. Briefly, EBs were first cultured in

StemPro-34 (Invitrogen) media containing BMP4 (1 ng/ml). On day 1, they were transferred to

the induction medium (StemPro-34, basic fibroblast growth factor (bFGF; 2.5 ng/ml), activin A

(6 ng/ml) and BMP4 (10 ng/ml)). On day 4, the EBs were removed from the induction medium

and re-cultured in StemPro-34 supplemented with vascular endothelial growth factor (VEGF; 10

ng/ml) and Inhibitor of Wnt production-2 (IWP2; 2 μM). On day 8, the medium was changed

again and the EBs were cultured in StemPro-34 containing VEGF (20 ng/ml) and bFGF (10

ng/ml) for the remainder of EB culture as well as for the biowire culture. EBs were maintained in

hypoxic environment (5% CO2, 5% O2) for the first 12 days and then transferred into a 5% CO2

for the remainder of the culture period. EBs were dissociated for seeding in biowires at day 23

(EBd23).

5.2.4 Generation of cardiac biowires

Cardiac cells (from neonatal rat isolation or hESC differentiation) were first suspended at 200

million /ml (unless specified otherwise) in Collagen Type I based gel (2.5 mg/ml of rat tail

collagen type I (BD Biosciences) neutralized by 1N NaOH and 10× M199 media as described by

the manufacturer) with the supplements of 4.5 μg/ml glucose, 1% (v/v) HEPES, 10% (v/v)

Matrigel (BD Biosciences), and 2 μg/ml NaHCO3. Suspended cardiac cells were then seeded

into the cell culture channel (3 μl per biowire). After 30 min incubation at 37 °C to induce the

gelation, appropriate media were added. Cardiac biowires were kept in culture for up to 14 days

with media change every 2-3 days.

Cardiac biowires starting with different cell densities (100 and 200 million/ml) were seeded to

study the effect of the cell seeding density. Collagen-based gel was seeded into the cell culture

channel without loading cardiac cells, to serve as a cell-free control. Ultra-long cardiac biowires

were generated with customized biowire bioreactor that was 5 cm long fabricated in a similar

manner as described above and seeded with neonatal rat cardiomyocytes.

Page 116: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

92

5.2.5 Quantification of compaction rate

After seeding, brightfield images of the biowires were taken every day (n = 3 per group) using an

optical microscope (Olympus CKX41) and the diameters of the biowires at five distinct locations

were averaged with image analysis.

5.2.6 Immunostaining and Fluorescent Microscopy

Biowires were fixed with 4% paraformaldehyde, permeablized by 0.25% Triton X-100, and

blocked by 10% bovine serum albumin (BSA). Immunostaining was performed using the

following antibodies: mouse anti-cardiac Troponin T (cTnT) (Abcam; 1:100), rabbit anti-

Connexin 43 (Cx-43) (Abcam; 1:200), mouse anti-α-actinin (Abcam; 1:200), goat anti-mouse-

Alexa Fluor 488 (Jackson Immuno Research; 1:400), anti-rabbit-TRITC (Invitrogen; 1:200),

anti-mouse-TRITC (Jackson Immuno Research; 1:200). Nuclei were counterstained with 4',6-

diamidino-2-phenylindole (DAPI) (Biotium; 1:100). Phalloidin-Alexa 660 (Introgen; 1:600) was

used to stain F-actin fibers. For confocal microscopy, the stained cardiac biowires were

visualized under an inverted confocal microscope (Olympus IX81) or an upright confocal

microscope (Zeiss LSM 510).

5.2.7 Quantification of nuclei elongation and alignment

Cell nuclei within the biowires were visualized by DAPI staining and z-stack images were

obtained by confocal microscopy with 3 μm interval. Each stack of the confocal images was

analyzed in ImageJ 1.45s (National Institutes of Health, USA) with an automated algorithm

described by Xu et al[503] with approximately 1000 nuclei analyzed per sample. Nuclei

elongations were characterized as nucleus aspect ratios, the ratio of long axis over short axis of

the nuclei, and nuclear alignment was characterized by orientation angles. In the control

monolayer group, orientation of the nuclei was characterized compared to an arbitrarily defined

orientation, while in the biowire group, the orientation of the suture templates was set as

reference.

5.2.8 Characterization of perfusable biowires

Neonatal rat cardiac cells were seeded into the perfusable biowire reactors with tubing template.

After cultivation for 7 days, the cardiac biowires were sectioned and visualized under

Page 117: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

93

environmental SEM (Hitachi S-3400 N). The biowires were imaged under variable pressure

mode at 70 Pa and 15 kV and the chamber temperature was −20°C.

To visualize the cross-section, perfusable cardiac biowires were stained with cTnT antibody and

then TRITC. Stained biowires were then cryo-sectioned into 500 μm thick sections using a

cryostat (Leica CM3050S) and mounted to Superfrost Plus glass slide (VWR). Images of the

cross-sectioned biowires were acquired by Olympus fluorescent microscope (Olympus IX81).

To demonstrate the feasibility of the perfusable biowire bioreactor, FITC-labeled polystyrene

beads (Spherotech Inc.) were added into the drug reservoir and perfused through the rat cardiac

biowire, while it beat spontaneously on day 8. Bright-field and fluorescent videos and images

were acquired with a fluorescence microscope (Olympus IX81).

5.2.9 Quantification of NO perfusion

Sodium nitroprusside (SNP) (Sigma) was dissolved in distilled water to make 200 mM SNP

solution and then added to the drug reservoir. Perfusion through the tubing template was driven

by the external peristaltic pump. Once the SNP solution perfused through the tubing, the

peristaltic pump was stopped and the entire perfusion system was kept in cell culture incubator.

NO amount in the cell culture channel (outside the PTFE tubing) was quantified with a

fluorometric Nitric Oxide Assay Kit (Calbiochem, 482655). In brief, samples collected from the

cell culture channels (8 μl, n = 3) at different time points (0.5 hr, 6 hr, and 24 hr) were converted

to nitrite by nitrate reductase and then developed into a fluorescent compound 1-H-

naphthotriazole. The fluorescent signals were quantified by a plate reader (Apollo LB 911,

Berthold Technologies) and compared to the nitrate standard.

5.2.10 NO treatment of human cardiac biowires

On day 7, the NO treatment of human cardiac biowire was initiated by perfusing the 200 mM

SNP solution and the peristaltic pump was stopped once the SNP solution was perfused through

the tubing. The beating activities of the human cardiac biowires were recorded at 16.67

frames/second before treatment and 24 hr post-treatment by Olympus IX81 while the biowires

were kept at 37°C. The beating activities of the human cardiac biowires were quantified by the

image analysis method described by Sage et al[504]. In brief, the movements of one spot at the

Page 118: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

94

same location on the human cardiac biowire before and after the NO treatment were

characterized.

5.2.11 Electrical stimulation

Different electrical stimulation conditions were applied to the rat cardiac biowires as described

previously [485]. The parallel stimulation chambers were fitted with two 1/4-inch-diameter

carbon rods (Ladd Research Industries) placed 2 cm apart, perpendicular to the biowires (such

that the electrical field was parallel with the biowire long axis), and connected to a stimulator

(S88X, Grass) with platinum wires (Ladd Research Industries). The perpendicular stimulation

chambers were built with two carbon rods 1 cm apart placed parallel with the biowires (i.e. the

filed was perpendicular to the long axis of the biowire). The biowires were pre-cultured for 4

days until the biowire structures were established and their spontaneous beating was

synchronized, and then subjected to the electrical field stimulation (biphasic, rectangular, 1 ms

duration, 1.2 Hz, 3.5-4 V/cm) for 4 days with 10 μM ascorbic acid supplemented in the culture

media while control biowires were cultured without electrical stimulation. At the end of

electrical stimulation, the rat cardiac biowires were double stained for cTnT with Alexa 488

conjugated antibody and Cx-43 with TRITC conjugated antibody, or their mechanical properties

were measured by atomic force microscopy (AFM). Confocal images were acquired using

identical microscope settings for all groups. Areas stained positive for cTnT staining (green

pixels) or Cx-43 staining (red pixels) were quantified by ImageJ using the identical thresholding

parameters in all groups.

For the human perfusable cardiac biowire, only parallel electrical stimulation was applied as

described above. Starting on day 4, electrical field stimulations (biphasic, rectangular, 1 ms

duration, 1 Hz, 3.5-4 V/cm) were applied for 4 days while control biowires were cultured

without electrical stimulation. Both stimulated and control biowires were perfused with culture

medium at a flow rate of 2 μl/min within the PTFE tubing driven by an external syringe pump

(PHD Ultra; Harvard Apparatus). At the end of electrical stimulation, the electrical properties of

the stimulated and control human cardiac biowires were characterized in terms of excitation

threshold (ET) and maximum capture rate (MCR) under external field pacing as previously

described [505].

Page 119: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

95

5.2.12 Atomic force microscopy (AFM)

After application of electrical stimulations for 4 days, rat cardiac biowires were tested using a

commercial AFM (Bioscope Catalyst; Bruker) mounted on an inverted optical microscope

(Nikon Eclipse-Ti). The force-indentation measurements were done with a spherical tip (radius =

5-10 μm) at nine distinct spots to evenly cover the center of the cardiac biowires with 5 nN

trigger force at 1 Hz indentation rate. The cantilever (MLCT-D, Bruker) had a nominal spring

constant of 0.03 N/m. Hertz model was applied to the force curves to estimate the Young's

modulus and detailed data analysis was described elsewhere[506]. All AFM measurements were

done in fluid environment at room temperature.

5.2.13 Statistical analysis

Statistical analysis was performed using SigmaPlot 11.0. Differences between experimental

groups were analyzed using t-test or one-way ANOVA with significant difference considered as

P < 0.05.

Page 120: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

96

5.3 Results

5.3.1 Generation and characterization of cardiac biowires

Figure 5-1 Cardiac bundles in native myocardium. (a) Schematic illustration of the structure of cardiac bundles

in native myocardium. Cardiomyocytes are elongated, aligned, and grouped into bundles around capillaries. (b)

Tangential section of adult rat myocardium with CD31 staining (brown). Nuclei were counterstained as light violet

(long arrows). The blood vessels were noted as asterisks while the capillaries were noted as short arrows. Scale bar =

200 μm. (c) Fluorescent image of tangential cryosection of neonatal rat myocardium. Cardiac troponin T (cTnT) was

stained against Alexa 488-labeled (green) antibody, showing its unique striation structure. Cell nuclei were

counterstained with DAPI (blue) and the long arrows indicate elongated nuclei. Scale bar = 20 μm.

The native myocardium has a highly anisotropic structure (Figure 5-1a) with a high density of

capillaries (Figure 5-1b) and surrounding elongated and aligned cells (Figure 5-1c). In the

native heart, extracellular matrix (ECM) serves as a template for cells to align and elongate[98].

Page 121: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

97

Figure 5-2 Generation of cardiac biowires with microfabricated bioreactor. (a) Within 7 days of cultivation,

neonatal rat cardiomyocytes (8.75 million cells/ml) remodeled the gel and compacted around the suture template to

form the biowire structure. Scale bar = 200 μm. (b) Device design of the microfabricated bioreactor with suspended

6-0 suture template. (c) A customized ultra-long cardiac biowire in the length of 5 cm (scale bar = 500 μm). (d)

Quantification of gel compaction and its dependence on initial seeding density of cardiomyocytes (mean ± SD, n =

3). With no cardiomyocytes seeded (gel only), the gel did not compact and form biowire structure. Biowires with

higher seeding density (200 million cells/ml) compacted faster than those with lower seeding density (100 million

cells/ml) during the remodelling.

Page 122: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

98

In addition, a structural correlation between directionality of capillaries and cardiomyocytes can

be readily observed[489]. We aimed to emulate this in our biowire bioreactor by introducing a

perfusable tubing template and by using a hydrogel, for cell seeding, consisting of ECM

molecules normally present in the native heart. Primary neonatal rat cardiomyocytes were used

to generate 3D, self-assembled cardiac biowires by seeding within type I collagen-based gel into

microfabricated PDMS platforms with suspended templates (Figure 5-2b). Seeded cells

remodeled and contracted the collagen gel matrix around the templates within a week (Figure 5-

2a, Figure 5-4a). The gel compaction only occurred with the presence of the seeded cells, as

cell-free gels did not compact or degrade during the culture time, and the compaction rate

positively correlated with the cell seeding density (Figure 5-2d). Cardiac biowires of different

dimensions could be generated by customizing the dimensions of the biowire bioreactor. Here,

we generated biowires as long as 5 cm (Figure 5-2c). Generation of longer biowires might be

possible; however it was not explored in this work.

Image analysis of the cell nuclei that counterstained with DAPI (Figure 5-3a, left) revealed

nuclei elongation and alignment along the axis of suture template. There was a significant

difference (p < 0.001) of nuclei aspect ratio between biowires and monolayer group (Figure 5-

3b). Compared to neonatal rat cardiomyocytes cultured in monolayer controls, biowires had a

lower frequency of cells in the smaller aspect ratio range and a higher frequency of cells in the

larger aspect ratio range (Figure 5-3c). Image analysis also revealed that cell nuclei in biowires

were oriented along with the axis of the suture template, while those in monolayers were

randomly distributed (Figure 5-3d).

Neonatal rat cardiac biowires started to beat spontaneously between 3 and 4 days post-seeding

and kept beating during gel compaction, demonstrating that the biowire bioreactor allowed for

electromechanical coupling of the cells within the hydrogel matrix. The spontaneous beating of

biowires with higher seeding density (200 million/ml) started earlier and was more prominent

than in those with lower seeding density (100 million/ml), which is thought to be a result of the

presence of more cardiomyocytes and better coupling. Immunohistochemistry staining showed

that the rat cardiac biowires expressed the sarcomeric protein, cTnT (Figure 5-3a, right).

Page 123: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

99

Figure 5-3 The suture template provides topographical cues in the biowires for the cardiomyocytes to

elongate and align. (a) Confocal images of the biowire with nuclei counterstained with DAPI and cardiac

Troponin-T (cTnT) stained with Alexa 488 (green). Scale bar = 100 μm. (b) Nuclei aspect ratios (~1000 nuclei

characterized per sample) of cardiac cells cultured as monolayer vs. seeded in the biowires plotted in box plot

showing the 1st quartile, median, and 3rd quartile with a significant difference between two groups (***, p < 0.001).

(c) Histogram showing the distribution of nuclei aspect ratios of biowire group and monolayer group (n = 3 per

group). There were significantly higher frequencies in the lower aspect ratio range in monolayer group (*, p < 0.05)

and higher frequencies in higher aspect ratio range in biowire group (#, p < 0.05). (d) Characterization of nucleus

orientation reveals random distribution of nuclei in the monolayer group (random direction as 0 ̊) and oriented

distribution of nuclei along with the suture template in the biowire group (orientation of suture template as 0 ̊).

Dashed lines indicate orientation angle, solid hemi-circular lines indicate the percentage gridlines, and grey

(monolayer) or blue (biowire) lines indicate the actual percentage of the nuclei in the specific orientation angle.

Page 124: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

100

5.3.2 Generation and characterization of perfusable cardiac biowires

Figure 5-4 Generation of perfusable cardiac biowires. (a) Neonatal rat cardiomyocytes (200 million cells/ml)

remodeled the gel and compacted around the tubing template (ID = 50.8 μm, OD = 152.4 μm). Scale bar = 200 μm.

A close-up view showing the tubing lumen at the end of the biowire is given at top-right. Scale bar = 100 μm. (b)

SEM images demonstrate that the cardiac tissue attached to the tubing surface and formed a uniform-thick layer

Page 125: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

101

after remodelling. (c) Representative phase contrast image (left) and confocal image (right) showing the circular

morphology of the cross section of the perfusable cardiac biowire with the expression of cardiac Troponin-T (cTnT).

Scale bar = 150 μm. (d) Set-up of the perfusion device. Two microfabricated modules were added to the bioreactor

system: a drug reservoir at one end of the biowire and a channel at the other end for connection to an external

negative pressure source. The entire biowire perfusion system was bonded on a glass slide (real-life image shown at

the top-left corner). (e) The tubing-templated biowire was perfused with FITC-labeled polystyrene beads (1 μm in

diameter). Dash lines illustrate the wall of the cell culture channel. FITC-labeled beads were indicated by arrows.

Asterisks indicate the auto fluorescence from the cardiomyocytes within the cardiac biowire. This image was over-

exposed to better visualize the fluorescent beads. Scale bar = 100 μm.

Primary neonatal rat and hESC-derived cardiomyocytes were used to generate perfusable cardiac

biowires with PTFE tubing template Figure 5-4d). Both cell types were able to form the cardiac

biowires and beat spontaneously (Figure 4a). As shown in SEM images, cells attached to the

smooth surface of the PTFE tubing after self-remodeling (Figure 5-4b). Cross sections of these

perfusable biowires showed that self-remodeled cells encircled the tubing template and expressed

cTnT (Figure 5-4c).

The feasibility of perfusable biowire bioreactor was demonstrated by perfusion with FITC-

labeled fluorescent beads. Perfusion rate driven by the peristaltic pump was quantified to be 2 ±

0.16 μL/min (n = 3). Bright field video showed both spontaneous beating activity of the rat

cardiac biowire and the perfusion of the fluorescent beads. The movement of the beads was

better visualized under fluorescent view. A snapshot of the video (Figure 5-4e) was overexposed

to provide better visualization of the fluorescent beads. The cardiac biowire was also visible in

this image due to the auto-fluorescence of cardiomyocytes.

5.3.3 NO treatment of human cardiac biowires by perfusion

To demonstrate feasibility of drug testing in the perfusable cardiac biowire, a pharmacological

agent, NO donor SNP, was applied to the culture media that was perfused through the tubing

lumen. As NO was generated in the tubing lumen, it diffused through the tubing wall reaching

the cell culture outer channel where the total amount of NO was quantified. The amount of NO

released from 200 mM SNP was quantified by a fluorometric assay which validated the

persistence of the NO release from SNP solution over several hours (Figure 5-5a). The

cumulative NO amount in the cell culture channel was 100 μM (800 pmol in 8μl), which

exceeded the physiological levels of NO in vivo[507].

Upon gel compaction, the hESC-derived cardiomyocytes within the biowires started spontaneous

beating. After NO treatment for 24 hr, performed by perfusion of NO-donor SNP through the

Page 126: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

102

tubing lumen, the spontaneous beating of human cardiac biowires slowed down and this was

further characterized by image analysis (Figure 5-5b, c). In order to compare beating frequency

changes between different biowires, the frequencies after 24 hr NO treatment were normalized to

the basal level (before treatment). The beating frequencies after NO treatment were significantly

lower than the basal level (74±3%, n = 3) while the control biowires remained the same (100±9%,

n = 3).

Figure 5-5 Nitric oxide (NO) treatment on human tubing-templated biowires. (a) Quantification of NO amount

passing through the tubing wall after perfusing SNP (200 mM) for 0.5 hr, 6 hr, and 24 hr. (b) 24 hr NO treatment

significantly slowed down the beating of biowires compared to the basal levels while there was no significant

change in the non-treated biowires (n = 3 per group, p < 0.01). (c) Quantified by image analysis, the beating rate of a

biowire after 24 hr NO treatment was less frequent compared to the basal level. (d) Confocal images showing the

disrupted α-actinin (green) structure within the NO-treated biowire (right) compared to the control (left) (top: lower

magnification, scale bar = 50 μm; bottom: higher magnification, scale bar = 10 μm).

Page 127: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

103

The degradation of cytoskeleton of cardiomyocytes within the biowires based on hESC derived

cardiomyocytes caused by NO treatment through perfusion was characterized using confocal

microscopy for α-actinin labeled with Alexa 488 conjugated antibody (green) and actin labeled

with Alexa 660 conjugated antibody (far red) (Figure 5-5d). It was possible to clearly discern

the striated pattern of sarcomeric Z-discs labeled with α-actinin in the control biowires, while the

NO treated biowires showed an overall punctate pattern.

5.3.4 Electrical stimulation of cardiac biowires

To demonstrate the versatility of the cardiac biowire bioreactor, electrical stimulation was

applied to further improve the phenotype of cardiomyocytes (Figure 5-6a).

Immunohistochemical staining showed that the rat cardiomyocytes in the biowires that

underwent electrical stimulation with the field parallel to the biowire long axis had more cTnT

positive structures oriented along with the axis of the suture template (indicated by the dashed

line), while those in non-stimulated biowires were randomly distributed and those in

perpendicular-stimulated biowires were found to be perpendicular to the suture template (Figure

5-6b). Moreover, the cardiomyocytes in the parallel- and perpendicular- stimulated biowires

showed stronger expression of Cx-43, a marker for the gap junctions between adjacent

cardiomyocytes, compared to the control biowires, indicating better coupling between the

cardiomyocytes in the stimulated groups (Figure 5-6b). This was also confirmed by comparing

the ratio of Cx-43 positive area over cTnT positive area under same magnification with identical

microscope settings (Figure 5-6c).

When handling the rat cardiac biowires outside the bioreactor, it was noticed that the parallel-

stimulated biowires were stiffer than the non-stimulated control. This was further assessed by

AFM analysis (n = 3 per group), which revealed significantly (p = 0.009) higher apparent

Young’s modulus of parallel-stimulated biowires compared to non-stimulated controls (Figure

5-6d).

The perfusable human cardiac biowires that underwent medium perfusion through the tubing and

parallel electrical stimulation at the same time showed improved electrical properties compared

to the non-stimulated controls as assessed by ET and MCR under electrical field stimulation. The

ET is the minimum electrical field voltage required for inducing synchronous contractions and

Page 128: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

104

the decreased ET of the stimulated biowires (Figure 5-6e) indicated better electrical excitability.

The MCR is the maximum beating frequency attainable while maintaining synchronous

contractions and the increased MCR (Figure 5-6f) of the stimulated biowires indicated improved

cell alignment and interconnectivity.

Figure 5-6 Electrical stimulation and perfusion of cardiac biowires. (a) Experimental set-up of biowires under

different electrical stimulation conditions. Carbon rods (in black) connected to an external stimulator provided either

parallel or perpendicular electrical field stimulation on cardiac biowires for 4 days starting on Day 4. (b)

Representative confocal images of biowires after application of different electrical stimulation conditions (left:

lower magnification, scale bar = 50 μm; right: higher magnification, scale bar = 10 μm). Parallel-stimulated biowires

showed more cTnT positive (green) structures oriented along with the suture template (indicated by dashed lines). (c)

Higher ratio of Cx-43 positive area over cTnT positive area indicated stronger expression of Cx-43 in both parallel-

and perpendicular-stimulated biowires compared to the non-stimulated controls (n = 4 per group, p < 0.05). (d)

Page 129: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

105

Young’s modulus of cardiac biowires characterized by AFM reveals that the parallel-stimulated biowires had higher

apparent Young’s modulus compared with the control biowires (**, p < 0.01). (e) Electrically stimulated perfused

biowires based on hESC derived cardiomyocytes had lower excitation threshold compared to the non-stimulated

controls (***, p < 0.001). (f) The electrically stimulated perfused biowires based on hESC derived cardiomyocytes

had higher maximum capture rate compared to the non-stimulated controls (*, p < 0.05).

5.4 Discussion

The native myocardium consists of spatially well-defined cardiac bundles with supporting

vasculature (Figure 1a) and the cardiomyocytes within the cardiac bundles are highly anisotropic

(Figure 1b). In this study, we have developed a microfabricated bioreactor to generate cardiac

biowires in vitro recapitulating the structure and function of native cardiac bundles. To the best

of our knowledge, this is the first study to examine the drug effects on cardiomyocytes by

perfusion within cardiac bundle model, which better mimics native myocardium mass transfer

properties compared to other engineered heart tissues. This bioreactor provided topographical

cues for the cardiac cells to elongate and align, and was also integrated with other cues, e.g.

electrical stimulation.

Gel compaction has been widely applied in tissue engineering to create 3D microtissue

constructs for in vivo implantation[508] and in vitro models[411, 509]. Compared to scaffold-

based constructs, the self-assembled constructs from gel compaction produce increased force of

contraction due to the higher cell density after the compaction[409]. Moreover, there is

increasing interest in microtissue constructs made by gel compaction as microarrays for drug

testing because they provide much higher throughput than conventional models[411, 412, 509,

510]. In this study, type I collagen was chosen as the main gel matrix as it is one of the main

ECM components of native myocardium. We noted that previous in vitro collagen-based models

only stayed intact for several days due to their poor mechanical properties[509]. In our

microfabricated system, with the mechanical support provided by the suspended templates, the

cardiac biowires remained stable in the bioreactor for weeks. We were able to generate cardiac

tissues in larger scale (up to 5 cm long) compared to other in vitro models and the dimensions of

the cell culture channel could be easily customized, which could render additional control over

the morphology of the cardiac biowires. The cell culture channels were initially designed to be

300 μm in height considering the limitations for oxygen and nutrient supply[511]. Moreover, the

presence of the templates enabled easy disassembly of the biowire from the bioreactor and facile

handling of the cardiac biowires at the end of cultivation for further characterization.

Page 130: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

106

Our microfabricated bioreactor was also able to generate cardiac biowires that are 5 cm long,

which is comparable to the height of the human heart. The feasibility of handling individual

cardiac biowire together with the ability to create macro-scale biowires raise up the prospect of

investigating the alignment of multiple cardiac biowires by bundling or weaving them together to

generate thicker structures, using similar methods as described by Onoe et al [512]. To

characterize the force generated by the cardiac biowires or cardiac biowire bundles, degradable

sutures could be used to generate template-free cardiac biowires which will be a topic of our

future studies.

To validate our microfabricated bioreactor, neonatal rat cardiomyocytes were used in preliminary

studies. Only when seeded at higher cell density (> 5×107 cells/ml), which is comparable to the

cell density in native rat myocardium (~108 cells/ml)[180]. the cardiac biowires started

spontaneous beating on day 3-4. The template provided contact guidance for the cells to elongate

and align along with, recapitulating the anisotropic properties of cardiomyocytes in the native

myocardium. The image analysis was done on cell nuclei due to the difficulty of defining cell

membranes within 3D tissue. However, nuclear alignment is a sufficient indication of cell

alignment and also one of the hallmarks of native myocardium (Figure 5-1c).

To further develop the biowire system, we used PTFE tubing as the template instead of the 6-0

silk suture. The commercially available PTFE tubing was chosen because it is biocompatible

(USP Class VI), extremely non-absorbent (ideal for drug testing), and micro-scale in dimension

(ID = 50 μm, OD = 150 μm), on the order of post-capillary venules in size[513]. Due to the small

size of the inner lumen, we used negative pressure to drive the perfusion instead of positive

pressure. Two microfabricated modules were added to the system to enable long-term perfusion

and incubation of the biowire system. Indicated by the shortening of biowire during self-

remodeling, the cell attachment on PTFE tubing was not as strong as that on silk suture, mainly

because of the smoothness of the PTFE tubing surface (Figure 5-4b). However, the cell-gel

composite was still able to assemble itself around the tubing with a circular cross-section.

In this study, NO was chosen as a model drug because of following reasons: (1) NO is produced

by endothelial cells in native myocardium, and then transported in the radial direction to

cardiomyocytes [514], the scenario we were trying to recapitulate in our biowire bioreactor; (2)

NO plays a critical role in regulating myocardial function, through both vascular-dependent and -

Page 131: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

107

independent effects[514]; (3) there is increasing evidence showing that NO is directly implicated

in cardiomyocyte disease development and prevention, such as in ischemia-reperfusion

injury[515]; (4) NO is a small gas molecular, which can readily pass the tubing wall. SNP was

chosen as the NO donor because it is a common NO donor used as vasodilator to treat pulmonary

hypertension and low cardiac output [516, 517]. Moreover, SNP aqueous solution was reported

to release NO at a constant rate over several hours in vitro[518].

For the NO treatment testing, we generated human cardiac biowires from hESC-derived

cardiomyocytes. The human cardiac biowire started spontaneous beating as early as day 1 and

the beating was synchronized within 7 days. After 24 hr of NO treatment, the beating frequencies

of the human cardiac biowires significantly slowed down compared to their basal level. This

result corresponds with the vasodilator effect of NO in vivo[519] and might be caused by

degradation of myofibrillar cytoskeleton, which has been seen by Chiusa et al[520]. However,

NO shows bi-polar inotropic effect at lower concentrations with diverse intracellular mechanisms

and there were discrepancies between studies due to the lack of standardization for in vitro

models[514]. Therefore our microfabricated bioreactor could serve as a novel platform to

uncover the effects of NO on cardiomyocytes at the tissue level.

To demonstrate the versatility of our biowire bioreactors, electrical stimulation was integrated

with the system as it has been reported to improve the phenotypes of cardiomyocytes[485, 501].

Because the cells in the cardiac biowires were anisotropic, we studied both parallel- and

perpendicular- field electrical stimulations on the rat biowires. The higher tissue stiffness under

parallel electrical stimulation, which was closer to isolated neonatal rat cardiac tissue (6.8 ± 2.8

kPa)[521], were attributed to more organized cellular contractile apparatus as characterized by

immunohistochemical staining. The perfusable human cardiac biowires were electrically

stimulated and perfused at the same time and this brings the prospect to study the interaction

between electrical stimulation and pharmaceutical agents delivered in a physiological manner. A

more detailed study on electrical stimulation alone of biowires based on human pluripotent

cardiomyocytes has been done in our group and indicated that electrical stimulation of

progressive frequency increase markedly improved the maturation of hPSC-derived

cardiomyocytes in terms of myofibril structure and electrical properties[413].

Page 132: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

108

Medium perfusion has been recognized to improve the viability and functionality of

cardiomyocytes within cardiac constructs in vitro since perfusion significantly improves oxygen

and nutrient supply[522]. In most of previous studies, bioreactors provided medium perfusion by

sandwiching cell-laden porous scaffold, while exposing the cardiomyocytes directly to the

flow[522-524]. This does not exactly recapitulate the native myocardium where the blood supply

flows through a dense vascular network that minimizes transport distances but also protects

cardiomyocytes from shear[525]. More recently, bioreactors were developed to provide the

electrical stimulation and medium perfusion simultaneously and it was shown that perfusion and

stimulation had a synergistic effect on improving the contractile functionality of the cardiac

constructs[525, 526]. However, the cardiac constructs in these systems were based on porous

scaffolds and therefore unable to provide the information about the effect of electrical

stimulation on anisotropic cardiac tissue.

Previous studies describe the design of perfusion bioreactors that enable high-throughput in vitro

drug testing on cardiac constructs[524, 527]. Kaneko et al designed a microchamber array chip to

evaluate single cell level interactions for drug testing[527]. Agarwal et al designed a bioreactor

composed of a microarray of cantilevers that was able to characterize diastolic and systolic

stresses generated by anisotropic cardiac microtissue in real-time and the bioreactor could

provide electrical stimulation on these cardiac microtissues[524]. These two studies

characterized cardiac function on either single cell or monolayer level, which might be

insufficient to provide accurate information of cardiac disease as in our complex native system.

Moreover, the drugs investigated in these studies were directly applied to the cells, instead to the

blood compartment, and the presence of flow generated shear stress on cardiomyocytes, both of

which contributed to the generation of an unphysiological environment compared to that

cardiomyocytes experience in the native heart.

There are several advantages of our microfabricated cardiac biowire bioreactors: (1) they are

better mimic of the native cardiac bundle structure with anisotropic alignment; (2) the presence

of the template enables easier handling for later characterization and keeps the entire structure

stable for weeks; (3) the device could be easily customized and applicable for high-throughput

drug screening; (4) the platform provides topographical stimulation by itself; (5) the platform is

versatile and could be integrated with other stimuli as well (e.g. mechanical stimulation); (6) the

Page 133: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

109

perfusable cardiac biowire system is the first platform to study pharmacological agents applied to

cardiomyocytes by perfusion through cardiac bundle mimic and could provide valuable

knowledge on cardiac disease development and therapeutics.

While this perfusable cardiac biowire platform provides us many opportunities, there are still

some limitations of our current platform. The permeability of the commercially available PTFE

tubing renders limitation on the drug candidates that can be tested, as only small molecules can

diffuse appreciably through the tubing wall and proteins cannot. Ideal tubing materials should be

microporous for better permeability. Further studies are required to investigate other relevant

pharmacological agents and seeding endothelial cells in the tubing lumen to study the interaction

between endothelial cells and cardiomyocytes could also be pursued.

5.5 Conclusion

In conclusion, we have developed a microfabricated cardiac biowire bioreactor that is capable of

testing pharmacological agents applied by perfusion through the lumen. The bioreactor provides

topographical cues for the cardiomyocytes to assemble and align and it could be integrated with

other stimuli to further improve the phenotypes of the cardiomyocytes. The engineered cardiac

biowires could serve as in vitro models that recapitulate the structure and function of the in vivo

cardiac bundles for studies of cardiac development and disease.

5.6 Acknowledgments

The authors would like to thank Zheng Gong for his kind help with preparing Figure 3d. This

work was funded by grants from Ontario Research Fund–Global Leadership Round 2 (ORF-

GL2), Natural Sciences and Engineering Research Council of Canada (NSERC) Strategic Grant

(STPGP 381002-09), Canadian Institutes of Health Research (CIHR) Operating Grant (MOP-

126027), NSERC-CIHR Collaborative Health Research, Grant (CHRPJ 385981-10), NSERC

Discovery Grant (RGPIN 326982-10), NSERC Discovery Accelerator Supplement (RGPAS

396125-10), National Institutes of Health grant 2R01 HL076485 and Heart and Stroke

Foundation grant (T6946).

Page 134: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

110

Chapter 6

6 Discussion and conclusions

6.1 Discussion

The work reported in this thesis was motivated by the need for an instructive matrix designed

with biochemical and biophysical cues to promote and regulate tissue morphogenesis in situ. The

focus of tissue engineering has evolved from replacing damaged tissues or organs with functional

tissues substitutes generated in vitro into creating an instructive microenvironment to regenerate

the impaired tissue in situ. Compared with generating tissues in vitro, where biochemical cues

can be precisely controlled in term of concentration and treatment time by changing culture

media, instructive matrix design for tissue generation in vivo is challenged by difficult control

over the presentation of biochemical and biophysical cues. To facilitate optimal tissue

morphogenesis, the instructive matrix design heavily relies on our understanding of cell-ECM

interactions and the ability to properly present different cues, both biochemical and biophysical,

to regulate cellular functions. This thesis aims to contribute to this scientific endeavor with a

particular focus on designing instructive matrix with immobilized biochemical cues and

microfabricated topographical guidance.

With ever-growing evidence showing the importance of growth factor regulation in tissue

regeneration, effective delivery of growth factors and growth-factor-derived peptides from

matrix represents an important approach to promote tissue regeneration. Conventional growth

factor delivery methods as soluble supplements are usually poorly controlled with a burst release

that may lead to severe systematic side effects [528]. Moreover, soluble growth factor delivery

often requires repetitive applications, which is not desirable in clinical applications. Covalent

immobilization brings the possibility of controlled release of the biochemical cues and their

prolonged presentation in the matrix. In this thesis, we presented evidence that single application

of collagen-based matrix with covalently immobilized growth factors or short sequence peptides

promoted tissue regeneration, with specific emphases on cardiac tissue engineering and wound

healing applications.

Page 135: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

111

In Chapter 3, angiogenic growth factors, VEGF and bFGF, and ang-1-derived peptide,

QHREDGS, were covalently immobilized onto collagen scaffolds using EDC chemistry. Our

results demonstrated efficient immobilization and prolonged release of both growth factors and

the peptide. Of note, the porous structure and the tensile strength of the collagen scaffolds were

not altered by the chemical modification. The porous structure of the collagen scaffold is

important for tissue regeneration in situ as it facilitates native cell infiltration and new blood

vessel formation. The mechanical properties, such as the tensile strength, are critical for tissue

engineering applications in a dynamic microenvironment, such as in the heart.

The next study [181] used the collagen scaffold immobilized with angiogenic growth factors to

rejuvenate MSCs from aged human patients. The modified collagen scaffold enhanced MSCs

proliferation in vitro and prolonged cell survival and improved angiogenesis to restore

ventricular morphology and function in vivo. Of note, the improvement was most prominent with

patches seeded with cells from old donors. This novel cytokine-conjugated, sustained-release

system provides a practical and promising platform for cardiac repair in elderly survivors of an

extensive myocardial infarction, an important advance in an increasingly aging society.

In Chapter 4, the pro-survival effect of QHREDGS peptide was demonstrated in human primary

keratinocytes for the first time. The QHREDGS peptide has been shown to promote cell survival

through integrin interaction and there is growing body of literature demonstrating improved

efficacy of integrin ligands presented as immobilized form rather than soluble supplements. This

motivated us to conjugate the peptide onto the backbone of chitosan and then form stable

systems (e.g. films and hydrogels) with collagen. When conjugated within chitosan-only coating,

the immobilized QHREDGS peptide promoted keratinocytes attachment, while the peptide

within chitosan-collagen films promoted cell survival against H2O2 stress, and collective

migration in vitro. We presented results from both normal and diabetic human keratinocytes

showing up-regulation of Akt and MAPKp42/44 phosphorylations.

We further assessed the efficacy of QHREDGS peptide in promoting wound healing in db/db

diabetic mice model that is clinically relevant. When immobilized in a chitosan-collagen

hydrogel system, the QHREDGS peptide accelerated diabetic wound healing in a manner that

was not directly associated with improved local angiogenesis. This study indicated that

QHREDGS peptide holds promise to augment the therapeutic outcomes of current diabetic

Page 136: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

112

chronic wound therapies, the majority of which focus on addressing impaired angiogenesis.

Together, our data on human primary keratinocytes, both normal and diabetic, and in a db/db

diabetic mouse model demonstrated clinical relevance and we propose QHREDGS peptide as a

therapeutic candidate for promoting diabetic wound healing.

Importantly, both the collagen patch immobilized growth factors and the chitosan-collagen

hydrogel immobilized QHREDGS peptide were applied only once at the beginning of both in

vivo studies. This highlights the contribution of covalent immobilization of biochemical cues in

instructive matrix design for tissue engineering applications where repetitive therapeutic

interventions are sometimes not accessible, such as in the heart. In general, single application is

preferable compared with repetitive applications because of less possibilities to disrupt the

regenerating microenvironment.

Another potential contribution of covalently immobilizing growth factors or peptides is to enable

dynamic presentation of the biochemical cues. The spatial and temporal presentation of different

stimuli is crucial during embryo development and has been extensively investigated and applied

in stem cell differentiations, which indicates their importance in tissue regeneration as well.

However, this was not explored in this thesis. Other studies presenting different biochemical cues

discriminately with spatiotemporal control have shown great promise in various tissue

engineering applications such as bone regeneration [529], new vessel formation [530, 531],

osteochondral tissue engineering [532, 533], muscle regeneration [534], and brain tissue

regeneration [535, 536].

The importance of biophysical cues in stem cell differentiation and tissue morphogenesis has

been increasingly recognized and different biophysical cues including matrix mechanical

properties and topographical guidance have been applied in instructive matrix design [89-91,

537-539]. These challenges in mechanisms and applications of topographical cues became

addressable with emerging material and analytical technologies that bring the possibility to

fabricate and characterize topographical cues with precise control up to nano-scale and increased

rigor and reproducibility [94]. These studies significantly contribute to our understanding of how

cells sense, adapt, and respond to their surrounding microenvironment, which in turn is central to

designing instructive matrix for tissue engineering.

Page 137: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

113

Moreover, empowered by the advances of microfabrication, a variety of functional micro-tissue

constructs were generated in vitro to recapitulate native tissues including lung [540], liver [541,

542], heart [410, 413, 487, 543], blood vessel [544], kidney [545, 546], intestine [547, 548], and

skin [549, 550]. These engineered micro-tissues and systems composed of multiple tissues [551,

552] have been proposed to be cogent platforms for pre-clinical drug screening studies with

better physiological relevance compared with animal models. The development of these systems

marked the emergence of the field of “Organ-on-a-Chip” or “Human-on-a-Chip” [553-555].

In Chapter 5, we developed a microfabricated cardiac biowire bioreactor that was capable of

testing pharmacological agents applied by perfusion through the lumen in the center of the

engineered cardiac micro-tissues. The bioreactor provided topographical cues for the cardiac

cells to assemble and align and it was integrated with electrical stimulation to further improve the

phenotypes of the cardiomyocytes. The engineered cardiac biowires could serve as in vitro

models that recapitulate the structure and function of the in vivo cardiac bundles for studies of

cardiac development and disease. The next study [413] utilized the original bioreactor with

suture template as platform to investigate the effect of electrical stimulation on maturation of

cardiomyocytes derived from human pluripotent stem cells.

Currently, topographical cues are mainly integrated in 2D substrates in vitro with few studies

demonstrating their promise in tissue regeneration in vivo [556]. Compared with biochemical

cues applied in vitro, the importance of biophysical cues is still underestimated due to the limited

feasibility to present different biophysical cues with temporal control. For example, different

protocols have been intensively investigated for stem cell differentiation induced by biochemical

cues as a result of effortless switching of biochemical cues by changing culture media during the

treatment regimen. Current technologies have enable presenting topographical cues with precise

control up to nano-scale [44], but not the flexibility of switching between different topographical

cues over time without disrupting the cell-matrix interaction yet. Therefore, the dynamic

presentation of topographical cues is a leading challenge yet to be overcome for instructive

matrix designs.

Page 138: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

114

6.2 Significant contributions

The body of work presented in this thesis addresses different strategies to regulate tissue

remodeling with biochemical or topographical cues. The first two studies were motivated by the

limited therapeutic outcome of existing treatments and the last study was motivated by the

limitation of existing drug-screening platforms at the time I started my study.

The QHREDGS peptide is a novel peptide derived from ang1 and has demonstrated pro-survival

effect on various cell types including cardiac cells, endothelial cells, iPSCs and osteoblasts in

different studies [7-13]. Here we showed covalent immobilization of QHREDGS peptide onto

collagen scaffold by EDC chemistry. Covalent immobilization significantly prolongs the

presence of the QHREDGS peptide in the matrix compared with delivery as a soluble

supplement. Moreover, the chemical modification did not alter the porous structure and

mechanical properties of the collagen scaffolds, which are important for their integration with the

native tissue. This is the first study to immobilize the QHREDGS peptide on collagen scaffold

and it might serve as good candidate for tissue engineering applications including cardiac

regeneration and wound healing.

MSCs derived from bone marrow serve as a good candidate for cell therapy because they can be

easily obtained from autologous tissue and be expanded in vitro rapidly. However, the use of

autologous MSCs is limited in the patients most in need, the elderly, because of their age-related

dysfunctions and impaired regeneration potential [4, 557]. Here, we designed a bioactive

collagen patch with covalently immobilized angiogenic growth factors (VEGF and bFGF) and

demonstrated its ability to rejuvenate MSCs from aged donors for the first time [181]. Seeded on

the collagen patches immobilized with angiogenic growth factors, the MSCs from aged donors

showed increased proliferation and decreased differentiation in vitro and exhibited improved cell

survival and cardiac functional regeneration in vivo after implantation. This study could

significantly contribute to current cell therapies for different tissue engineering applications and

implement their applications to the patients need them the most.

Current approaches to treat diabetic wound ulcers mainly focus on angiogenesis and render

limited therapeutic outcome. Here we presented a novel approach that will provide means to

recapitulate key aspects of scarless embryonic wound healing by 1) promoting effective

Page 139: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

115

keratinocyte migration, 2) protecting the wound-bed cells against oxidative stress and 3)

providing a new matrix for cell attachment. We used a chitosan-collagen hydrogel modified with

the QHREDGS peptide and demonstrated that the immobilized QHREDGS peptide promoted

cell survival under hydrogen peroxide stress, collective cell migration of both normal and

diabetic human primary keratinocytes. Importantly, application of a single treatment with a low

dose of QHREDGS-immobilized chitosan-collagen hydrogel accelerated wound closure and

granulation tissue formation in a type 2 diabetic mice model. Importantly, the QHREDGS

peptide can be synthesized at a cost that is 20000 times cheaper than human recombinant growth

factors that have been investigated in a number of previous studies. Together the findings in this

study propose the QHREDGS peptide as a therapeutic candidate for promoting diabetic wound

healing and augmenting current therapies.

Cardiovascular complications are the leading cause of drug withdrawal from the market and

there is a growing interest in developing physiologically relevant models with human cells to be

implemented in drug development [53]. Here we presented perfusable cardiac microtissues

generated in microfabricated bioreactors with suspended template providing topographical

guidance. This is the first platform that is able to test drug candidates by perfusion through the

center of cardiac microtissue and we validated its application by perfusing NO-releasing solution.

This work laid the foundation of the “bio-wire” system [413] and together with following

technologies our lab started TARA Biosystems Inc. to produce physiologically relevant mature

heart tissue that can be interrogated to measure physical and biological factors capable of

accurately predicting cardiotoxicity for pre-clinical applications.

Together, the novel matrix systems designed in this thesis regulate cell-matrix interactions by

providing different biochemical cues or topographical guidance. These matrix systems regulate

tissue regeneration and remodeling in applications including cardiac remodeling, wound healing,

and generating cardiac micro-tissues, and resulted in improved therapeutic outcomes in vivo or

physiological relevance in vitro. The findings from these studies would significantly contribute

to our knowledge of how to design and present biochemical and topographical cues to regulate

cell-matrix interaction and tissue remodeling both in vitro and in vivo.

Page 140: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

116

6.3 Conclusion

In conclusion, the studies in this thesis demonstrated that cell-matrix interactions can be

facilitated by topographical and biochemical cues provided by surrounding matrix, which

regulate cell assembly, cell functions, and tissue morphogenesis. Specifically, we demonstrated

the feasibility of these instructive cues in both tissue regeneration in vivo and creating complex

micro-tissues in vitro. With immobilized biochemical cues, cardiac and cutaneous tissue

regenerations were promoted by single application of the designed instructive matrix. With

microfabricated topographical guidance, perfusable cardiac micro-tissues were generated with

improved physiological relevance. These findings, in turn, contribute to our knowledge of cell-

matrix interaction, which is critical for instructive biomaterials design. Designing instructive

matrix that promotes tissue morphogenesis in situ would significantly contribute to the transition

of tissue engineering from the laboratory bench to the patient’s bedside.

Page 141: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

117

Chapter 7

7 Recommendations for future work

7.1 Investigate cardiac regeneration by collagen patch immobilized with QHREDGS peptide

In Chapter 3, we immobilized angiogenic growth factors, VEGF and bFGF, on collagen

scaffolds and applied them in rejuvenating human MSCs from aged patients for cardiac

regeneration after SVR. The growth-factor-modified collagen patches demonstrated clear

efficacy in improving the phenotype of aged MSCs both in vitro and in vivo. We also

immobilized QHREDGS peptide on collagen scaffolds and the peptide has been reported to

promote cardiac cell survival in vitro [8, 9] and cardiac regeneration in vivo [13]. Therefore, it

would be interesting to investigate the QHREDGS-modified collagen patch and compare its

efficacy in promoting cardiac regeneration with QHREDGS-conjugated hydrogel system.

7.2 Determine the mechanism of accelerated keratinocyte collective migration promoted by QHREDGS peptide

In Chapter 4, we presented the results showing that QHREDGS peptide immobilized in

chitosan-collagen substrates promoted keratinocyte collective migration in vitro while the

underlying mechanism remains unknown. With emerging experimental techniques (e.g. laser

ablation) and analytical tools (e.g. individual cell tracking in time-lapse imaging), different

mechanisms have been proposed for collective epithelial cell migration, including local tractions

pulling cooperatively towards unfilled space (termed “kenotaxis”) [455], mechanical exclusion

interactions between cells [456], and intercellular adhesion and tension [457]. We propose using

a high throughput imaging system to acquire time-lapse images of the human primary

keratinocyte collective migration and applying individual cell tracking to look at cell speed (total

travel distance per time), cell velocity (net displacement per time), persistence (velocity per

displacement), and angle of migration of each cell. These parameters will provide insight into the

effects of QHREDGS peptide on cell locomotion, particularly whether the accelerated collective

migration is caused by directed migration (chemotactic) or increased motility in random

directions (chemokinetic).

Page 142: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

118

Future studies can compare keratinocyte migration with QHREDGS supplemented in medium or

immobilized in substrate as our results showed no significant acceleration of migration when the

peptide was supplemented as soluble form. Understanding the mechanism on promoting

keratinocyte collective migration would be important for the clinical transition of QHREDGS

peptide.

7.3 Improve the perfusable biowire for drug candidates with high molecular weight

In Chapter 5, we presented the design of perfusable biowire and its capability to deliver

perfused nitric oxide to the surrounding cardiac tissue. However, poor permeability of the PTFE

micro-tubing significantly limited its application in testing other drug candidates with higher

molecular weight, which is particularly relevant for pharmaceutical agents screening. Therefore,

it is necessary to replace the PTFE micro-tubing with other tubular template made of

biocompatible material with better permeability. Ideally, the template material should have nano-

or micro-scale porous structure that enables efficient mass transfer and cell migration that

mimics the native blood vessel walls. Poly(octamethylene maleate (anhydride) citrate) (POMaC)

is a biocompatible material and has been microfabricated into micro-channels in our lab with

interconnected nano-pores and microfabricated micro-pores [558]. Moreover, POMaC material is

well suited for cardiac tissue engineering because it is an elastomer that can be dynamically

stretched with tunable elasticity in the range of adult human myocardium (200-500 kPa).

Therefore, microfabricated POMaC tubing would be a good candidate to replace the PTFE

tubing template in our current design. Drug candidates with high molecular weight and complex

structure, such as growth factors, can then be investigated.

7.4 Investigate the synergy between biochemical cues and topographical cues

In this thesis, our results demonstrated that topographical and biochemical cues, applied

independently in three studies, can regulate cell-matrix interaction to promote tissue regeneration

in vivo or create micro-tissue with increased complexity in vitro. It is therefore interesting to

investigate their synergistic effects and potentially provide topographical and biochemical cues

simultaneously to facilitate cell assembly, cell function and tissue morphogenesis in tissue

Page 143: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

119

engineering applications. Specifically, keratinocyte collective migration may be accelerated by

topographical guidance as well. By microfabrication, we can create micro-grooves on the culture

substrate and immobilize QHREDGS peptide afterwards, and then investigate the keratinocyte

collective migration with topographical and biochemical cues together.

Moreover, the cardiac micro-tissues usually risk poor cell survival in the center due to limited

mass transfer, which could be potentially improved by supplementing QHREDGS peptide in the

culture medium or immobilizing QHREDGS peptide in the collagen-based hydrogel. It would be

interesting to investigate the effect of QHREDGS peptide on self-remodeling during the micro-

tissue formation in vitro as well.

Page 144: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

120

References

1. Langer, R. and J.P. Vacanti, Tissue engineering. Science, 1993. 260(5110): p. 920-6.

2. Place, E.S., N.D. Evans and M.M. Stevens, Complexity in biomaterials for tissue

engineering. Nature Materials, 2009. 8(6): p. 457-470.

3. Segers, V.F. and R.T. Lee, Stem-cell therapy for cardiac disease. Nature, 2008.

451(7181): p. 937-42.

4. Perin, E.C., G.V. Silva, T.D. Henry, M.G. Cabreira-Hansen, W.H. Moore, S.A. Coulter,

et al., A randomized study of transendocardial injection of autologous bone marrow

mononuclear cells and cell function analysis in ischemic heart failure (FOCUS-HF). Am

Heart J, 2011. 161(6): p. 1078-87 e3.

5. Gurtner, G.C., S. Werner, Y. Barrandon and M.T. Longaker, Wound repair and

regeneration. Nature, 2008. 453(7193): p. 314-321.

6. Vincent, F., Wound healing and its impairment in the diabetic foot. Lancet, 2005.

366(9498): p. 1736-1743.

7. Rask, F., S.M. Dallabrida, N.S. Ismail, Z. Amoozgar, Y. Yeo, M.A. Rupnick, et al.,

Photocrosslinkable chitosan modified with angiopoietin-1 peptide, QHREDGS, promotes

survival of neonatal rat heart cells. Journal of Biomedical Materials Research Part A,

2010. 95A(1): p. 105-117.

8. Rask, F., A. Mihic, L. Reis, S.M. Dallabrida, N.S. Ismail, K. Sider, et al., Hydrogels

modified with QHREDGS peptide support cardiomyocyte survival in vitro and after sub-

cutaneous implantation. Soft Matter, 2010. 6(20): p. 5089-5099.

9. Reis, L.A., L.L.Y. Chiu, Y. Liang, K. Hyunh, A. Momen and M. Radisic, A peptide-

modified chitosan-collagen hydrogel for cardiac cell culture and delivery. Acta

biomaterialia, 2012. 8(3): p. 1022-36.

10. Miklas, J.W., S.M. Dallabrida, L.a. Reis, N. Ismail, M. Rupnick and M. Radisic,

QHREDGS Enhances Tube Formation, Metabolism and Survival of Endothelial Cells in

Collagen-Chitosan Hydrogels. PLoS ONE, 2013. 8(8): p. e72956-e72956.

11. Dang, L.T., N.T. Feric, C. Laschinger, W.Y. Chang, B. Zhang, G.A. Wood, et al.,

Inhibition of apoptosis in human induced pluripotent stem cells during expansion in a

defined culture using angiopoietin-1 derived peptide QHREDGS. Biomaterials, 2014.

35(27): p. 7786-99.

12. Feric, N.T., C.C.H. Cheng, M.C. Goh, V. Dudnyk, V. Di Tizio and M. Radisic,

Angiopoietin-1 peptide QHREDGS promotes osteoblast differentiation, bone matrix

deposition and mineralization on biomedical materials. Biomaterials Science, 2014. 2(10):

p. 1384-1398.

13. Reis, L.A., L.L. Chiu, J. Wu, N. Feric, C. Laschinger, A. Momen, et al., Hydrogels With

Integrin-Binding Angiopoietin-1-Derived Peptide, QHREDGS, for Treatment of Acute

Myocardial Infarction. Circ Heart Fail, 2015. 8(2): p. 333-41.

Page 145: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

121

14. Zhang, B., Y. Xiao, A. Hsieh, N. Thavandiran and M. Radisic, Micro- and

nanotechnology in cardiovascular tissue engineering. Nanotechnology, 2011. 22(49): p.

494003-494003.

15. Sun, B.K., Z. Siprashvili and P.A. Khavari, Advances in skin grafting and treatment of

cutaneous wounds. Science, 2014. 346(6212): p. 941-5.

16. Fulco, I., S. Miot, M.D. Haug, A. Barbero, A. Wixmerten, S. Feliciano, et al., Engineered

autologous cartilage tissue for nasal reconstruction after tumour resection: an

observational first-in-human trial. Lancet, 2014. 384(9940): p. 337-46.

17. Warnke, P.H., I.N. Springer, J. Wiltfang, Y. Acil, H. Eufinger, M. Wehmoller, et al.,

Growth and transplantation of a custom vascularised bone graft in a man. Lancet, 2004.

364(9436): p. 766-70.

18. McAllister, T.N., M. Maruszewski, S.A. Garrido, W. Wystrychowski, N. Dusserre, A.

Marini, et al., Effectiveness of haemodialysis access with an autologous tissue-engineered

vascular graft: a multicentre cohort study. Lancet, 2009. 373(9673): p. 1440-6.

19. Olausson, M., P.B. Patil, V.K. Kuna, P. Chougule, N. Hernandez, K. Methe, et al.,

Transplantation of an allogeneic vein bioengineered with autologous stem cells: a proof-

of-concept study. Lancet, 2012. 380(9838): p. 230-7.

20. Elkins, R.C., P.E. Dawson, S. Goldstein, S.P. Walsh and K.S. Black, Decellularized

human valve allografts. Ann Thorac Surg, 2001. 71(5 Suppl): p. S428-32.

21. Schmidt, C.E. and J.B. Leach, Neural tissue engineering: strategies for repair and

regeneration. Annu Rev Biomed Eng, 2003. 5: p. 293-347.

22. Atala, A., S.B. Bauer, S. Soker, J.J. Yoo and A.B. Retik, Tissue-engineered autologous

bladders for patients needing cystoplasty. Lancet, 2006. 367(9518): p. 1241-6.

23. Macchiarini, P., P. Jungebluth, T. Go, M.A. Asnaghi, L.E. Rees, T.A. Cogan, et al.,

Clinical transplantation of a tissue-engineered airway. Lancet, 2008. 372(9655): p. 2023-

30.

24. Raya-Rivera, A., D.R. Esquiliano, J.J. Yoo, E. Lopez-Bayghen, S. Soker and A. Atala,

Tissue-engineered autologous urethras for patients who need reconstruction: an

observational study. Lancet, 2011. 377(9772): p. 1175-82.

25. Raya-Rivera, A.M., D. Esquiliano, R. Fierro-Pastrana, E. Lopez-Bayghen, P. Valencia, R.

Ordorica-Flores, et al., Tissue-engineered autologous vaginal organs in patients: a pilot

cohort study. Lancet, 2014. 384(9940): p. 329-36.

26. Vunjak-Novakovic, G., K.O. Lui, N. Tandon and K.R. Chien, Bioengineering Heart

Muscle: A Paradigm for Regenerative Medicine. Annual Review of Biomedical

Engineering, 2011. 13(1): p. 245-267.

27. Madigan, N.N., S. McMahon, T. O'Brien, M.J. Yaszemski and A.J. Windebank, Current

tissue engineering and novel therapeutic approaches to axonal regeneration following

spinal cord injury using polymer scaffolds. Respiratory Physiology & Neurobiology,

2009. 169(2): p. 183-199.

Page 146: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

122

28. Kim, H., M.J. Cooke and M.S. Shoichet, Creating permissive microenvironments for

stem cell transplantation into the central nervous system. Trends in Biotechnology, 2012.

30(1): p. 55-63.

29. Luyten, F.P. and J. Vanlauwe, Tissue engineering approaches for osteoarthritis. Bone,

2012. 51(2): p. 289-296.

30. Jakob, F., R. Ebert, A. Ignatius, T. Matsushita, Y. Watanabe, J. Groll, et al., Bone tissue

engineering in osteoporosis. Maturitas, 2013. 75(2): p. 118-124.

31. Amer, L.D., M.J. Mahoney and S.J. Bryant, Tissue Engineering Approaches to Cell-

Based Type 1 Diabetes Therapy. Tissue Engineering Part B-Reviews, 2014. 20(5): p.

455-467.

32. Ananthanarayanan, A., B.C. Narmada, X.J. Mo, M. McMillian and H. Yu, Purpose-

driven biomaterials research in liver-tissue engineering. Trends in Biotechnology, 2011.

29(3): p. 110-118.

33. Hynes, S.R. and E.B. Lavik, A tissue-engineered approach towards retinal repair:

Scaffolds for cell transplantation to the subretinal space. Graefes Archive for Clinical and

Experimental Ophthalmology, 2010. 248(6): p. 763-778.

34. Lysaght, M.J., Product development in tissue engineering. Tissue Eng, 1995. 1(2): p. 221-

8.

35. Lysaght, M.J., N.A. Nguy and K. Sullivan, An economic survey of the emerging tissue

engineering industry. Tissue Eng, 1998. 4(3): p. 231-8.

36. Lysaght, M.J. and J. Reyes, The growth of tissue engineering. Tissue Eng, 2001. 7(5): p.

485-93.

37. Lysaght, M.J. and A.L. Hazlehurst, Tissue engineering: the end of the beginning. Tissue

Eng, 2004. 10(1-2): p. 309-20.

38. Lysaght, M.J., A. Jaklenec and E. Deweerd, Great expectations: Private sector activity in

tissue engineering, regenerative medicine, and stem cell therapeutics. Tissue Engineering

Part A, 2008. 14(2): p. 305-U57.

39. Jaklenec, A., A. Stamp, E. Deweerd, A. Sherwin and R. Langer, Progress in the Tissue

Engineering and Stem Cell Industry "Are we there yet?". Tissue Engineering Part B-

Reviews, 2012. 18(3): p. 155-166.

40. Bouchie, A., Tissue engineering firms go under. Nat Biotechnol, 2002. 20(12): p. 1178-9.

41. Discher, D.E., D.J. Mooney and P.W. Zandstra, Growth factors, matrices, and forces

combine and control stem cells. Science, 2009. 324(5935): p. 1673-7.

42. Nerem, R.M., Regenerative medicine: the emergence of an industry. J R Soc Interface,

2010. 7 Suppl 6: p. S771-5.

43. Vacanti, J.P. and R. Langer, Tissue engineering: the design and fabrication of living

replacement devices for surgical reconstruction and transplantation. Lancet, 1999. 354

Suppl 1: p. SI32-4.

Page 147: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

123

44. Thavandiran, N., S.S. Nunes, Y. Xiao and M. Radisic, Topological and electrical control

of cardiac differentiation and assembly. Stem cell research & therapy, 2013. 4(1): p. 14-

14.

45. Pashuck, E.T. and M.M. Stevens, Designing Regenerative Biomaterial Therapies for the

Clinic. Science Translational Medicine, 2012. 4(160): p. 1-12.

46. Atala, A., F.K. Kasper and A.G. Mikos, Engineering complex tissues. Sci Transl Med,

2012. 4(160): p. 160rv12.

47. Lutolf, M.P. and J.A. Hubbell, Synthetic biomaterials as instructive extracellular

microenvironments for morphogenesis in tissue engineering. Nature Biotechnology, 2005.

23(1): p. 47-55.

48. Siani-Rose, M., R.S. Kellar, G.K. Naughton and S.K. Williams, Use of cultured three-

dimensional tissue for treating congestive heart failure. 2007, Theregen, Inc.

49. Parenteau-Bareil, R., R. Gauvin and F. Berthod, Collagen-Based Biomaterials for Tissue

Engineering Applications. Materials, 2010. 3(3): p. 1863-1887.

50. Steven, F.S. and D.S. Jackson, Purification and amino acid composition of monomeric

and polymeric collagens. Biochem J, 1967. 104(2): p. 534-6.

51. Gross, J., J.H. Highberger and F.O. Schmitt, Extraction of Collagen from Connective

Tissue by Neutral Salt Solutions. Proc Natl Acad Sci U S A, 1955. 41(1): p. 1-7.

52. Drake, M.P., P.F. Davison, S. Bump and F.O. Schmitt, Action of proteolytic enzymes on

tropocollagen and insoluble collagen. Biochemistry, 1966. 5(1): p. 301-12.

53. Huyer, L.D., M. Montgomery, Y. Zhao, Y. Xiao, G. Conant, A. Korolj, et al., Biomaterial

based cardiac tissue engineering and its applications. Biomedical materials, 2015. 10(3):

p. 034004-034004.

54. Rinaudo, M., Chitin and chitosan: Properties and applications. Progress in Polymer

Science, 2006. 31(7): p. 603-632.

55. Pavinatto, F.J., L. Caseli and O.N. Oliveira, Chitosan in Nanostructured Thin Films.

Biomacromolecules, 2010. 11(8): p. 1897-1908.

56. Zheng, L.Y. and J.A.F. Zhu, Study on antimicrobial activity of chitosan with different

molecular weights. Carbohydrate Polymers, 2003. 54(4): p. 527-530.

57. Sogias, I.A., A.C. Williams and V.V. Khutoryanskiy, Why is chitosan mucoadhesive?

Biomacromolecules, 2008. 9(7): p. 1837-1842.

58. Okamoto, Y., K. Kawakami, K. Miyatake, M. Morimoto, Y. Shigemasa and S. Minami,

Analgesic effects of chitin and chitosan. Carbohydrate Polymers, 2002. 49(3): p. 249-252.

59. Wedmore, I., J.G. McManus, A.E. Pusateri and J.B. Holcomb, A special report on the

chitosan-based hemostatic dressing: Experience in current combat operations. Journal of

Trauma-Injury Infection and Critical Care, 2006. 60(3): p. 655-658.

60. Pillai, C.K.S., W. Paul and C.P. Sharma, Chitin and chitosan polymers: Chemistry,

solubility and fiber formation. Progress in Polymer Science, 2009. 34(7): p. 641-678.

Page 148: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

124

61. Tomihata, K. and Y. Ikada, In vitro and in vivo degradation of films of chitin and its

deacetylated derivatives. Biomaterials, 1997. 18(7): p. 567-575.

62. Jayakumar, R., M. Prabaharan, P.T.S. Kumar, S.V. Nair and H. Tamura, Biomaterials

based on chitin and chitosan in wound dressing applications. Biotechnology Advances,

2011. 29(3): p. 322-337.

63. Bhattarai, N., J. Gunn and M.Q. Zhang, Chitosan-based hydrogels for controlled,

localized drug delivery. Advanced Drug Delivery Reviews, 2010. 62(1): p. 83-99.

64. Di Martino, A., M. Sittinger and M.V. Risbud, Chitosan: A versatile biopolymer for

orthopaedic tissue-engineering. Biomaterials, 2005. 26(30): p. 5983-5990.

65. Meng, S., Z.J. Liu, L. Shen, Z. Guo, L.S.L. Chou, W. Zhong, et al., The effect of a layer-

by-layer chitosan-heparin coating on the endothelialization and coagulation properties of

a coronary stent system. Biomaterials, 2009. 30(12): p. 2276-2283.

66. Wurm, F.M., Production of recombinant protein therapeutics in cultivated mammalian

cells. Nat Biotechnol, 2004. 22(11): p. 1393-8.

67. Yanagisawamiwa, A., Y. Uchida, F. Nakamura, T. Tomaru, H. Kido, T. Kamijo, et al.,

Salvage of Infarcted Myocardium by Angiogenic Action of Basic Fibroblast Growth-

Factor. Science, 1992. 257(5075): p. 1401-1403.

68. Takeshita, S., L.P. Zheng, E. Brogi, M. Kearney, L.Q. Pu, S. Bunting, et al., Therapeutic

angiogenesis. A single intraarterial bolus of vascular endothelial growth factor augments

revascularization in a rabbit ischemic hind limb model. J Clin Invest, 1994. 93(2): p. 662-

70.

69. Takeshita, S., L.Q. Pu, L.A. Stein, A.D. Sniderman, S. Bunting, N. Ferrara, et al.,

Intramuscular administration of vascular endothelial growth factor induces dose-

dependent collateral artery augmentation in a rabbit model of chronic limb ischemia.

Circulation, 1994. 90(5 Pt 2): p. II228-34.

70. Smiell, J.M., T.J. Wieman, D.L. Steed, B.H. Perry, A.R. Sampson and B.H. Schwab,

Efficacy and safety of becaplermin (recombinant human platelet-derived growth factor-

BB) in patients with nonhealing, lower extremity diabetic ulcers: a combined analysis of

four randomized studies. Wound repair and regeneration : official publication of the

Wound Healing Society [and] the European Tissue Repair Society, 1999. 7(5): p. 335-46.

71. Burkus, J.K., M.F. Gornet, C.A. Dickman and T.A. Zdeblick, Anterior lumbar interbody

fusion using rhBMP-2 with tapered interbody cages. Journal of Spinal Disorders &

Techniques, 2002. 15(5): p. 337-349.

72. Govender, S., C. Csimma, H.K. Genant, A. Valentin-Opran and B.S. Grp, Recombinant

human bone morphogenetic protein-2 for treatment of open tibial fractures - A

prospective, controlled, randomized study of four hundred and fifty patients. Journal of

Bone and Joint Surgery-American Volume, 2002. 84a(12): p. 2123-2134.

73. Boyne, P.J., R.E. Marx, M. Nevins, G. Triplett, E. Lazaro, L.C. Lilly, et al., A feasibility

study evaluating rhBMP-2/absorbable collagen sponge for maxillary sinus floor

augmentation. International Journal of Periodontics & Restorative Dentistry, 1997. 17(1):

p. 10-25.

Page 149: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

125

74. Boyne, P.J., L.C. Lilly, R.E. Marx, P.K. Moy, M. Nevins, D.B. Spagnoli, et al., De novo

bone induction by recombinant human bone morphogenetic protein-2 (rhBMP-2) in

maxillary sinus floor augmentation. Journal of Oral and Maxillofacial Surgery, 2005.

63(12): p. 1693-1707.

75. Wong, S., M.S. Shim and Y.J. Kwon, Synthetically designed peptide-based biomaterials

with stimuli-responsive and membrane-active properties for biomedical applications.

Journal of Materials Chemistry B, 2014. 2(6): p. 595-615.

76. French, K.M., I. Somasuntharam and M.E. Davis, Self-assembling peptide-based delivery

of therapeutics for myocardial infarction. Adv Drug Deliv Rev, 2016. 96: p. 40-53.

77. Pierschbacher, M.D. and E. Ruoslahti, Cell Attachment Activity of Fibronectin Can Be

Duplicated by Small Synthetic Fragments of the Molecule. Nature, 1984. 309(5963): p.

30-33.

78. Underwood, P.A., F.A. Bennett, A. Kirkpatrick, P.A. Bean and B.A. Moss, Evidence for

the Location of a Binding Sequence for the Alpha-2-Beta-1 Integrin of Endothelial-Cells,

in the Beta-1 Subunit of Laminin. Biochemical Journal, 1995. 309: p. 765-771.

79. Xu, J.S., D. Rodriguez, E. Petitclerc, J.J. Kim, M. Hangai, Y.S. Moon, et al., Proteolytic

exposure of a cryptic site within collagen type IV is required for angiogenesis and tumor

growth in vivo. Journal of Cell Biology, 2001. 155(5): p. 859-859.

80. Malmsten, M., M. Davoudi, B. Walse, V. Rydengard, M. Pasupuleti, M. Morgelin, et al.,

Antimicrobial peptides derived from growth factors. Growth Factors, 2007. 25(1): p. 60-

70.

81. Furstenberger, G. and H.J. Senn, Insulin-like growth factors and cancer. Lancet Oncol,

2002. 3(5): p. 298-302.

82. Richardson, T.P., M.C. Peters, A.B. Ennett and D.J. Mooney, Polymeric system for dual

growth factor delivery. Nat Biotechnol, 2001. 19(11): p. 1029-34.

83. Sohier, J., T.J. Vlugt, N. Cabrol, C. Van Blitterswijk, K. de Groot and J.M. Bezemer,

Dual release of proteins from porous polymeric scaffolds. J Control Release, 2006.

111(1-2): p. 95-106.

84. Ferrara, N. and K. Alitalo, Clinical applications of angiogenic growth factors and their

inhibitors. Nat Med, 1999. 5(12): p. 1359-64.

85. Martino, M.M., P.S. Briquez, E. Guc, F. Tortelli, W.W. Kilarski, S. Metzger, et al.,

Growth Factors Engineered for Super-Affinity to the Extracellular Matrix Enhance

Tissue Healing. Science, 2014. 343(6173): p. 885-888.

86. Kuhl, P.R. and L.G. Griffith-Cima, Tethered epidermal growth factor as a paradigm for

growth factor-induced stimulation from the solid phase. Nat Med, 1996. 2(9): p. 1022-7.

87. Chiu, L.L.Y., R.D. Weisel, R.-K. Li and M. Radisic, Defining conditions for covalent

immobilization of angiogenic growth factors onto scaffolds for tissue engineering.

Journal of Tissue Engineering and Regenerative Medicine, 2011. 5(1): p. 69-84.

88. Xiao, Y., L.A. Reis, Y. Zhao and M. Radisic, Modifications of collagen-based

biomaterials with immobilized growth factors or peptides. Methods, 2015. 84: p. 44-52.

Page 150: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

126

89. Elosegui-Artola, A., E. Bazellieres, M.D. Allen, I. Andreu, R. Oria, R. Sunyer, et al.,

Rigidity sensing and adaptation through regulation of integrin types. Nature Materials,

2014. 13(6): p. 631-637.

90. Engler, A.J., S. Sen, H.L. Sweeney and D.E. Discher, Matrix elasticity directs stem cell

lineage specification. Cell, 2006. 126(4): p. 677-89.

91. Yang, C., M.W. Tibbitt, L. Basta and K.S. Anseth, Mechanical memory and dosing

influence stem cell fate. Nature Materials, 2014. 13(6): p. 645-652.

92. Chen, W.Q., L.G. Villa-Diaz, Y.B. Sun, S.N. Weng, J.K. Kim, R.H.W. Lam, et al.,

Nanotopography Influences Adhesion, Spreading, and Self-Renewal of Human

Embryonic Stem Cells. Acs Nano, 2012. 6(5): p. 4094-4103.

93. Jaggy, M., P. Zhang, A.M. Greiner, T.J. Autenrieth, V. Nedashkivska, A.N. Efremov, et

al., Hierarchical Micro-Nano Surface Topography Promotes Long-Term Maintenance of

Undifferentiated Mouse Embryonic Stem Cells. Nano Lett, 2015. 15(10): p. 7146-54.

94. Mechanobiology in harness. Nat Mater, 2014. 13(6): p. 531.

95. Bursac, N., K.K. Parker, S. Iravanian and L. Tung, Cardiomyocyte cultures with

controlled macroscopic anisotropy: a model for functional electrophysiological studies of

cardiac muscle. Circ Res, 2002. 91(12): p. e45-54.

96. Khademhosseini, A., G. Eng, J. Yeh, P.A. Kucharczyk, R. Langer, G. Vunjak-Novakovic,

et al., Microfluidic patterning for fabrication of contractile cardiac organoids. Biomed

Microdevices, 2007. 9(2): p. 149-57.

97. Badie, N. and N. Bursac, Novel micropatterned cardiac cell cultures with realistic

ventricular microstructure. Biophys J, 2009. 96(9): p. 3873-85.

98. Kim, D.-H., E.A. Lipke, P. Kim, R. Cheong, S. Thompson, M. Delannoy, et al.,

Nanoscale cues regulate the structure and function of macroscopic cardiac tissue

constructs. Proceedings of the National Academy of Sciences, 2010. 107(2): p. 565-570.

99. Engelmayr, G.C., M. Cheng, C.J. Bettinger, J.T. Borenstein, R. Langer and L.E. Freed,

Accordion-like honeycombs for tissue engineering of cardiac anisotropy. Nat Mater,

2008. 7(12): p. 1003-1010.

100. Madden, L.R., D.J. Mortisen, E.M. Sussman, S.K. Dupras, J.A. Fugate, J.L. Cuy, et al.,

Proangiogenic scaffolds as functional templates for cardiac tissue engineering.

Proceedings of the National Academy of Sciences, 2010. 107(34): p. 15211-15216.

101. Daley, G.Q., The promise and perils of stem cell therapeutics. Cell Stem Cell, 2012. 10(6):

p. 740-9.

102. Wang, S., X. Qu and R.C. Zhao, Clinical applications of mesenchymal stem cells. J

Hematol Oncol, 2012. 5: p. 19.

103. Pittenger, M.F., A.M. Mackay, S.C. Beck, R.K. Jaiswal, R. Douglas, J.D. Mosca, et al.,

Multilineage potential of adult human mesenchymal stem cells. Science, 1999. 284(5411):

p. 143-147.

Page 151: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

127

104. Zuk, P.A., M. Zhu, P. Ashjian, D.A. De Ugarte, J.I. Huang, H. Mizuno, et al., Human

adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell, 2002.

13(12): p. 4279-4295.

105. In 't Anker, P.S., S.A. Scherjon, C. Kleijburg-van der Keur, G.M. de Groot-Swings, F.H.

Claas, W.E. Fibbe, et al., Isolation of mesenchymal stem cells of fetal or maternal origin

from human placenta. Stem Cells, 2004. 22(7): p. 1338-45.

106. Shih, D.T.B., D.C. Lee, S.C. Chen, R.Y. Tsai, C.T. Huang, C.C. Tsai, et al., Isolation and

characterization of neurogenic mesenchymal stem cells in human scalp tissue. Stem Cells,

2005. 23(7): p. 1012-1020.

107. Erices, A., P. Conget and J.J. Minguell, Mesenchymal progenitor cells in human

umbilical cord blood. Br J Haematol, 2000. 109(1): p. 235-42.

108. Sarugaser, R., D. Lickorish, D. Baksh, M.M. Hosseini and J.E. Davies, Human umbilical

cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem Cells, 2005.

23(2): p. 220-9.

109. Wang, H.S., S.C. Hung, S.T. Peng, C.C. Huang, H.M. Wei, Y.J. Guo, et al.,

Mesenchymal stem cells in the Wharton's jelly of the human umbilical cord. Stem Cells,

2004. 22(7): p. 1330-7.

110. Gronthos, S., M. Mankani, J. Brahim, P.G. Robey and S. Shi, Postnatal human dental

pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A, 2000. 97(25): p.

13625-30.

111. Nadri, S. and M. Soleimani, Comparative analysis of mesenchymal stromal cells from

murine bone marrow and amniotic fluid. Cytotherapy, 2007. 9(8): p. 729-37.

112. De Bari, C., F. Dell'Accio, P. Tylzanowski and F.P. Luyten, Multipotent mesenchymal

stem cells from adult human synovial membrane. Arthritis Rheum, 2001. 44(8): p. 1928-

42.

113. Patki, S., S. Kadam, V. Chandra and R. Bhonde, Human breast milk is a rich source of

multipotent mesenchymal stem cells. Human Cell, 2010. 23(2): p. 35-40.

114. Santos, F., P.Z. Andrade, M.M. Abecasis, J.M. Gimble, L.G. Chase, A.M. Campbell, et

al., Toward a clinical-grade expansion of mesenchymal stem cells from human sources: a

microcarrier-based culture system under xeno-free conditions. Tissue Eng Part C

Methods, 2011. 17(12): p. 1201-10.

115. Quarto, R., M. Mastrogiacomo, R. Cancedda, S.M. Kutepov, V. Mukhachev, A.

Lavroukov, et al., Repair of large bone defects with the use of autologous bone marrow

stromal cells. New England Journal of Medicine, 2001. 344(5): p. 385-386.

116. Haleem, A.M., A.A. El Singergy, D. Sabry, H.M. Atta, L.A. Rashed, C.R. Chu, et al.,

The Clinical Use of Human Culture-Expanded Autologous Bone Marrow Mesenchymal

Stem Cells Transplanted on Platelet-Rich Fibrin Glue in the Treatment of Articular

Cartilage Defects: A Pilot Study and Preliminary Results. Cartilage, 2010. 1(4): p. 253-

261.

117. Noiseux, N., M. Gnecchi, M. Lopez-Ilasaca, L. Zhang, S.D. Solomon, A. Deb, et al.,

Mesenchymal stem cells overexpressing Akt dramatically repair infarcted myocardium

Page 152: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

128

and improve cardiac function despite infrequent cellular fusion or differentiation. Mol

Ther, 2006. 14(6): p. 840-50.

118. Kemp, K., D. Gordon, D.C. Wraith, E. Mallam, E. Hartfield, J. Uney, et al., Fusion

between human mesenchymal stem cells and rodent cerebellar Purkinje cells.

Neuropathology and Applied Neurobiology, 2011. 37(2): p. 166-178.

119. Nauta, A.J. and W.E. Fibbe, Immunomodulatory properties of mesenchymal stromal cells.

Blood, 2007. 110(10): p. 3499-506.

120. Chen, P.M., M.L. Yen, K.J. Liu, H.K. Sytwu and B.L. Yen, Immunomodulatory

properties of human adult and fetal multipotent mesenchymal stem cells. J Biomed Sci,

2011. 18: p. 49.

121. Ranganath, S.H., O. Levy, M.S. Inamdar and J.M. Karp, Harnessing the mesenchymal

stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell, 2012.

10(3): p. 244-58.

122. Yew, T.L., Y.T. Hung, H.Y. Li, H.W. Chen, L.L. Chen, K.S. Tsai, et al., Enhancement of

Wound Healing by Human Multipotent Stromal Cell Conditioned Medium: The

Paracrine Factors and p38 MAPK Activation. Cell Transplantation, 2011. 20(5): p. 693-

706.

123. Rustad, K.C., V.W. Wong, M. Sorkin, J.P. Glotzbach, M.R. Major, J. Rajadas, et al.,

Enhancement of mesenchymal stem cell angiogenic capacity and stemness by a

biomimetic hydrogel scaffold. Biomaterials, 2012. 33(1): p. 80-90.

124. Guan, J., Z. Zhu, R.C. Zhao, Z. Xiao, C. Wu, Q. Han, et al., Transplantation of human

mesenchymal stem cells loaded on collagen scaffolds for the treatment of traumatic brain

injury in rats. Biomaterials, 2013. 34(24): p. 5937-46.

125. Manassero, M., V. Viateau, M. Deschepper, K. Oudina, D. Logeart-Avramoglou, H.

Petite, et al., Bone Regeneration in Sheep Using Acropora Coral, a Natural Resorbable

Scaffold, and Autologous Mesenchymal Stem Cells. Tissue Engineering Part A, 2013.

19(13-14): p. 1554-1563.

126. Guo, R.J., C.L. Ward, J.M. Davidson, C.L. Duvall, J.C. Wenke and S.A. Guelcher, A

transient cell-shielding method for viable MSC delivery within hydrophobic scaffolds

polymerized in situ. Biomaterials, 2015. 54: p. 21-33.

127. Klopp, A.H., A. Gupta, E. Spaeth, M. Andreeff and F. Marini, Concise Review:

Dissecting a Discrepancy in the Literature: Do Mesenchymal Stem Cells Support or

Suppress Tumor Growth? Stem Cells, 2011. 29(1): p. 11-19.

128. Sethe, S., A. Scutt and A. Stolzing, Aging of mesenchymal stem cells. Ageing Res Rev,

2006. 5(1): p. 91-116.

129. Bui, A.L., T.B. Horwich and G.C. Fonarow, Epidemiology and risk profile of heart

failure. Nat Rev Cardiol, 2011. 8(1): p. 30-41.

130. Nerem, R.M. and A. Sambanis, Tissue engineering: from biology to biological substitutes.

Tissue Eng, 1995. 1(1): p. 3-13.

131. Muller-Ehmsen, J., K.L. Peterson, L. Kedes, P. Whittaker, J.S. Dow, T.I. Long, et al.,

Rebuilding a damaged heart: long-term survival of transplanted neonatal rat

Page 153: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

129

cardiomyocytes after myocardial infarction and effect on cardiac function. Circulation,

2002. 105(14): p. 1720-6.

132. Reinecke, H., M. Zhang, T. Bartosek and C.E. Murry, Survival, integration, and

differentiation of cardiomyocyte grafts: a study in normal and injured rat hearts.

Circulation, 1999. 100(2): p. 193-202.

133. Leor, J., S. Aboulafia-Etzion, A. Dar, L. Shapiro, I.M. Barbash, A. Battler, et al.,

Bioengineered cardiac grafts: A new approach to repair the infarcted myocardium?

Circulation, 2000. 102(19 Suppl 3): p. III56-61.

134. Zong, X., H. Bien, C.Y. Chung, L. Yin, D. Fang, B.S. Hsiao, et al., Electrospun fine-

textured scaffolds for heart tissue constructs. Biomaterials, 2005. 26(26): p. 5330-8.

135. Li, R.K., Z.Q. Jia, R.D. Weisel, D.A. Mickle, A. Choi and T.M. Yau, Survival and

function of bioengineered cardiac grafts. Circulation, 1999. 100(19 Suppl): p. II63-9.

136. Kehat, I., D. Kenyagin-Karsenti, M. Snir, H. Segev, M. Amit, A. Gepstein, et al., Human

embryonic stem cells can differentiate into myocytes with structural and functional

properties of cardiomyocytes. Journal of Clinical Investigation, 2001. 108(3): p. 407-414.

137. Kattman, S.J., T.L. Huber and G.M. Keller, Multipotent Flk-1(+) cardiovascular

progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle

lineages. Developmental Cell, 2006. 11(5): p. 723-732.

138. Yang, L., M.H. Soonpaa, E.D. Adler, T.K. Roepke, S.J. Kattman, M. Kennedy, et al.,

Human cardiovascular progenitor cells develop from a KDR plus embryonic-stem-cell-

derived population. Nature, 2008. 453(7194): p. 524-U6.

139. Kouskoff, V., G. Lacaud, S. Schwantz, H.J. Fehling and G. Keller, Sequential

development of hematopoietic and cardiac mesoderm during embryonic stem cell

differentiation. Proc Natl Acad Sci U S A, 2005. 102(37): p. 13170-5.

140. Guo, X.M., Y.S. Zhao, H.X. Chang, C.Y. Wang, L.L. E, X.A. Zhang, et al., Creation of

engineered cardiac tissue in vitro from mouse embryonic stem cells. Circulation, 2006.

113(18): p. 2229-37.

141. Stevens, K.R., L. Pabon, V. Muskheli and C.E. Murry, Scaffold-free human cardiac

tissue patch created from embryonic stem cells. Tissue Eng Part A, 2009. 15(6): p. 1211-

22.

142. Stevens, K.R., K.L. Kreutziger, S.K. Dupras, F.S. Korte, M. Regnier, V. Muskheli, et al.,

Physiological function and transplantation of scaffold-free and vascularized human

cardiac muscle tissue. Proc Natl Acad Sci U S A, 2009. 106(39): p. 16568-73.

143. Takahashi, K. and S. Yamanaka, Induction of pluripotent stem cells from mouse

embryonic and adult fibroblast cultures by defined factors. Cell, 2006. 126(4): p. 663-76.

144. Mauritz, C., K. Schwanke, M. Reppel, S. Neef, K. Katsirntaki, L.S. Maier, et al.,

Generation of functional murine cardiac myocytes from induced pluripotent stem cells.

Circulation, 2008. 118(5): p. 507-17.

145. Zwi, L., O. Caspi, G. Arbel, I. Huber, A. Gepstein, I.H. Park, et al., Cardiomyocyte

differentiation of human induced pluripotent stem cells. Circulation, 2009. 120(15): p.

1513-23.

Page 154: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

130

146. Zhang, J., G.F. Wilson, A.G. Soerens, C.H. Koonce, J. Yu, S.P. Palecek, et al., Functional

cardiomyocytes derived from human induced pluripotent stem cells. Circ Res, 2009.

104(4): p. e30-41.

147. Tulloch, N.L., V. Muskheli, M.V. Razumova, F.S. Korte, M. Regnier, K.D. Hauch, et al.,

Growth of engineered human myocardium with mechanical loading and vascular

coculture. Circ Res, 2011. 109(1): p. 47-59.

148. Murry, C.E., M.A. Kay, T. Bartosek, S.D. Hauschka and S.M. Schwartz, Muscle

differentiation during repair of myocardial necrosis in rats via gene transfer with MyoD. J

Clin Invest, 1996. 98(10): p. 2209-17.

149. Efe, J.A., S. Hilcove, J. Kim, H. Zhou, K. Ouyang, G. Wang, et al., Conversion of mouse

fibroblasts into cardiomyocytes using a direct reprogramming strategy. Nature Cell

Biology, 2011. 13(3): p. 215-U61.

150. Jung, Y., G. Bauer and J.A. Nolta, Concise review: Induced pluripotent stem cell-derived

mesenchymal stem cells: progress toward safe clinical products. Stem Cells, 2012. 30(1):

p. 42-7.

151. Lee, A.S., C. Tang, M.S. Rao, I.L. Weissman and J.C. Wu, Tumorigenicity as a clinical

hurdle for pluripotent stem cell therapies. Nature Medicine, 2013. 19(8): p. 998-1004.

152. Miller, D.M., S.D. Thomas, A. Islam, D. Muench and K. Sedoris, c-Myc and cancer

metabolism. Clin Cancer Res, 2012. 18(20): p. 5546-53.

153. Hacein-Bey-Abina, S., C. Von Kalle, M. Schmidt, M.P. McCormack, N. Wulffraat, P.

Leboulch, et al., LMO2-associated clonal T cell proliferation in two patients after gene

therapy for SCID-X1. Science, 2003. 302(5644): p. 415-9.

154. Cavazzana-Calvo, M., E. Payen, O. Negre, G. Wang, K. Hehir, F. Fusil, et al.,

Transfusion independence and HMGA2 activation after gene therapy of human beta-

thalassaemia. Nature, 2010. 467(7313): p. 318-22.

155. Gore, A., Z. Li, H.L. Fung, J.E. Young, S. Agarwal, J. Antosiewicz-Bourget, et al.,

Somatic coding mutations in human induced pluripotent stem cells. Nature, 2011.

471(7336): p. 63-7.

156. Bergmann, O., R.D. Bhardwaj, S. Bernard, S. Zdunek, F. Barnabe-Heider, S. Walsh, et

al., Evidence for Cardiomyocyte Renewal in Humans. Science, 2009. 324(5923): p. 98-

102.

157. Kajstura, J., K. Urbanek, S. Perl, T. Hosoda, H. Zheng, B. Ogorek, et al.,

Cardiomyogenesis in the adult human heart. Circ Res, 2010. 107(2): p. 305-15.

158. Ruvinov, E., T. Harel-Adar and S. Cohen, Bioengineering the Infarcted Heart by

Applying Bio-inspired Materials. Journal of Cardiovascular Translational Research,

2011. 4(5): p. 559-574.

159. Urbanek, K., F. Quaini, G. Tasca, D. Torella, C. Castaldo, B. Nadal-Ginard, et al., Intense

myocyte formation from cardiac stem cells in human cardiac hypertrophy. Proceedings of

the National Academy of Sciences of the United States of America, 2003. 100(18): p.

10440-10445.

Page 155: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

131

160. Urbanek, K., D. Torella, F. Sheikh, A. De Angelis, D. Nurzynska, F. Silvestri, et al.,

Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart

failure. Proceedings of the National Academy of Sciences of the United States of America,

2005. 102(24): p. 8692-8697.

161. Urbanek, K., D. Cesselli, M. Rota, A. Nascimbene, A. De Angelis, T. Hosoda, et al.,

Stem cell niches in the adult mouse heart. Proc Natl Acad Sci U S A, 2006. 103(24): p.

9226-31.

162. Laugwitz, K.L., A. Moretti, J. Lam, P. Gruber, Y.H. Chen, S. Woodard, et al., Postnatal

isl1+cardioblasts enter fully differentiated cardiomyocyte lineages. Nature, 2005.

433(7026): p. 647-653.

163. Segers, V.F.M. and R.T. Lee, Stem-cell therapy for cardiac disease. Nature, 2008.

451(7181): p. 937-942.

164. Passier, R., L.W. van Laake and C.L. Mummery, Stem-cell-based therapy and lessons

from the heart. Nature, 2008. 453(7193): p. 322-329.

165. Beltrami, A.P., L. Barlucchi, D. Torella, M. Baker, F. Limana, S. Chimenti, et al., Adult

cardiac stem cells are multipotent and support myocardial regeneration. Cell, 2003.

114(6): p. 763-776.

166. Domian, I.J., M. Chiravuri, P. van der Meer, A.W. Feinberg, X. Shi, Y. Shao, et al.,

Generation of Functional Ventricular Heart Muscle from Mouse Ventricular Progenitor

Cells. Science, 2009. 326(5951): p. 426-429.

167. Messina, E., L. De Angelis, G. Frati, S. Morrone, S. Chimenti, F. Fiordaliso, et al.,

Isolation and expansion of adult cardiac stem cells from human and murine heart.

Circulation Research, 2004. 95(9): p. 911-921.

168. Johnston, P.V., T. Sasano, K. Mills, R. Evers, S.T. Lee, R.R. Smith, et al., Engraftment,

Differentiation, and Functional Benefits of Autologous Cardiosphere-Derived Cells in

Porcine Ischemic Cardiomyopathy. Circulation, 2009. 120(12): p. 1075-U95.

169. Chimenti, I., R.R. Smith, T.S. Li, G. Gerstenblith, E. Messina, A. Giacomello, et al.,

Relative roles of direct regeneration versus paracrine effects of human cardiosphere-

derived cells transplanted into infarcted mice. Circ Res, 2010. 106(5): p. 971-80.

170. Orlic, D., J. Kajstura, S. Chimenti, I. Jakoniuk, S.M. Anderson, B. Li, et al., Bone

marrow cells regenerate infarcted myocardium. Nature, 2001. 410(6829): p. 701-5.

171. Jackson, K.A., S.M. Majka, H. Wang, J. Pocius, C.J. Hartley, M.W. Majesky, et al.,

Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J

Clin Invest, 2001. 107(11): p. 1395-402.

172. Deb, A., S.H. Wang, K.A. Skelding, D. Miller, D. Simper and N.M. Caplice, Bone

marrow-derived cardiomyocytes are present in adult human heart - A study of gender-

mismatched bone marrow transplantation patients. Circulation, 2003. 107(9): p. 1247-

1249.

173. Orlic, D., J. Kajstura, S. Chimenti, D.M. Bodine, A. Leri and P. Anversa, Bone marrow

stem cells regenerate infarcted myocardium. Pediatric Transplantation, 2003. 7: p. 86-88.

Page 156: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

132

174. Orlic, D., J. Kajstura, S. Chimenti, F. Limana, I. Jakoniuk, F. Quaini, et al., Mobilized

bone marrow cells repair the infarcted heart, improving function and survival.

Proceedings of the National Academy of Sciences of the United States of America, 2001.

98(18): p. 10344-10349.

175. Chen, S.L., W. Fang, F. Ye, Y.H. Liu, J. Qian, S. Shan, et al., Effect on left ventricular

function of intracoronary transplantation of autologous bone marrow mesenchymal stem

cell in patients with acute myocardial infarction. American Journal of Cardiology, 2004.

94(1): p. 92-95.

176. Fraser, J.K., I. Wulur, Z. Alfonso and M.H. Hedrick, Fat tissue: an underappreciated

source of stem cells for biotechnology. Trends in Biotechnology, 2006. 24(4): p. 150-154.

177. Bai, X.W. and E. Alt, Myocardial regeneration potential of adipose tissue-derived stem

cells. Biochemical and Biophysical Research Communications, 2010. 401(3): p. 321-326.

178. Bai, X.W., Y.S. Yan, Y.H. Song, M. Seidensticker, B. Rabinovich, R. Metzele, et al.,

Both cultured and freshly isolated adipose tissue-derived stem cells enhance cardiac

function after acute myocardial infarction. European Heart Journal, 2010. 31(4): p. 489-

501.

179. Gaebel, R., D. Furlani, H. Sorg, B. Polchow, J. Frank, K. Bieback, et al., Cell Origin of

Human Mesenchymal Stem Cells Determines a Different Healing Performance in

Cardiac Regeneration. Plos One, 2011. 6(2).

180. Radisic, M., M. Euloth, L. Yang, R. Langer, L.E. Freed and G. Vunjak-Novakovic, High-

density seeding of myocyte cells for cardiac tissue engineering. Biotechnology and

Bioengineering, 2003. 82(4): p. 403-414.

181. Kang, K., L. Sun, Y. Xiao, S.-H. Li, J. Wu, J. Guo, et al., Aged human cells rejuvenated

by cytokine enhancement of biomaterials for surgical ventricular restoration. Journal of

the American College of Cardiology, 2012. 60(21): p. 2237-49.

182. Bursac, N., M. Papadaki, R.J. Cohen, F.J. Schoen, S.R. Eisenberg, R. Carrier, et al.,

Cardiac muscle tissue engineering: toward an in vitro model for electrophysiological

studies. Am J Physiol, 1999. 277(2 Pt 2): p. H433-44.

183. Shin, M., O. Ishii, T. Sueda and J.P. Vacanti, Contractile cardiac grafts using a novel

nanofibrous mesh. Biomaterials, 2004. 25(17): p. 3717-3723.

184. Ozawa, T., D.A.G. Mickle, R.D. Weisel, N. Koyama, S. Ozawa and R.K. Li, Optimal

biomaterial for creation of autologous cardiac grafts. Circulation, 2002. 106(13): p. I176-

I182.

185. Rockwood, D.N., R.E. Akins, Jr., I.C. Parrag, K.A. Woodhouse and J.F. Rabolt, Culture

on electrospun polyurethane scaffolds decreases atrial natriuretic peptide expression by

cardiomyocytes in vitro. Biomaterials, 2008. 29(36): p. 4783-91.

186. Hidalgo-Bastida, L.A., J.J. Barry, N.M. Everitt, F.R. Rose, L.D. Buttery, I.P. Hall, et al.,

Cell adhesion and mechanical properties of a flexible scaffold for cardiac tissue

engineering. Acta Biomater, 2007. 3(4): p. 457-62.

Page 157: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

133

187. Ott, H.C., T.S. Matthiesen, S.K. Goh, L.D. Black, S.M. Kren, T.I. Netoff, et al.,

Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial heart.

Nat Med, 2008. 14(2): p. 213-21.

188. Dai, W., L.E. Wold, J.S. Dow and R.A. Kloner, Thickening of the infarcted wall by

collagen injection improves left ventricular function in rats: a novel approach to preserve

cardiac function after myocardial infarction. J Am Coll Cardiol, 2005. 46(4): p. 714-9.

189. Suuronen, E.J., J.P. Veinot, S. Wong, V. Kapila, J. Price, M. Griffith, et al., Tissue-

engineered injectable collagen-based matrices for improved cell delivery and

vascularization of ischemic tissue using CD133+progenitors expanded from the

peripheral blood. Circulation, 2006. 114: p. I138-I144.

190. Kofidis, T., A. Lenz, J. Boublik, P. Akhyari, B. Wachsmann, K. Mueller-Stahl, et al.,

Pulsatile perfusion and cardiomyocyte viability in a solid three-dimensional matrix.

Biomaterials, 2003. 24(27): p. 5009-14.

191. Christman, K.L., A.J. Vardanian, Q. Fang, R.E. Sievers, H.H. Fok and R.J. Lee,

Injectable fibrin scaffold improves cell transplant survival, reduces infarct expansion, and

induces neovasculature formation in ischemic myocardium. J Am Coll Cardiol, 2004.

44(3): p. 654-60.

192. Wang, H., X. Zhang, Y. Li, Y. Ma, Y. Zhang, Z. Liu, et al., Improved myocardial

performance in infarcted rat heart by co-injection of basic fibroblast growth factor with

temperature-responsive chitosan hydrogel. J Heart Lung Transplant, 2010. 29(8): p. 881-

7.

193. Liu, Z., H. Wang, Y. Wang, Q. Lin, A. Yao, F. Cao, et al., The influence of chitosan

hydrogel on stem cell engraftment, survival and homing in the ischemic myocardial

microenvironment. Biomaterials, 2012. 33(11): p. 3093-106.

194. Landa, N., L. Miller, M.S. Feinberg, R. Holbova, M. Shachar, I. Freeman, et al., Effect of

injectable alginate implant on cardiac remodeling and function after recent and old

infarcts in rat. Circulation, 2008. 117(11): p. 1388-96.

195. Leor, J., S. Tuvia, V. Guetta, F. Manczur, D. Castel, U. Willenz, et al., Intracoronary

injection of in situ forming alginate hydrogel reverses left ventricular remodeling after

myocardial infarction in Swine. J Am Coll Cardiol, 2009. 54(11): p. 1014-23.

196. Kofidis, T., D.R. Lebl, E.C. Martinez, G. Hoyt, M. Tanaka and R.C. Robbins, Novel

injectable bioartificial tissue facilitates targeted, less invasive, large-scale tissue

restoration on the beating heart after myocardial injury. Circulation, 2005. 112(9): p.

I173-I177.

197. Laflamme, M.A., K.Y. Chen, A.V. Naumova, V. Muskheli, J.A. Fugate, S.K. Dupras, et

al., Cardiomyocytes derived from human embryonic stem cells in pro-survival factors

enhance function of infarcted rat hearts. Nature Biotechnology, 2007. 25(9): p. 1015-1024.

198. Dobner, S., D. Bezuidenhout, P. Govender, P. Zilla and N. Davies, A synthetic non-

degradable polyethylene glycol hydrogel retards adverse post-infarct left ventricular

remodeling. J Card Fail, 2009. 15(7): p. 629-36.

199. Fujimoto, K.L., Z.W. Ma, D.M. Nelson, R. Hashizume, J.J. Guan, K. Tobita, et al.,

Synthesis, characterization and therapeutic efficacy of a biodegradable, thermoresponsive

Page 158: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

134

hydrogel designed for application in chronic infarcted myocardium. Biomaterials, 2009.

30(26): p. 4357-4368.

200. Li, X., J. Zhou, Z.Q. Liu, J. Chen, S.H. Lu, H.Y. Sun, et al., A PNIPAAm-based

thermosensitive hydrogel containing SWCNTs for stem cell transplantation in myocardial

repair. Biomaterials, 2014. 35(22): p. 5679-5688.

201. Atzet, S., S. Curtin, P. Trinh, S. Bryant and B. Ratner, Degradable Poly(2-hydroxyethyl

methacrylate)-co-polycaprolactone Hydrogels for Tissue Engineering Scaffolds.

Biomacromolecules, 2008. 9(12): p. 3370-3377.

202. Furth, M.E., A. Atala and M.E. Van Dyke, Smart biomaterials design for tissue

engineering and regenerative medicine. Biomaterials, 2007. 28(34): p. 5068-5073.

203. Bouten, C.V.C., P.Y.W. Dankers, A. Driessen-Mol, S. Pedron, A.M.A. Brizard and F.P.T.

Baaijens, Substrates for cardiovascular tissue engineering. Advanced Drug Delivery

Reviews, 2011. 63(4-5): p. 221-241.

204. Reis, L.A., L.L. Chiu, N. Feric, L. Fu and M. Radisic, Biomaterials in myocardial tissue

engineering. J Tissue Eng Regen Med, 2016. 10(1): p. 11-28.

205. Zhao, Y., N.T. Feric, N. Thavandiran, S.S. Nunes and M. Radisic, The role of tissue

engineering and biomaterials in cardiac regenerative medicine. Can J Cardiol, 2014.

30(11): p. 1307-22.

206. Chan, B.P. and K.W. Leong, Scaffolding in tissue engineering: general approaches and

tissue-specific considerations. Eur Spine J, 2008. 17 Suppl 4: p. 467-79.

207. Naderi, H., M.M. Matin and A.R. Bahrami, Review paper: critical issues in tissue

engineering: biomaterials, cell sources, angiogenesis, and drug delivery systems. J

Biomater Appl, 2011. 26(4): p. 383-417.

208. Radisic, M., H. Park, S. Gerecht, C. Cannizzaro, R. Langer and G. Vunjak-Novakovic,

Biomimetic approach to cardiac tissue engineering. Philos Trans R Soc Lond B Biol Sci,

2007. 362(1484): p. 1357-68.

209. Coirault, C., J.L. Samuel, D. Chemla, J.C. Pourny, F. Lambert, F. Marotte, et al.,

Increased compliance in diaphragm muscle of the cardiomyopathic Syrian hamster. J

Appl Physiol (1985), 1998. 85(5): p. 1762-9.

210. Omens, J.H., Stress and strain as regulators of myocardial growth. Progress in Biophysics

& Molecular Biology, 1998. 69(2-3): p. 559-572.

211. Weis, S.M., J.L. Emery, K.D. Becker, D.J. McBride, Jr., J.H. Omens and A.D.

McCulloch, Myocardial mechanics and collagen structure in the osteogenesis imperfecta

murine (oim). Circ Res, 2000. 87(8): p. 663-9.

212. Nagueh, S.F., G. Shah, Y. Wu, G. Torre-Amione, N.M. King, S. Lahmers, et al., Altered

titin expression, myocardial stiffness, and left ventricular function in patients with dilated

cardiomyopathy. Circulation, 2004. 110(2): p. 155-62.

213. Nelson, D.M., Z.W. Ma, K.L. Fujimoto, R. Hashizume and W.R. Wagner, Intra-

myocardial biomaterial injection therapy in the treatment of heart failure: Materials,

outcomes and challenges. Acta Biomaterialia, 2011. 7(1): p. 1-15.

Page 159: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

135

214. Chuong, C.J., M.S. Sacks, G. Templeton, F. Schwiep and R.L. Johnson, Regional

Deformation and Contractile Function in Canine Right Ventricular Free Wall. American

Journal of Physiology, 1991. 260(4): p. H1224-H1235.

215. Sacks, M.S. and C.J. Chuong, Biaxial Mechanical-Properties of Passive Right-

Ventricular Free Wall Myocardium. Journal of Biomechanical Engineering-Transactions

of the Asme, 1993. 115(2): p. 202-205.

216. Rappaport, D., D. Adam, P. Lysyansky and S. Riesner, Assessment of myocardial

regional strain and strain rate by tissue tracking in B-mode echocardiograms. Ultrasound

Med Biol, 2006. 32(8): p. 1181-92.

217. Demer, L.L. and F.C. Yin, Passive biaxial mechanical properties of isolated canine

myocardium. J Physiol, 1983. 339: p. 615-30.

218. Chuong, C.J., M.S. Sacks, G. Templeton, F. Schwiep and R.L. Johnson, Jr., Regional

deformation and contractile function in canine right ventricular free wall. Am J Physiol,

1991. 260(4 Pt 2): p. H1224-35.

219. Holmes, J.W., T.K. Borg and J.W. Covell, Structure and mechanics of healing

myocardial infarcts. Annu Rev Biomed Eng, 2005. 7: p. 223-53.

220. Engelmayr, G.C., Jr., M. Cheng, C.J. Bettinger, J.T. Borenstein, R. Langer and L.E.

Freed, Accordion-like honeycombs for tissue engineering of cardiac anisotropy. Nat

Mater, 2008. 7(12): p. 1003-10.

221. Fomovsky, G.M., J.R. Macadangdang, G. Ailawadi and J.W. Holmes, Model-based

design of mechanical therapies for myocardial infarction. J Cardiovasc Transl Res, 2011.

4(1): p. 82-91.

222. Fomovsky, G.M., S.A. Clark, K.M. Parker, G. Ailawadi and J.W. Holmes, Anisotropic

reinforcement of acute anteroapical infarcts improves pump function. Circ Heart Fail,

2012. 5(4): p. 515-22.

223. Jawad, H., N.N. Ali, A.R. Lyon, Q.Z. Chen, S.E. Harding and A.R. Boccaccini,

Myocardial tissue engineering: a review. J Tissue Eng Regen Med, 2007. 1(5): p. 327-42.

224. Anderson, J.M., Biological responses to materials. Annual Review of Materials Research,

2001. 31: p. 81-110.

225. Baudis, S., F. Nehl, S.C. Ligon, A. Nigisch, H. Bergmeister, D. Bernhard, et al.,

Elastomeric degradable biomaterials by photopolymerization-based CAD-CAM for

vascular tissue engineering. Biomed Mater, 2011. 6(5): p. 055003.

226. Anderson, J.M. and M.S. Shive, Biodegradation and biocompatibility of PLA and PLGA

microspheres. Advanced Drug Delivery Reviews, 2012. 64: p. 72-82.

227. Franz, S., S. Rammelt, D. Scharnweber and J.C. Simon, Immune responses to implants -

a review of the implications for the design of immunomodulatory biomaterials.

Biomaterials, 2011. 32(28): p. 6692-709.

228. Anderson, J.M., A. Rodriguez and D.T. Chang, Foreign body reaction to biomaterials.

Seminars in Immunology, 2008. 20(2): p. 86-100.

Page 160: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

136

229. Palmer, J.A., K.M. Abberton, G.M. Mitchell and W.A. Morrison, Macrophage Phenotype

in Response to Implanted Synthetic Scaffolds: An Immunohistochemical Study in the Rat.

Cells Tissues Organs, 2014. 199(2-3): p. 169-183.

230. Griffith, L.G. and G. Naughton, Tissue engineering - Current challenges and expanding

opportunities. Science, 2002. 295(5557): p. 1009-+.

231. Montgomery, M., B.Y. Zhang and M. Radisic, Cardiac Tissue Vascularization: From

Angiogenesis to Microfluidic Blood Vessels. Journal of Cardiovascular Pharmacology

and Therapeutics, 2014. 19(4): p. 382-393.

232. Madden, L.R., D.J. Mortisen, E.M. Sussman, S.K. Dupras, J.A. Fugate, J.L. Cuy, et al.,

Proangiogenic scaffolds as functional templates for cardiac tissue engineering.

Proceedings of the National Academy of Sciences of the United States of America, 2010.

107(34): p. 15211-15216.

233. Chen, Q.Z., S.L. Liang and G.A. Thouas, Elastomeric biomaterials for tissue engineering.

Progress in Polymer Science, 2013. 38(3-4): p. 584-671.

234. Tran, R.T., P. Thevenot, D. Gyawali, J.C. Chiao, L.P. Tang and J. Yang, Synthesis and

characterization of a biodegradable elastomer featuring a dual crosslinking mechanism.

Soft Matter, 2010. 6(11): p. 2449-2461.

235. Eming, S.A., P. Martin and M. Tomic-Canic, Wound repair and regeneration:

mechanisms, signaling, and translation. Sci Transl Med, 2014. 6(265): p. 265sr6.

236. Kirsner, R.S. and W.H. Eaglstein, The wound healing process. Dermatologic clinics,

1993. 11(4): p. 629-40.

237. Nelson, R.L., K. Thomas, J. Morgan and A. Jones, Non surgical therapy for anal fissure.

Cochrane Database Syst Rev, 2012. 2: p. CD003431.

238. Brem, H. and M. Tomic-Canic, Cellular and molecular basis of wound healing in diabetes.

The Journal of Clinical Investigation, 2007. 117(5): p. 1219-1222.

239. Tack, C.J., P.J. van Gurp, C. Holmes and D.S. Goldstein, Local sympathetic denervation

in painful diabetic neuropathy. Diabetes, 2002. 51(12): p. 3545-3553.

240. Loomans, C.J., E.J. de Koning, F.J. Staal, M.B. Rookmaaker, C. Verseyden, H.C. de

Boer, et al., Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis

of vascular complications of type 1 diabetes. Diabetes, 2004. 53(1): p. 195-9.

241. Gallagher, K.A., Z.J. Liu, M. Xiao, H. Chen, L.J. Goldstein, D.G. Buerk, et al., Diabetic

impairments in NO-mediated endothelial progenitor cell mobilization and homing are

reversed by hyperoxia and SDF-1 alpha. J Clin Invest, 2007. 117(5): p. 1249-59.

242. Soneja, A., M. Drews and T. Malinski, Role of nitric oxide, nitroxidative and oxidative

stress in wound healing. Pharmacol Rep, 2005. 57 Suppl: p. 108-19.

243. Brownlee, M., Advanced Protein Glycosylation in Diabetes and Aging. Annual Review of

Medicine, 1995. 46: p. 223-234.

244. Signorelli, S.S., G. Malaponte, M. Libra, L. Di Pino, G. Celotta, V. Bevelacqua, et al.,

Plasma levels and zymographic activities of matrix metalloproteinases 2 and 9 in type II

Page 161: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

137

diabetics with peripheral arterial disease. Vascular medicine (London, England), 2005.

10(1): p. 1-6.

245. Hasan, A., H. Murata, A. Falabella, S. Ochoa, L. Zhou, E. Badiavas, et al., Dermal

fibroblasts from venous ulcers are unresponsive to the action of transforming growth

factor-beta 1. Journal of dermatological science, 1997. 16(1): p. 59-66.

246. Agren, M.S., H.H. Steenfos, S. Dabelsteen, J.B. Hansen and E. Dabelsteen, Proliferation

and mitogenic response to PDGF-BB of fibroblasts isolated from chronic venous leg

ulcers is ulcer-age dependent. The Journal of investigative dermatology, 1999. 112(4): p.

463-9.

247. Loot, M.A.M., S.B. Kenter, F.L. Au, W.J.M. van Galen, E. Middelkoop, J.D. Bos, et al.,

Fibroblasts derived from chronic diabetic ulcers differ in their response to stimulation

with EGF, IGF-I, bFGF and PDGF-AB compared to controls. European journal of cell

biology, 2002. 81(3): p. 153-60.

248. Stanley, A.C., N.N. Fernandez, K.M. Lounsbury, K. Corrow, T. Osler, C. Healey, et al.,

Pressure-induced cellular senescence: a mechanism linking venous hypertension to

venous ulcers. The Journal of surgical research, 2005. 124(1): p. 112-7.

249. Zykova, S.N., T.G. Jenssen, M. Berdal, R. Olsen, R. Myklebust and R. Seljelid, Altered

cytokine and nitric oxide secretion in vitro by macrophages from diabetic type II-like

db/db mice. Diabetes, 2000. 49(9): p. 1451-8.

250. Loots, M.A., E.N. Lamme, J. Zeegelaar, J.R. Mekkes, J.D. Bos and E. Middelkoop,

Differences in cellular infiltrate and extracellular matrix of chronic diabetic and venous

ulcers versus acute wounds. The Journal of investigative dermatology, 1998. 111(5): p.

850-7.

251. Lan, C.C., C.S. Wu, H.Y. Kuo, S.M. Huang and G.S. Chen, Hyperglycaemic conditions

hamper keratinocyte locomotion via sequential inhibition of distinct pathways: new

insights on poor wound closure in patients with diabetes. Br J Dermatol, 2009. 160(6): p.

1206-14.

252. Xu, K.P., Y. Li, A.V. Ljubimov and F.S. Yu, High glucose suppresses epidermal growth

factor receptor/phosphatidylinositol 3-kinase/Akt signaling pathway and attenuates

corneal epithelial wound healing. Diabetes, 2009. 58(5): p. 1077-85.

253. Spravchikov, N., G. Sizyakov, M. Gartsbein, D. Accili, T. Tennenbaum and E.

Wertheimer, Glucose effects on skin keratinocytes: implications for diabetes skin

complications. Diabetes, 2001. 50(7): p. 1627-35.

254. Deveci, M., R.R. Gilmont, W.R. Dunham, B.P. Mudge, D.J. Smith and C.L. Marcelo,

Glutathione enhances fibroblast collagen contraction and protects keratinocytes from

apoptosis in hyperglycaemic culture. Br J Dermatol, 2005. 152(2): p. 217-24.

255. Winter, G.D., Formation of the scab and the rate of epithelization of superficial wounds

in the skin of the young domestic pig. Nature, 1962. 193: p. 293-4.

256. Hinman, C.D. and H. Maibach, Effect of Air Exposure and Occlusion on Experimental

Human Skin Wounds. Nature, 1963. 200: p. 377-8.

Page 162: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

138

257. Fan, K., J. Tang, J. Escandon and R.S. Kirsner, State of the art in topical wound-healing

products. Plastic and reconstructive surgery, 2011. 127 Suppl 1: p. 44S-59S.

258. Pawar, H.V., J. Tetteh and J.S. Boateng, Preparation, optimisation and characterisation of

novel wound healing film dressings loaded with streptomycin and diclofenac. Colloids

and Surfaces B-Biointerfaces, 2013. 102: p. 102-110.

259. Aoyagi, S., H. Onishi and Y. Machida, Novel chitosan wound dressing loaded with

minocycline for the treatment of severe burn wounds. Int J Pharm, 2007. 330(1-2): p.

138-45.

260. Stinner, D.J., S.P. Noel, W.O. Haggard, J.T. Watson and J.C. Wenke, Local Antibiotic

Delivery Using Tailorable Chitosan Sponges: The Future of Infection Control? Journal of

Orthopaedic Trauma, 2010. 24(9): p. 592-597.

261. Labovitiadi, O., K. Matthews and A. Lamb, Modified Franz diffusion for the in-vitro

determination of the efficacy of antimicrobial wafers against methicillin-resistant

Staphylococcus aureus. Journal of Pharmacy and Pharmacology, 2009. 61: p. A19-A20.

262. Unnithan, A.R., N.A.M. Barakat, P.B.T. Pichiah, G. Gnanasekaran, R. Nirmala, Y.S. Cha,

et al., Wound-dressing materials with antibacterial activity from electrospun

polyurethane-dextran nanofiber mats containing ciprofloxacin HCl. Carbohydrate

Polymers, 2012. 90(4): p. 1786-1793.

263. Sinha, M., R.M. Banik, C. Haldar and P. Maiti, Development of ciprofloxacin

hydrochloride loaded poly(ethylene glycol)/chitosan scaffold as wound dressing. Journal

of Porous Materials, 2013. 20(4): p. 799-807.

264. Schairer, D.O., J.S. Chouake, J.D. Nosanchuk and A.J. Friedman, The potential of nitric

oxide releasing therapies as antimicrobial agents. Virulence, 2012. 3(3): p. 271-279.

265. Efem, S.E., Clinical observations on the wound healing properties of honey. Br J Surg,

1988. 75(7): p. 679-81.

266. Ranzato, E., S. Martinotti and B. Burlando, Epithelial mesenchymal transition traits in

honey-driven keratinocyte wound healing: Comparison among different honeys. Wound

Repair and Regeneration, 2012. 20(5): p. 778-785.

267. Holmes, C., J.S. Wrobel, M.P. Maceachern and B.R. Boles, Collagen-based wound

dressings for the treatment of diabetes-related foot ulcers: a systematic review. Diabetes

Metab Syndr Obes, 2013. 6: p. 17-29.

268. Czaja, W.K., D.J. Young, M. Kawecki and R.M. Brown, The future prospects of

microbial cellulose in biomedical applications. Biomacromolecules, 2007. 8(1): p. 1-12.

269. Seetharaman, S., S. Natesan, R.S. Stowers, C. Mullens, D.G. Baer, L.J. Suggs, et al., A

PEGylated fibrin-based wound dressing with antimicrobial and angiogenic activity. Acta

Biomater, 2011. 7(7): p. 2787-96.

270. Kanokpanont, S., S. Damrongsakkul, J. Ratanavaraporn and P. Aramwit, An innovative

bi-layered wound dressing made of silk and gelatin for accelerated wound healing.

International Journal of Pharmaceutics, 2012. 436(1-2): p. 141-153.

Page 163: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

139

271. Dongargaonkar, A.A., G.L. Bowlin and H. Yang, Electrospun Blends of Gelatin and

Gelatin-Dendrimer Conjugates As a Wound-Dressing and Drug-Delivery Platform.

Biomacromolecules, 2013. 14(11): p. 4038-4045.

272. Sayag, J., S. Meaume and S. Bohbot, Healing properties of calcium alginate dressings. J

Wound Care, 1996. 5(8): p. 357-62.

273. Banerjee, J., P. Das Ghatak, S. Roy, S. Khanna, E.K. Sequin, K. Bellman, et al.,

Improvement of human keratinocyte migration by a redox active bioelectric dressing.

PLoS One, 2014. 9(3): p. e89239.

274. Chester, D.L., D.S. Balderson and R.P. Papini, A review of keratinocyte delivery to the

wound bed. J Burn Care Rehabil, 2004. 25(3): p. 266-75.

275. Nuschke, A., Activity of mesenchymal stem cells in therapies for chronic skin wound

healing. Organogenesis, 2014. 10(1): p. 29-37.

276. Kondo, S. and Y. Kuroyanagi, Development of a wound dressing composed of

hyaluronic acid and collagen sponge with epidermal growth factor. J Biomater Sci Polym

Ed, 2012. 23(5): p. 629-43.

277. Xie, Z.W., C.B. Paras, H. Weng, P. Punnakitikashem, L.C. Su, K. Vu, et al., Dual growth

factor releasing multi-functional nanofibers for wound healing. Acta Biomaterialia, 2013.

9(12): p. 9351-9359.

278. Yang, Y., T. Xia, W. Zhi, L. Wei, J. Weng, C. Zhang, et al., Promotion of skin

regeneration in diabetic rats by electrospun core-sheath fibers loaded with basic fibroblast

growth factor. Biomaterials, 2011. 32(18): p. 4243-4254.

279. Puolakkainen, P.A., D.R. Twardzik, J.E. Ranchalis, S.C. Pankey, M.J. Reed and W.R.

Gombotz, The Enhancement in Wound-Healing by Transforming Growth Factor-Beta(1)

(Tgf-Beta(1)) Depends on the Topical Delivery System. Journal of Surgical Research,

1995. 58(3): p. 321-329.

280. Cho, C.-H., R.A. Kammerer, H.J. Lee, M.O. Steinmetz, Y.S. Ryu, S.H. Lee, et al.,

COMP-Ang1: a designed angiopoietin-1 variant with nonleaky angiogenic activity.

Proceedings of the National Academy of Sciences of the United States of America, 2004.

101(15): p. 5547-52.

281. Tokunou, T., R. Miller, P. Patwari, M.E. Davis, V.F. Segers, A.J. Grodzinsky, et al.,

Engineering insulin-like growth factor-1 for local delivery. FASEB J, 2008. 22(6): p.

1886-93.

282. Martino, M.M., F. Tortelli, M. Mochizuki, S. Traub, D. Ben-David, G.A. Kuhn, et al.,

Engineering the Growth Factor Microenvironment with Fibronectin Domains to Promote

Wound and Bone Tissue Healing. Science Translational Medicine, 2011. 3(100): p.

100ra89.

283. Sheffield, P., Tissue oxygen measurements, in Problem Wounds: The Role of Oxygen.

1988, Elsevier: New York. p. 17-52.

284. Brimson, C.H. and Y. Nigam, The role of oxygen-associated therapies for the healing of

chronic wounds, particularly in patients with diabetes. J Eur Acad Dermatol Venereol,

2013. 27(4): p. 411-8.

Page 164: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

140

285. Fischer, B.H., Topical Hyperbaric Oxygen Treatment of Pressure Sores and Skin Ulcers.

Lancet, 1969. 2(7617): p. 405-9.

286. Rabkin, J. and T. Hunt, Infection and oxygen, in Problem Wounds: The Role of Oxygen.

1988, Elsevier: New York. p. 1-16.

287. Gallagher, K.A., Z.J. Liu, M. Xiao, H.Y. Chen, L.J. Goldstein, D.G. Buerk, et al.,

Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and

homing are reversed by hyperoxia and SDF-1 alpha. Journal of Clinical Investigation,

2007. 117(5): p. 1249-1259.

288. Brismar, K., F. Lind and G. Kratz, Dose-dependent hyperbaric oxygen stimulation of

human fibroblast proliferation. Wound Repair Regen, 1997. 5(2): p. 147-50.

289. Hartmann, M., K. Jonsson and B. Zederfeldt, Effect of tissue perfusion and oxygenation

on accumulation of collagen in healing wounds. Randomized study in patients after major

abdominal operations. Eur J Surg, 1992. 158(10): p. 521-6.

290. Tandara, A.A. and T.A. Mustoe, Oxygen in wound healing - More than a nutrient. World

Journal of Surgery, 2004. 28(3): p. 294-300.

291. Wang, G.L., B.H. Jiang, E.A. Rue and G.L. Semenza, Hypoxia-Inducible Factor-1 Is a

Basic-Helix-Loop-Helix-Pas Heterodimer Regulated by Cellular O-2 Tension.

Proceedings of the National Academy of Sciences of the United States of America, 1995.

92(12): p. 5510-5514.

292. Botusan, I.R., V.G. Sunkari, O. Savu, A.I. Catrina, J. Grunler, S. Lindberg, et al.,

Stabilization of HIF-1 alpha is critical to improve wound healing in diabetic mice.

Proceedings of the National Academy of Sciences of the United States of America, 2008.

105(49): p. 19426-19431.

293. Palmer, R.M., A.G. Ferrige and S. Moncada, Nitric oxide release accounts for the

biological activity of endothelium-derived relaxing factor. Nature, 1987. 327(6122): p.

524-6.

294. Friedman, A. and J. Friedman, New biomaterials for the sustained release of nitric oxide:

past, present and future. Expert opinion on drug delivery, 2009. 6(10): p. 1113-22.

295. Luo, J.D. and A.F. Chen, Nitric oxide: a newly discovered function on wound healing.

Acta Pharmacol Sin, 2005. 26(3): p. 259-64.

296. Hevel, J.M., K.A. White and M.A. Marletta, Purification of the Inducible Murine

Macrophage Nitric-Oxide Synthase - Identification as a Flavoprotein. Journal of

Biological Chemistry, 1991. 266(34): p. 22789-22791.

297. Mayer, B., M. John, B. Heinzel, E.R. Werner, H. Wachter, G. Schultz, et al., Brain nitric

oxide synthase is a biopterin- and flavin-containing multi-functional oxido-reductase.

FEBS Lett, 1991. 288(1-2): p. 187-91.

298. Stuehr, D.J., H.J. Cho, N.S. Kwon, M.F. Weise and C.F. Nathan, Purification and

characterization of the cytokine-induced macrophage nitric oxide synthase: an FAD- and

FMN-containing flavoprotein. Proceedings of the National Academy of Sciences of the

United States of America, 1991. 88(17): p. 7773-7.

Page 165: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

141

299. Andrew, P.J. and B. Mayer, Enzymatic function of nitric oxide synthases. Cardiovasc

Res, 1999. 43(3): p. 521-31.

300. RomeroGraillet, C., E. Aberdam, N. Biagoli, W. Massabni, J.P. Ortonne and R. Ballotti,

Ultraviolet B radiation acts through the nitric oxide and cGMP signal transduction

pathway to stimulate melanogenesis in human melanocytes. Journal of Biological

Chemistry, 1996. 271(45): p. 28052-28056.

301. Baudouin, J.E. and P. Tachon, Constitutive nitric oxide synthase is present in normal

human keratinocytes. The Journal of investigative dermatology, 1996. 106(3): p. 428-31.

302. Wang, R.J., A. Ghahary, Y.J. Shen, P.G. Scott and E.E. Tredget, Human dermal

fibroblasts produce nitric oxide and express both constitutive and inducible nitric oxide

synthase isoforms. Journal of Investigative Dermatology, 1996. 106(3): p. 419-427.

303. Shimizu, Y., M. Sakai, Y. Umemura and H. Ueda, Immunohistochemical localization of

nitric oxide synthase in normal human skin: expression of endothelial-type and inducible-

type nitric oxide synthase in keratinocytes. The Journal of dermatology, 1997. 24(2): p.

80-7.

304. Sirsjo, A., M. Karlsson, A. Gidlof, O. Rollman and H. Torma, Increased expression of

inducible nitric oxide synthase in psoriatic skin and cytokine-stimulated cultured

keratinocytes. Br J Dermatol, 1996. 134(4): p. 643-8.

305. Bruch-Gerharz, D., K. Fehsel, C. Suschek, G. Michel, T. Ruzicka and V. Kolb-Bachofen,

A proinflammatory activity of interleukin 8 in human skin: expression of the inducible

nitric oxide synthase in psoriatic lesions and cultured keratinocytes. The Journal of

experimental medicine, 1996. 184(5): p. 2007-12.

306. Kuhn, A., K. Fehsel, P. Lehmann, J. Krutmann, T. Ruzicka and V. Kolb-Bachofen,

Aberrant timing in epidermal expression of inducible nitric oxide synthase after UV

irradiation in cutaneous lupus erythematosus. The Journal of investigative dermatology,

1998. 111(1): p. 149-53.

307. Schaffer, M.R., U. Tantry, S.S. Gross, H.L. Wasserkrug and A. Barbul, Nitric oxide

regulates wound healing. Journal of Surgical Research, 1996. 63(1): p. 237-240.

308. Frank, S., M. Madlener, J. Pfeilschifter and S. Werner, Induction of inducible nitric oxide

synthase and its corresponding tetrahydrobiopterin-cofactor-synthesizing enzyme GTP-

cyclohydrolase I during cutaneous wound repair. The Journal of investigative

dermatology, 1998. 111(6): p. 1058-64.

309. Luo, J.D., Y.Y. Wang, W.L. Fu, J. Wu and A.F. Chen, Gene therapy of endothelial nitric

oxide synthase and manganese superoxide dismutase restores delayed wound healing in

type 1 diabetic mice. Circulation, 2004. 110(16): p. 2484-2493.

310. Boissel, J.P., D. Ohly, M. Bros, U. Godtel-Armbrust, U. Forstermann and S. Frank, The

neuronal nitric oxide synthase is upregulated in mouse skin repair and in response to

epidermal growth factor in human HaCaT keratinocytes. Journal of Investigative

Dermatology, 2004. 123(1): p. 132-139.

311. Yamasaki, K., H.D.J. Edington, C. McClosky, E. Tzeng, A. Lizonova, I. Kovesdi, et al.,

Reversal of impaired wound repair in iNOS-deficient mice by topical adenoviral-

mediated iNOS gene transfer. Journal of Clinical Investigation, 1998. 101(5): p. 967-971.

Page 166: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

142

312. Lee, P.C., A.N. Salyapongse, G.A. Bragdon, L.L. Shears, S.C. Watkins, H.D.J. Edington,

et al., Impaired wound healing and angiogenesis in eNOS-deficient mice. American

Journal of Physiology-Heart and Circulatory Physiology, 1999. 277(4): p. H1600-H1608.

313. Schäffer, M.R., U. Tantry, P.A. Efron, G.M. Ahrendt, F.J. Thornton and A. Barbul,

Diabetes-impaired healing and reduced wound nitric oxide synthesis: a possible

pathophysiologic correlation. Surgery, 1997. 121(5): p. 513-9.

314. Stallmeyer, B., M. Anhold, C. Wetzler, K. Kahlina, J. Pfeilschifter and S. Frank,

Regulation of eNOS in normal and diabetes-impaired skin repair: implications for tissue

regeneration. Nitric oxide : biology and chemistry / official journal of the Nitric Oxide

Society, 2002. 6(2): p. 168-77.

315. Andrew, P.J., H. Harant and I.J.D. Lindley, Nitric-Oxide Regulates Il-8 Expression in

Melanoma-Cells at the Transcriptional Level. Biochemical and Biophysical Research

Communications, 1995. 214(3): p. 949-956.

316. Vodovotz, Y., L. Chesler, H. Chong, S.J. Kim, J.T. Simpson, W. DeGraff, et al.,

Regulation of transforming growth factor beta1 by nitric oxide. Cancer research, 1999.

59(9): p. 2142-9.

317. Belenky, S.N., R.A. Robbins and I. Rubinstein, Nitric oxide synthase inhibitors attenuate

human monocyte chemotaxis in vitro. Journal of leukocyte biology, 1993. 53(5): p. 498-

503.

318. Ziche, M., L. Morbidelli, R. Choudhuri, H.T. Zhang, S. Donnini, H.J. Granger, et al.,

Nitric oxide synthase lies downstream from vascular endothelial growth factor-induced

but not basic fibroblast growth factor-induced angiogenesis. The Journal of clinical

investigation, 1997. 99(11): p. 2625-34.

319. Ziche, M., L. Morbidelli, E. Masini, S. Amerini, H.J. Granger, C.A. Maggi, et al., Nitric

oxide mediates angiogenesis in vivo and endothelial cell growth and migration in vitro

promoted by substance P. The Journal of clinical investigation, 1994. 94(5): p. 2036-44.

320. Stallmeyer, B., H. Kampfer, N. Kolb, J. Pfeilschifter and S. Frank, The function of nitric

oxide in wound repair: inhibition of inducible nitric oxide-synthase severely impairs

wound reepithelialization. J Invest Dermatol, 1999. 113(6): p. 1090-8.

321. Seo, S.J., H.G. Choi, H.J. Chung and C.K. Hong, Time course of expression of mRNA of

inducible nitric oxide synthase and generation of nitric oxide by ultraviolet B in

keratinocyte cell lines. The British journal of dermatology, 2002. 147(4): p. 655-62.

322. Schaffer, M.R., P.A. Efron, F.J. Thornton, K. Klingel, S.S. Gross and A. Barbul, Nitric

oxide, an autocrine regulator of wound fibroblast synthetic function. J Immunol, 1997.

158(5): p. 2375-81.

323. Thornton, F.J., M.R. Schaffer, M.B. Witte, L.L. Moldawer, S.L. MacKay, A. Abouhamze,

et al., Enhanced collagen accumulation following direct transfection of the inducible

nitric oxide synthase gene in cutaneous wounds. Biochem Biophys Res Commun, 1998.

246(3): p. 654-9.

324. Han, G., L.N. Nguyen, C. Macherla, Y.L. Chi, J.M. Friedman, J.D. Nosanchuk, et al.,

Nitric Oxide-Releasing Nanoparticles Accelerate Wound Healing by Promoting

Page 167: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

143

Fibroblast Migration and Collagen Deposition. American Journal of Pathology, 2012.

180(4): p. 1465-1473.

325. Seabra, A.B., A. Fitzpatrick, J. Paul, M.G. De Oliveira and R. Weller, Topically applied

S-nitrosothiol-containing hydrogels as experimental and pharmacological nitric oxide

donors in human skin. The British journal of dermatology, 2004. 151(5): p. 977-83.

326. Li, Y. and P.I. Lee, Controlled nitric oxide delivery platform based on S-nitrosothiol

conjugated interpolymer complexes for diabetic wound healing. Molecular

pharmaceutics, 2010. 7(1): p. 254-66.

327. Masters, K.S.B., S.J. Leibovich, P. Belem, J.L. West and L.A. Poole-Warren, Effects of

nitric oxide releasing poly(vinyl alcohol) hydrogel dressings on dermal wound healing in

diabetic mice. Wound Repair and Regeneration, 2002. 10(5): p. 286-94.

328. Lowe, A., J. Bills, R. Verma, L. Lavery, K. Davis and K.J. Balkus, Jr., Electrospun nitric

oxide releasing bandage with enhanced wound healing. Acta Biomater, 2015. 13: p. 121-

30.

329. Martinez, L.R., G. Han, M. Chacko, M.R. Mihu, M. Jacobson, P. Gialanella, et al.,

Antimicrobial and healing efficacy of sustained release nitric oxide nanoparticles against

Staphylococcus aureus skin infection. The Journal of investigative dermatology, 2009.

129(10): p. 2463-9.

330. Blecher, K., L.R. Martinez, C. Tuckman-Vernon, P. Nacharaju, D. Schairer, J. Chouake,

et al., Nitric oxide-releasing nanoparticles accelerate wound healing in NOD-SCID mice.

Nanomedicine : nanotechnology, biology, and medicine, 2012.

331. Branski, L.K., G.G. Gauglitz, D.N. Herndon and M.G. Jeschke, A review of gene and

stem cell therapy in cutaneous wound healing. Burns, 2009. 35(2): p. 171-80.

332. Herweijer, H. and J.A. Wolff, Progress and prospects: naked DNA gene transfer and

therapy. Gene Ther, 2003. 10(6): p. 453-8.

333. McCaffrey, A.P., L. Meuse, T.T.T. Pham, D.S. Conklin, G.J. Hannon and M.A. Kay,

Gene expression - RNA interference in adult mice. Nature, 2002. 418(6893): p. 38-39.

334. Scholz, C. and E. Wagner, Therapeutic plasmid DNA versus siRNA delivery: common

and different tasks for synthetic carriers. J Control Release, 2012. 161(2): p. 554-65.

335. Steinstraesser, L., M.C. Lam, F. Jacobsen, P.E. Porporato, K.K. Chereddy, M. Becerikli,

et al., Skin electroporation of a plasmid encoding hCAP-18/LL-37 host defense peptide

promotes wound healing. Mol Ther, 2014. 22(4): p. 734-42.

336. Albrecht-Schgoer, K., W. Schgoer, M. Theurl, U. Stanzl, D. Lener, D. Dejaco, et al.,

Topical secretoneurin gene therapy accelerates diabetic wound healing by interaction

between heparan-sulfate proteoglycans and basic FGF. Angiogenesis, 2014. 17(1): p. 27-

36.

337. Kwon, M.J., S. An, S. Choi, K. Nam, H.S. Jung, C.S. Yoon, et al., Effective healing of

diabetic skin wounds by using nonviral gene therapy based on minicircle vascular

endothelial growth factor DNA and a cationic dendrimer. Journal of Gene Medicine,

2012. 14(4): p. 272-278.

Page 168: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

144

338. Dou, C., F. Lay, A.M. Ansari, D.J. Rees, A.K. Ahmed, O. Kovbasnjuk, et al.,

Strengthening the skin with topical delivery of keratinocyte growth factor-1 using a novel

DNA plasmid. Mol Ther, 2014. 22(4): p. 752-61.

339. Pola, R., L.E. Ling, M. Silver, M.J. Corbley, M. Kearney, R.B. Pepinsky, et al., The

morphogen Sonic hedgehog is an indirect angiogenic agent upregulating two families of

angiogenic growth factors. Nature Medicine, 2001. 7(6): p. 706-711.

340. Asai, J., H. Takenaka, K.F. Kusano, M. Ii, C. Luedemann, C. Curry, et al., Topical sonic

hedgehog gene therapy accelerates wound healing in diabetes by enhancing endothelial

progenitor cell-mediated microvascular remodeling. Circulation, 2006. 113(20): p. 2413-

24.

341. Park, H.J., J. Lee, M.J. Kim, T.J. Kang, Y. Jeong, S.H. Um, et al., Sonic hedgehog

intradermal gene therapy using a biodegradable poly(beta-amino esters) nanoparticle to

enhance wound healing. Biomaterials, 2012. 33(35): p. 9148-56.

342. Powell, R.J., P. Goodney, F.O. Mendelsohn, E.K. Moen, B.H. Annex and H.G.F.T.

Investigators, Safety and efficacy of patient specific intramuscular injection of HGF

plasmid gene therapy on limb perfusion and wound healing in patients with ischemic

lower extremity ulceration: results of the HGF-0205 trial. J Vasc Surg, 2010. 52(6): p.

1525-30.

343. Morishita, R., H. Makino, M. Aoki, N. Hashiya, K. Yamasaki, J. Azuma, et al., Phase

I/IIa clinical trial of therapeutic angiogenesis using hepatocyte growth factor gene

transfer to treat critical limb ischemia. Arterioscler Thromb Vasc Biol, 2011. 31(3): p.

713-20.

344. Shigematsu, H., K. Yasuda, T. Sasajima, T. Takano, T. Miyata, T. Ohta, et al.,

Transfection of human HGF plasmid DNA improves limb salvage in Buerger's disease

patients with critical limb ischemia. Int Angiol, 2011. 30(2): p. 140-9.

345. Gu, Y., J. Zhang, L. Guo, S. Cui, X. Li, D. Ding, et al., A phase I clinical study of naked

DNA expressing two isoforms of hepatocyte growth factor to treat patients with critical

limb ischemia. J Gene Med, 2011. 13(11): p. 602-10.

346. Kulkarni, M.M., U. Greiser, T. O'Brien and A. Pandit, A temporal gene delivery system

based on fibrin microspheres. Mol Pharm, 2011. 8(2): p. 439-46.

347. Yang, Y., T. Xia, F. Chen, W. Wei, C. Liu, S. He, et al., Electrospun fibers with plasmid

bFGF polyplex loadings promote skin wound healing in diabetic rats. Mol Pharm, 2012.

9(1): p. 48-58.

348. Kim, H.S. and H.S. Yoo, In vitro and in vivo epidermal growth factor gene therapy for

diabetic ulcers with electrospun fibrous meshes. Acta Biomater, 2013. 9(7): p. 7371-80.

349. Lin, A., A. Hokugo, J. Choi and I. Nishimura, Small Cytoskeleton-Associated Molecule,

Fibroblast Growth Factor Receptor 1 Oncogene Partner 2/Wound Inducible Transcript-

3.0 (FGFR1OP2/wit3.0), Facilitates Fibroblast-Driven Wound Closure. American

Journal of Pathology, 2010. 176(1): p. 108-121.

350. Li, P., P. Liu, R.P. Xiong, X.Y. Chen, Y. Zhao, W.P. Lu, et al., Ski, a modulator of

wound healing and scar formation in the rat skin and rabbit ear. J Pathol, 2011. 223(5): p.

659-71.

Page 169: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

145

351. Burnett, J.C. and J.J. Rossi, RNA-based therapeutics: current progress and future

prospects. Chem Biol, 2012. 19(1): p. 60-71.

352. Ozpolat, B., A.K. Sood and G. Lopez-Berestein, Liposomal siRNA nanocarriers for

cancer therapy. Adv Drug Deliv Rev, 2014. 66: p. 110-6.

353. Chen, L., A. Endler, K. Uchida, S. Horiguchi, Y. Morizane, O. Iijima, et al., Int6/eIF3e

Silencing Promotes Functional Blood Vessel Outgrowth and Enhances Wound Healing

by Upregulating Hypoxia-Induced Factor 2 alpha Expression. Circulation, 2010. 122(9):

p. 910-919.

354. Nguyen, P.D., J.P. Tutela, V.D. Thanik, D. Knobel, R.J. Allen, C.C. Chang, et al.,

Improved diabetic wound healing through topical silencing of p53 is associated with

augmented vasculogenic mediators. Wound Repair and Regeneration, 2010. 18(6): p.

553-559.

355. Wetterau, M., F. George, A. Weinstein, P.D. Nguyen, J.P. Tutela, D. Knobel, et al.,

Topical prolyl hydroxylase domain-2 silencing improves diabetic murine wound closure.

Wound Repair Regen, 2011. 19(4): p. 481-6.

356. Lee, J.W., J.P. Tutela, R.A. Zoumalan, V.D. Thanik, P.D. Nguyen, L. Varjabedian, et al.,

Inhibition of Smad3 expression in radiation-induced fibrosis using a novel method for

topical transcutaneous gene therapy. Arch Otolaryngol Head Neck Surg, 2010. 136(7): p.

714-9.

357. Wang, Y.W., N.H. Liou, J.H. Cherng, S.J. Chang, K.H. Ma, E. Fu, et al., siRNA-

targeting transforming growth factor-beta type I receptor reduces wound scarring and

extracellular matrix deposition of scar tissue. J Invest Dermatol, 2014. 134(7): p. 2016-25.

358. Liu, X., L. Ma, J. Liang, B. Zhang, J. Teng and C. Gao, RNAi functionalized collagen-

chitosan/silicone membrane bilayer dermal equivalent for full-thickness skin regeneration

with inhibited scarring. Biomaterials, 2013. 34(8): p. 2038-48.

359. Zhao, R., Q.T. Yan, H.L. Huang, J.Y. Lv and W.L. Ma, Transdermal siRNA-TGF beta 1-

337 patch for hypertrophic scar treatment. Matrix Biology, 2013. 32(5): p. 265-276.

360. Kim, H.S. and H.S. Yoo, Matrix metalloproteinase-inspired suicidal treatments of

diabetic ulcers with siRNA-decorated nanofibrous meshes. Gene Therapy, 2013. 20(4): p.

378-385.

361. Charafeddine, R.A., J. Makdisi, D. Schairer, B.P. O'Rourke, J.D. Diaz-Valencia, J.

Chouake, et al., Fidgetin-Like 2: A Microtubule-Based Regulator of Wound Healing. J

Invest Dermatol, 2015. 135(9): p. 2309-18.

362. Randeria, P.S., M.A. Seeger, X.Q. Wang, H. Wilson, D. Shipp, C.A. Mirkin, et al.,

siRNA-based spherical nucleic acids reverse impaired wound healing in diabetic mice by

ganglioside GM3 synthase knockdown. Proc Natl Acad Sci U S A, 2015. 112(18): p.

5573-8.

363. Novina, C.D. and P.A. Sharp, The RNAi revolution. Nature, 2004. 430(6996): p. 161-4.

364. Banerjee, J., Y.C. Chan and C.K. Sen, MicroRNAs in skin and wound healing. Physiol

Genomics, 2011. 43(10): p. 543-56.

Page 170: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

146

365. Jin, Y., S.D. Tymen, D. Chen, Z.J. Fang, Y. Zhao, D. Dragas, et al., MicroRNA-99

family targets AKT/mTOR signaling pathway in dermal wound healing. PLoS One, 2013.

8(5): p. e64434.

366. Suarez, Y., C. Fernandez-Hernando, J.S. Pober and W.C. Sessa, Dicer dependent

microRNAs regulate gene expression and functions in human endothelial cells. Circ Res,

2007. 100(8): p. 1164-73.

367. Bonauer, A., G. Carmona, M. Iwasaki, M. Mione, M. Koyanagi, A. Fischer, et al.,

MicroRNA-92a controls angiogenesis and functional recovery of ischemic tissues in mice.

Science, 2009. 324(5935): p. 1710-3.

368. Chan, Y.C., S. Roy, S. Khanna and C.K. Sen, Downregulation of endothelial microRNA-

200b supports cutaneous wound angiogenesis by desilencing GATA binding protein 2

and vascular endothelial growth factor receptor 2. Arterioscler Thromb Vasc Biol, 2012.

32(6): p. 1372-82.

369. Chan, Y.C., S. Roy, Y. Huang, S. Khanna and C.K. Sen, The microRNA miR-199a-5p

down-regulation switches on wound angiogenesis by derepressing the v-ets

erythroblastosis virus E26 oncogene homolog 1-matrix metalloproteinase-1 pathway. J

Biol Chem, 2012. 287(49): p. 41032-43.

370. Xu, J., W. Wu, L. Zhang, W. Dorset-Martin, M.W. Morris, M.E. Mitchell, et al., The role

of microRNA-146a in the pathogenesis of the diabetic wound-healing impairment:

correction with mesenchymal stem cell treatment. Diabetes, 2012. 61(11): p. 2906-12.

371. Biswas, S., S. Roy, J. Banerjee, S.R. Hussain, S. Khanna, G. Meenakshisundaram, et al.,

Hypoxia inducible microRNA 210 attenuates keratinocyte proliferation and impairs

closure in a murine model of ischemic wounds. Proc Natl Acad Sci U S A, 2010. 107(15):

p. 6976-81.

372. Wang, T., L.L. Zhang, C.M. Shi, H.Q. Sun, J.P. Wang, R. Li, et al., TGF-beta-induced

miR-21 negatively regulates the antiproliferative activity but has no effect on EMT of

TGF-beta in HaCaT cells. International Journal of Biochemistry & Cell Biology, 2012.

44(2): p. 366-376.

373. Yang, X., J. Wang, S.L. Guo, K.J. Fan, J. Li, Y.L. Wang, et al., miR-21 promotes

keratinocyte migration and re-epithelialization during wound healing. Int J Biol Sci, 2011.

7(5): p. 685-90.

374. Wang, T., Y. Feng, H. Sun, L. Zhang, L. Hao, C. Shi, et al., miR-21 regulates skin wound

healing by targeting multiple aspects of the healing process. Am J Pathol, 2012. 181(6): p.

1911-20.

375. Pastar, I., A.A. Khan, O. Stojadinovic, E.A. Lebrun, M.C. Medina, H. Brem, et al.,

Induction of specific microRNAs inhibits cutaneous wound healing. J Biol Chem, 2012.

287(35): p. 29324-35.

376. Viticchie, G., A.M. Lena, F. Cianfarani, T. Odorisio, M. Annicchiarico-Petruzzelli, G.

Melino, et al., MicroRNA-203 contributes to skin re-epithelialization. Cell Death Dis,

2012. 3: p. e435.

Page 171: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

147

377. Sundaram, G.M., J.E. Common, F.E. Gopal, S. Srikanta, K. Lakshman, D.P. Lunny, et al.,

'See-saw' expression of microRNA-198 and FSTL1 from a single transcript in wound

healing. Nature, 2013. 495(7439): p. 103-6.

378. Li, D., X. Li, A. Wang, F. Meisgen, A. Pivarcsi, E. Sonkoly, et al., MicroRNA-31

Promotes Skin Wound Healing by Enhancing Keratinocyte Proliferation and Migration. J

Invest Dermatol, 2015. 135(6): p. 1676-85.

379. Li, D., A. Wang, X. Liu, F. Meisgen, J. Grunler, I.R. Botusan, et al., MicroRNA-132

enhances transition from inflammation to proliferation during wound healing. J Clin

Invest, 2015. 125(8): p. 3008-26.

380. Caskey, R.C., C. Zgheib, M. Morris, M. Allukian, W. Dorsett-Martin, J. Xu, et al.,

Dysregulation of collagen production in diabetes following recurrent skin injury:

contribution to the development of a chronic wound. Wound Repair Regen, 2014. 22(4):

p. 515-20.

381. Kato, M., J. Zhang, M. Wang, L. Lanting, H. Yuan, J.J. Rossi, et al., MicroRNA-192 in

diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via

inhibition of E-box repressors. Proc Natl Acad Sci U S A, 2007. 104(9): p. 3432-7.

382. Maurer, B., J. Stanczyk, A. Jungel, A. Akhmetshina, M. Trenkmann, M. Brock, et al.,

MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis

Rheum, 2010. 62(6): p. 1733-43.

383. Cheng, J., Y.L. Wang, D.M. Wang and Y.N. Wu, Identification of Collagen 1 as a Post-

transcriptional Target of miR-29b in Skin Fibroblasts: Therapeutic Implication for Scar

Reduction. American Journal of the Medical Sciences, 2013. 346(2): p. 98-103.

384. Yang, L.L., J.Q. Liu, X.Z. Bai, L. Fan, F. Han, W.B. Jia, et al., Acute downregulation of

miR-155 at wound sites leads to a reduced fibrosis through attenuating inflammatory

response. Biochemical and Biophysical Research Communications, 2014. 453(1): p. 153-

159.

385. Dengler, J., H. Song, N. Thavandiran, S. Masse, G.A. Wood, K. Nanthakumar, et al.,

Engineered heart tissue enables study of residual undifferentiated embryonic stem cell

activity in a cardiac environment. Biotechnol Bioeng, 2011. 108(3): p. 704-19.

386. Song, H., C. Yoon, S.J. Kattman, J. Dengler, S. Masse, T. Thavaratnam, et al.,

Interrogating functional integration between injected pluripotent stem cell-derived cells

and surrogate cardiac tissue. Proc Natl Acad Sci U S A, 2010. 107(8): p. 3329-34.

387. Song, H., P.W. Zandstra and M. Radisic, Engineered heart tissue model of diabetic

myocardium. Tissue Eng Part A, 2011. 17(13-14): p. 1869-78.

388. Kattman, S.J., A.D. Witty, M. Gagliardi, N.C. Dubois, M. Niapour, A. Hotta, et al.,

Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac

differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell, 2011. 8(2):

p. 228-40.

389. Bauwens, C.L., H. Song, N. Thavandiran, M. Ungrin, S. Masse, K. Nanthakumar, et al.,

Geometric control of cardiomyogenic induction in human pluripotent stem cells. Tissue

Eng Part A, 2011. 17(15-16): p. 1901-9.

Page 172: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

148

390. Murry, C.E. and G. Keller, Differentiation of embryonic stem cells to clinically relevant

populations: lessons from embryonic development. Cell, 2008. 132(4): p. 661-80.

391. Lian, X., C. Hsiao, G. Wilson, K. Zhu, L.B. Hazeltine, S.M. Azarin, et al., Robust

cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation

of canonical Wnt signaling. Proc Natl Acad Sci U S A, 2012. 109(27): p. E1848-57.

392. Xu, C., S. Police, M. Hassanipour, Y. Li, Y. Chen, C. Priest, et al., Efficient generation

and cryopreservation of cardiomyocytes derived from human embryonic stem cells.

Regen Med, 2011. 6(1): p. 53-66.

393. Dubois, N.C., A.M. Craft, P. Sharma, D.A. Elliott, E.G. Stanley, A.G. Elefanty, et al.,

SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human

pluripotent stem cells. Nat Biotechnol, 2011. 29(11): p. 1011-8.

394. Uosaki, H., H. Fukushima, A. Takeuchi, S. Matsuoka, N. Nakatsuji, S. Yamanaka, et al.,

Efficient and scalable purification of cardiomyocytes from human embryonic and

induced pluripotent stem cells by VCAM1 surface expression. PLoS One, 2011. 6(8): p.

e23657.

395. Zhang, B., J.V. Green, S.K. Murthy and M. Radisic, Label-free enrichment of functional

cardiomyocytes using microfluidic deterministic lateral flow displacement. PLoS One,

2012. 7(5): p. e37619.

396. Caspi, O., I. Itzhaki, I. Kehat, A. Gepstein, G. Arbel, I. Huber, et al., In vitro

electrophysiological drug testing using human embryonic stem cell derived

cardiomyocytes. Stem Cells Dev, 2009. 18(1): p. 161-72.

397. Itzhaki, I., L. Maizels, I. Huber, L. Zwi-Dantsis, O. Caspi, A. Winterstern, et al.,

Modelling the long QT syndrome with induced pluripotent stem cells. Nature, 2011.

471(7337): p. 225-9.

398. Liang, P., F. Lan, A.S. Lee, T. Gong, V. Sanchez-Freire, Y. Wang, et al., Drug screening

using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals

disease-specific patterns of cardiotoxicity. Circulation, 2013. 127(16): p. 1677-91.

399. Karp, J.M., Y. Yeo, W. Geng, C. Cannizarro, K. Yan, D.S. Kohane, et al., A

photolithographic method to create cellular micropatterns. Biomaterials, 2006. 27(27): p.

4755-64.

400. Guo, L., R.M. Abrams, J.E. Babiarz, J.D. Cohen, S. Kameoka, M.J. Sanders, et al.,

Estimating the risk of drug-induced proarrhythmia using human induced pluripotent stem

cell-derived cardiomyocytes. Toxicol Sci, 2011. 123(1): p. 281-9.

401. Guo, L., L. Coyle, R.M. Abrams, R. Kemper, E.T. Chiao and K.L. Kolaja, Refining the

human iPSC-cardiomyocyte arrhythmic risk assessment model. Toxicol Sci, 2013. 136(2):

p. 581-94.

402. Lamore, S.D., H.W. Kamendi, C.W. Scott, Y.P. Dragan and M.F. Peters, Cellular

impedance assays for predictive preclinical drug screening of kinase inhibitor

cardiovascular toxicity. Toxicol Sci, 2013. 135(2): p. 402-13.

Page 173: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

149

403. Natarajan, A., M. Stancescu, V. Dhir, C. Armstrong, F. Sommerhage, J.J. Hickman, et al.,

Patterned cardiomyocytes on microelectrode arrays as a functional, high information

content drug screening platform. Biomaterials, 2011. 32(18): p. 4267-74.

404. Wang, J., A. Chen, D.K. Lieu, I. Karakikes, G. Chen, W. Keung, et al., Effect of

engineered anisotropy on the susceptibility of human pluripotent stem cell-derived

ventricular cardiomyocytes to arrhythmias. Biomaterials, 2013. 34(35): p. 8878-86.

405. Chen, A., E. Lee, R. Tu, K. Santiago, A. Grosberg, C. Fowlkes, et al., Integrated platform

for functional monitoring of biomimetic heart sheets derived from human pluripotent

stem cells. Biomaterials, 2014. 35(2): p. 675-83.

406. Grosberg, A., P.W. Alford, M.L. McCain and K.K. Parker, Ensembles of engineered

cardiac tissues for physiological and pharmacological study: heart on a chip. Lab Chip,

2011. 11(24): p. 4165-73.

407. Wang, G., M.L. McCain, L. Yang, A. He, F.S. Pasqualini, A. Agarwal, et al., Modeling

the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell

and heart-on-chip technologies. Nat Med, 2014. 20(6): p. 616-23.

408. Ma, Z., S. Koo, M.A. Finnegan, P. Loskill, N. Huebsch, N.C. Marks, et al., Three-

dimensional filamentous human diseased cardiac tissue model. Biomaterials, 2014. 35(5):

p. 1367-77.

409. Baar, K., R. Birla, M.O. Boluyt, G.H. Borschel, E.M. Arruda and R.G. Dennis, Self-

organization of rat cardiac cells into contractile 3-D cardiac tissue. FASEB journal, 2005.

19(2): p. 275-7.

410. Thavandiran, N., N. Dubois, A. Mikryukov, S. Masse, B. Beca, C.A. Simmons, et al.,

Design and formulation of functional pluripotent stem cell-derived cardiac microtissues.

Proc Natl Acad Sci U S A, 2013. 110(49): p. E4698-707.

411. Hansen, A., A. Eder, M. Bönstrup, M. Flato, M. Mewe, S. Schaaf, et al., Development of

a drug screening platform based on engineered heart tissue. Circulation research, 2010.

107(1): p. 35-44.

412. Boudou, T., W.R. Legant, A. Mu, M.A. Borochin, N. Thavandiran, M. Radisic, et al., A

Microfabricated Platform to Measure and Manipulate the Mechanics of Engineered

Cardiac Microtissues. Tissue engineering. Part A, 2012. 18(9-10): p. 910-9.

413. Nunes, S.S., J.W. Miklas, J. Liu, R. Aschar-Sobbi, Y. Xiao, B. Zhang, et al., Biowire: a

platform for maturation of human pluripotent stem cell–derived cardiomyocytes. Nature

Methods, 2013. 10: p. 781-7.

414. Xiao, Y., B. Zhang, H. Liu, J.W. Miklas, M. Gagliardi, A.Q. Pahnke, et al.,

Microfabricated perfusable cardiac biowire: a platform that mimics native cardiac bundle.

Lab on a Chip, 2014. 14(5): p. 869-882.

415. Sakiyama-Elbert, S.E. and J.A. Hubbell, Functional biomaterials: Design of novel

biomaterials. Annual Review of Materials Research, 2001. 31: p. 183-201.

416. Administration, U.S.F.a.D. FDA Announces New Labeling Changes for Regranex:

Product to carry boxed warning. 2008; Available from:

http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2008/ucm116909.htm.

Page 174: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

150

417. Hollister, S.J., Porous scaffold design for tissue engineering. Nat Mater, 2005. 4(7): p.

518-24.

418. Lee, K.Y. and D.J. Mooney, Hydrogels for tissue engineering. Chemical Reviews, 2001.

101(7): p. 1869-1879.

419. Xiao, Y., B. Zhang, H. Liu, J.W. Miklas, M. Gagliardi, A. Pahnke, et al., Microfabricated

perfusable cardiac biowire: a platform that mimics native cardiac bundle. Lab Chip, 2014.

14(5): p. 869-82.

420. Miklas, J.W., S.S. Nunes, A. Sofla, L.A. Reis, A. Pahnke, Y. Xiao, et al., Bioreactor for

modulation of cardiac microtissue phenotype by combined static stretch and electrical

stimulation. Biofabrication, 2014. 6(2): p. 024113.

421. Graber, M.L., D.C. Dilillo, B.L. Friedman and E. Pastorizamunoz, Characteristics of

Fluoroprobes for Measuring Intracellular Ph. Analytical Biochemistry, 1986. 156(1): p.

202-212.

422. Grover, C.N., R.E. Cameron and S.M. Best, Investigating the morphological, mechanical

and degradation properties of scaffolds comprising collagen, gelatin and elastin for use in

soft tissue engineering. Journal of the Mechanical Behavior of Biomedical Materials,

2012. 10: p. 62-74.

423. Edvardsen, T., B.L. Gerber, J. Garot, D.A. Bluemke, J.A. Lima and O.A. Smiseth,

Quantitative assessment of intrinsic regional myocardial deformation by Doppler strain

rate echocardiography in humans: validation against three-dimensional tagged magnetic

resonance imaging. Circulation, 2002. 106(1): p. 50-6.

424. D'Hooge, J., L. Herbots and G.R. Sutherland, Quantitative assessment of intrinsic

regional myocardial deformation by Doppler strain rate echocardiography in humans.

Circulation, 2003. 107(7): p. e49; author reply e49.

425. Chiu, L.L.Y. and M. Radisic, Scaffolds with covalently immobilized VEGF and

Angiopoietin-1 for vascularization of engineered tissues. Biomaterials, 2010. 31(2): p.

226-241.

426. Singer, A.J. and R.A. Clark, Cutaneous wound healing. N Engl J Med, 1999. 341(10): p.

738-46.

427. Singh, N., D.G. Armstrong and B.A. Lipsky, Preventing foot ulcers in patients with

diabetes. JAMA, 2005. 293(2): p. 217-28.

428. Stojadinovic, O., I. Pastar, S. Vukelic, M.G. Mahoney, D. Brennan, A. Krzyzanowska, et

al., Deregulation of keratinocyte differentiation and activation: a hallmark of venous

ulcers. J Cell Mol Med, 2008. 12(6B): p. 2675-90.

429. Martin, P. and J. Lewis, Actin cables and epidermal movement in embryonic wound

healing. Nature, 1992. 360(6400): p. 179-83.

430. Fernandez-Gonzalez, R. and J.A. Zallen, Wounded cells drive rapid epidermal repair in

the early Drosophila embryo. Mol Biol Cell, 2013. 24(20): p. 3227-37.

431. Spravchikov, N., G. Sizyakov, M. Gartsbein, D. Accili, T. Tennenbaum and E.

Wertheimer, Glucose effects on skin keratinocytes - Implications for diabetes skin

complications. Diabetes, 2001. 50(7): p. 1627-1635.

Page 175: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

151

432. Kumin, A., C. Huber, T. Rulicke, E. Wolf and S. Werner, Peroxiredoxin 6 is a potent

cytoprotective enzyme in the epidermis. American Journal of Pathology, 2006. 169(4): p.

1194-1205.

433. Lan, C.C.E., C.S. Wu, S.M. Huang, I.H. Wu and G.S. Chen, High-Glucose Environment

Enhanced Oxidative Stress and Increased Interleukin-8 Secretion From Keratinocytes

New Insights Into Impaired Diabetic Wound Healing. Diabetes, 2013. 62(7): p. 2530-

2538.

434. Tarnuzzer, R.W. and G.S. Schultz, Biochemical analysis of acute and chronic wound

environments. Wound Repair Regen, 1996. 4(3): p. 321-5.

435. Ismail, N.S., E.A. Pravda, D. Li, S.-C. Shih and S.M. Dallabrida, Angiopoietin-1 Reduces

H2O2-Induced Increases in Reactive Oxygen Species and Oxidative Damage to Skin

Cells. J Invest Dermatol, 2010. 130(5): p. 1307-1317.

436. Stupack, D.G. and D.A. Cheresh, Get a ligand, get a life: integrins, signaling and cell

survival. J Cell Sci, 2002. 115(Pt 19): p. 3729-38.

437. Hersel, U., C. Dahmen and H. Kessler, RGD modified polymers: biomaterials for

stimulated cell adhesion and beyond. Biomaterials, 2003. 24(24): p. 4385-415.

438. Chiu, L.L.Y. and M. Radisic, Controlled release of thymosin β4 using collagen-chitosan

composite hydrogels promotes epicardial cell migration and angiogenesis. Journal of

controlled release : official journal of the Controlled Release Society, 2011. 155(3): p.

376-85.

439. Pastar, I., O. Stojadinovic, N.C. Yin, H. Ramirez, A.G. Nusbaum, A. Sawaya, et al.,

Epithelialization in Wound Healing: A Comprehensive Review. Adv Wound Care (New

Rochelle), 2014. 3(7): p. 445-464.

440. Schafer, M. and S. Werner, Oxidative stress in normal and impaired wound repair.

Pharmacol Res, 2008. 58(2): p. 165-71.

441. Scherer, S.S., G. Pietramaggiori, J.C. Mathews, R. Chan, P. Fiorina and D.P. Orgill,

Wound healing kinetics of the genetically diabetic mouse. Wounds, 2008. 20(1): p. 18-28.

442. Hinz, B., Formation and function of the myofibroblast during tissue repair. J Invest

Dermatol, 2007. 127(3): p. 526-37.

443. Tsang, M.W., W.K. Wong, C.S. Hung, K.M. Lai, W. Tang, E.Y. Cheung, et al., Human

epidermal growth factor enhances healing of diabetic foot ulcers. Diabetes Care, 2003.

26(6): p. 1856-61.

444. Hong, J.P., H.D. Jung and Y.W. Kim, Recombinant human epidermal growth factor

(EGF) to enhance healing for diabetic foot ulcers. Ann Plast Surg, 2006. 56(4): p. 394-8;

discussion 399-400.

445. Tsuboi, R. and D.B. Rifkin, Recombinant basic fibroblast growth factor stimulates wound

healing in healing-impaired db/db mice. J Exp Med, 1990. 172(1): p. 245-51.

446. Obara, K., M. Ishihara, T. Ishizuka, M. Fujita, Y. Ozeki, T. Maehara, et al.,

Photocrosslinkable chitosan hydrogel containing fibroblast growth factor-2 stimulates

wound healing in healing-impaired db/db mice. Biomaterials, 2003. 24(20): p. 3437-44.

Page 176: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

152

447. Galiano, R.D., O.M. Tepper, C.R. Pelo, K.A. Bhatt, M. Callaghan, N. Bastidas, et al.,

Topical Vascular Endothelial Growth Factor Accelerates Diabetic Wound Healing

through Increased Angiogenesis and by Mobilizing and Recruiting Bone Marrow-

Derived Cells. The American Journal of Pathology, 2004. 164(6): p. 1935-1947.

448. Saaristo, A., T. Tammela, A. Farkkila, M. Karkkainen, E. Suominen, S. Yla-Herttuala, et

al., Vascular endothelial growth factor-C accelerates diabetic wound healing. Am J

Pathol, 2006. 169(3): p. 1080-7.

449. Bitto, A., L. Minutoli, M.R. Galeano, D. Altavilla, F. Polito, T. Fiumara, et al.,

Angiopoietin-1 gene transfer improves impaired wound healing in genetically diabetic

mice without increasing VEGF expression. Clinical Science, 2008. 114(11-12): p. 707-

718.

450. Jackson, W.M., L.J. Nesti and R.S. Tuan, Concise review: clinical translation of wound

healing therapies based on mesenchymal stem cells. Stem Cells Transl Med, 2012. 1(1): p.

44-50.

451. Mizuno, H., M. Tobita and A.C. Uysal, Concise Review: Adipose-Derived Stem Cells as

a Novel Tool for Future Regenerative Medicine. Stem Cells, 2012. 30(5): p. 804-810.

452. Sasso, F.C., D. Torella, O. Carbonara, G.M. Ellison, M. Torella, M. Scardone, et al.,

Increased vascular endothelial growth factor expression but impaired vascular endothelial

growth factor receptor signaling in the myocardium of type 2 diabetic patients with

chronic coronary heart disease. J Am Coll Cardiol, 2005. 46(5): p. 827-34.

453. Hazarika, S., A.O. Dokun, Y. Li, A.S. Popel, C.D. Kontos and B.H. Annex, Impaired

angiogenesis after hindlimb ischemia in type 2 diabetes mellitus: differential regulation of

vascular endothelial growth factor receptor 1 and soluble vascular endothelial growth

factor receptor 1. Circ Res, 2007. 101(9): p. 948-56.

454. Eaglstein, W.H., R.S. Kirsner and M.C. Robson, Food and Drug Administration (FDA)

drug approval end points for chronic cutaneous ulcer studies. Wound Repair and

Regeneration, 2012. 20(6): p. 793-796.

455. Kim, J.H., X. Serra-Picamal, D.T. Tambe, E.H.H. Zhou, C.Y. Park, M. Sadati, et al.,

Propulsion and navigation within the advancing monolayer sheet. Nature Materials, 2013.

12(9): p. 856-863.

456. Londono, C., M.J. Loureiro, B. Slater, P.B. Lucker, J. Soleas, S. Sathananthan, et al.,

Nonautonomous contact guidance signaling during collective cell migration. Proceedings

of the National Academy of Sciences, 2014.

457. Vedula, S.R., H. Hirata, M.H. Nai, A. Brugues, Y. Toyama, X. Trepat, et al., Epithelial

bridges maintain tissue integrity during collective cell migration. Nat Mater, 2014. 13(1):

p. 87-96.

458. Davis, S., T.H. Aldrich, P.F. Jones, A. Acheson, D.L. Compton, V. Jain, et al., Isolation

of Angiopoietin-1, a Ligand for the TIE2 Receptor, by Secretion-Trap Expression

Cloning. Cell, 1996. 87(7): p. 1161-1169.

459. Brindle, N.P., P. Saharinen and K. Alitalo, Signaling and functions of angiopoietin-1 in

vascular protection. Circulation Research, 2006. 98(8): p. 1014-23.

Page 177: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

153

460. Dallabrida, S.M., N. Ismail, J.R. Oberle, B.E. Himes and M.A. Rupnick, Angiopoietin-1

Promotes Cardiac and Skeletal Myocyte Survival Through Integrins. Circulation

Research, 2005. 96(4): p. e8-e24.

461. Dallabrida, S.M., N.S. Ismail, E.A. Pravda, E.M. Parodi, R. Dickie, E.M. Durand, et al.,

Integrin binding angiopoietin-1 monomers reduce cardiac hypertrophy. The FASEB

Journal, 2008. 22(8): p. 3010-3023.

462. Takahashi, K., Y. Ito, M. Morikawa, M. Kobune, J. Huang, M. Tsukamoto, et al.,

Adenoviral-delivered angiopoietin-1 reduces the infarction and attenuates the progression

of cardiac dysfunction in the rat model of acute myocardial infarction. Mol Ther, 2003.

8(4): p. 584-92.

463. Gamble, J.R., J. Drew, L. Trezise, A. Underwood, M. Parsons, L. Kasminkas, et al.,

Angiopoietin-1 is an antipermeability and anti-inflammatory agent in vitro and targets

cell junctions. Circulation Research, 2000. 87(7): p. 603-607.

464. Kim, I., S.O. Moon, S.K. Park, S.W. Chae and G.Y. Koh, Angiopoietin-1 reduces VEGF-

stimulated leukocyte adhesion to endothelial cells by reducing ICAM-1, VCAM-1, and

E-selectin expression. Circ Res, 2001. 89(6): p. 477-9.

465. Fuxe, J., S. Tabruyn, K. Colton, H. Zaid, A. Adams, P. Baluk, et al., Pericyte requirement

for anti-leak action of angiopoietin-1 and vascular remodeling in sustained inflammation.

Am J Pathol, 2011. 178(6): p. 2897-909.

466. Oike, Y., K. Yasunaga, Y. Ito, S. Matsumoto, H. Maekawa, T. Morisada, et al.,

Angiopoietin-related growth factor (AGF) promotes epidermal proliferation, remodeling,

and regeneration. Proc Natl Acad Sci U S A, 2003. 100(16): p. 9494-9.

467. Cho, C.-H., H.-K. Sung, K.-T. Kim, H.G. Cheon, G.T. Oh, H.J. Hong, et al., COMP-

angiopoietin-1 promotes wound healing through enhanced angiogenesis,

lymphangiogenesis, and blood flow in a diabetic mouse model. Proceedings of the

National Academy of Sciences of the United States of America, 2006. 103(13): p. 4946-

4951.

468. Van Slyke, P., J. Alami, D. Martin, M. Kuliszewski, H. Leong-Poi, M.V. Sefton, et al.,

Acceleration of diabetic wound healing by an angiopoietin peptide mimetic. Tissue

engineering. Part A, 2009. 15(6): p. 1269-80.

469. Rask, F., A. Mihic, L. Reis, S.M. Dallabrida, N.S. Ismail, K. Sider, et al., Hydrogels

modified with QHREDGS peptide support cardiomyocyte survival in vitro and after sub-

cutaneous implantation. Soft Matter, 2010. 6(20): p. 5089-5099.

470. Goldfinger, L.E., S.B. Hopkinson, G.W. deHart, S. Collawn, J.R. Couchman and J.C.

Jones, The alpha3 laminin subunit, alpha6beta4 and alpha3beta1 integrin coordinately

regulate wound healing in cultured epithelial cells and in the skin. J Cell Sci, 1999. 112

( Pt 16): p. 2615-29.

471. Shang, M., N. Koshikawa, S. Schenk and V. Quaranta, The LG3 module of laminin-5

harbors a binding site for integrin alpha3beta1 that promotes cell adhesion, spreading,

and migration. J Biol Chem, 2001. 276(35): p. 33045-53.

Page 178: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

154

472. Manohar, A., S.G. Shome, J. Lamar, L. Stirling, V. Iyer, K. Pumiglia, et al., Alpha 3 beta

1 integrin promotes keratinocyte cell survival through activation of a MEK/ERK

signaling pathway. Journal of cell science, 2004. 117(Pt 18): p. 4043-54.

473. Sreejalekshmi, K.G. and P.D. Nair, Biomimeticity in tissue engineering scaffolds through

synthetic peptide modifications-Altering chemistry for enhanced biological response.

Journal of Biomedical Materials Research Part A, 2011. 96a(2): p. 477-491.

474. Martinez, A., M.D. Blanco, N. Davidenko and R.E. Cameron, Tailoring chitosan/collagen

scaffolds for tissue engineering: Effect of composition and different crosslinking agents

on scaffold properties. Carbohydr Polym, 2015. 132: p. 606-19.

475. Sen, C.K., G.M. Gordillo, S. Roy, R. Kirsner, L. Lambert, T.K. Hunt, et al., Human skin

wounds: a major and snowballing threat to public health and the economy. Wound Repair

Regen, 2009. 17(6): p. 763-71.

476. Ragnarson Tennvall, G. and J. Apelqvist, Health-economic consequences of diabetic foot

lesions. Clin Infect Dis, 2004. 39 Suppl 2: p. S132-9.

477. Galiano, R.D., J. Michaels, M. Dobryansky, J.P. Levine and G.C. Gurtner, Quantitative

and reproducible murine model of excisional wound healing. Wound repair and

regeneration, 2004. 12(4): p. 485-92.

478. Hanft, J.R., R.A. Pollak, A. Barbul, C. van Gils, P.S. Kwon, S.M. Gray, et al., Phase I

trial on the safety of topical rhVEGF on chronic neuropathic diabetic foot ulcers. J

Wound Care, 2008. 17(1): p. 30-2, 34-7.

479. FDA Announces New Labeling Changes for Regranex: Product to carry boxed warning,

U.S.F.a.D. Administration, Editor. 2008.

480. Chan, R.K., P.H. Liu, G. Pietramaggiori, S.I. Ibrahim, H.B. Hechtman and D.P. Orgill,

Effect of recombinant platelet-derived growth factor (Regranex) on wound closure in

genetically diabetic mice. J Burn Care Res, 2006. 27(2): p. 202-5.

481. Gabbiani, G., The myofibroblast in wound healing and fibrocontractive diseases. The

Journal of pathology, 2003. 200(4): p. 500-3.

482. Alves, N.M. and J.F. Mano, Chitosan derivatives obtained by chemical modifications for

biomedical and environmental applications. International Journal of Biological

Macromolecules, 2008. 43(5): p. 401-414.

483. Dash, M., F. Chiellini, R.M. Ottenbrite and E. Chiellini, Chitosan-A versatile semi-

synthetic polymer in biomedical applications. Progress in Polymer Science, 2011. 36(8):

p. 981-1014.

484. Go, A.S., D. Mozaffarian, V.L. Roger, E.J. Benjamin, J.D. Berry, W.B. Borden, et al.,

Heart disease and stroke statistics--2013 update: a report from the American Heart

Association. Circulation, 2013. 127(1): p. e6-e245.

485. Radisic, M., H. Park, H. Shing, T. Consi, F.J. Schoen, R. Langer, et al., Functional

assembly of engineered myocardium by electrical stimulation of cardiac myocytes

cultured on scaffolds. Proceedings of the National Academy of Sciences, 2004. 101(52): p.

18129-34.

Page 179: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

155

486. Zimmermann, W.H., K. Schneiderbanger, P. Schubert, M. Didie, F. Munzel, J.F.

Heubach, et al., Tissue engineering of a differentiated cardiac muscle construct.

Circulation Research, 2002. 90(2): p. 223-30.

487. Shimizu, T., M. Yamato, Y. Isoi, T. Akutsu, T. Setomaru, K. Abe, et al., Fabrication of

Pulsatile Cardiac Tissue Grafts Using a Novel 3-Dimensional Cell Sheet Manipulation

Technique and Temperature-Responsive Cell Culture Surfaces. Circulation Research,

2002. 90(3): p. e40-e48.

488. Sinnecker, D., A. Goedel, K.-L. Laugwitz and A. Moretti, Induced pluripotent stem cell-

derived cardiomyocytes: a versatile tool for arrhythmia research. Circulation research,

2013. 112(6): p. 961-8.

489. Ho, S.Y., Anatomy and myoarchitecture of the left ventricular wall in normal and in

disease. European Journal of Echocardiography, 2009. 10(8): p. iii3-iii7.

490. Hunter, P.J., A.J. Pullan and B.H. Smaill, Modeling total heart function. Annual review of

biomedical engineering, 2003. 5: p. 147-77.

491. Fermini, B. and A.a. Fossa, The impact of drug-induced QT interval prolongation on drug

discovery and development. Nature reviews. Drug discovery, 2003. 2(6): p. 439-47.

492. Administration, U.S.F.a.D., Recalls, Market Withdrawals & Safety Alerts. Recalls,

Market Withdrawals & Safety Alerts, 2012.

493. Piccini, J.P., D.J. Whellan, B.R. Berridge, J.K. Finkle, S.D. Pettit, N. Stockbridge, et al.,

Current challenges in the evaluation of cardiac safety during drug development:

translational medicine meets the Critical Path Initiative. American heart journal, 2009.

158(3): p. 317-26.

494. Yazawa, M., B. Hsueh, X. Jia, A.M. Pasca, J.A. Bernstein, J. Hallmayer, et al., Using

induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome.

Nature, 2011. 471(7337): p. 230-4.

495. Moretti, A., M. Bellin, A. Welling, C.B. Jung, J.T. Lam, L. Bott-Flügel, et al., Patient-

specific induced pluripotent stem-cell models for long-QT syndrome. The New England

journal of medicine, 2010. 363(15): p. 1397-409.

496. Carvajal-Vergara, X., A. Sevilla, S.L. D'Souza, Y.-S. Ang, C. Schaniel, D.-F. Lee, et al.,

Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome.

Nature, 2010. 465(7299): p. 808-12.

497. Sun, N., M. Yazawa, J. Liu, L. Han, V. Sanchez-Freire, O.J. Abilez, et al., Patient-

specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy.

Science translational medicine, 2012. 4(130): p. 130ra47-130ra47.

498. Shapira-Schweitzer, K., M. Habib, L. Gepstein and D. Seliktar, A photopolymerizable

hydrogel for 3-D culture of human embryonic stem cell-derived cardiomyocytes and rat

neonatal cardiac cells. Journal of molecular and cellular cardiology, 2009. 46(2): p. 213-

24.

499. Serena, E., E. Cimetta, S. Zatti, T. Zaglia, M. Zagallo, G. Keller, et al., Micro-arrayed

human embryonic stem cells-derived cardiomyocytes for in vitro functional assay. PloS

one, 2012. 7(11): p. e48483-e48483.

Page 180: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

156

500. Christoforou, N., B. Liau, S. Chakraborty, M. Chellapan, N. Bursac and K.W. Leong,

Induced Pluripotent Stem Cell-Derived Cardiac Progenitors Differentiate to

Cardiomyocytes and Form Biosynthetic Tissues. PLoS ONE, 2013. 8(6): p. e65963-

e65963.

501. Heidi Au, H.T., B. Cui, Z.E. Chu, T. Veres and M. Radisic, Cell culture chips for

simultaneous application of topographical and electrical cues enhance phenotype of

cardiomyocytes. Lab on a Chip, 2009. 9(4).

502. Yang, L., M.H. Soonpaa, E.D. Adler, T.K. Roepke, S.J. Kattman, M. Kennedy, et al.,

Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-

derived population. Nature, 2008. 453(7194): p. 524-8.

503. Xu, F., T. Beyazoglu, E. Hefner, U.A. Gurkan and U. Demirci, Automated and adaptable

quantification of cellular alignment from microscopic images for tissue engineering

applications. Tissue engineering. Part C, Methods, 2011. 17(6): p. 641-9.

504. Sage, D., F.R. Neumann, F. Hediger, S.M. Gasser and M. Unser, Automatic tracking of

individual fluorescence particles: application to the study of chromosome dynamics.

IEEE transactions on image processing, 2005. 14(9): p. 1372-83.

505. Chiu, L.L., R.K. Iyer, J.P. King and M. Radisic, Biphasic electrical field stimulation aids

in tissue engineering of multicell-type cardiac organoids. Tissue engineering. Part A,

2011. 17(11-12): p. 1465-1477.

506. Liu, H., Y. Sun and C.A. Simmons, Determination of local and global elastic moduli of

valve interstitial cells cultured on soft substrates. Journal of biomechanics, 2013. 46(11):

p. 1967-1971.

507. Hall, C.N. and J. Garthwaite, What is the real physiological NO concentration in vivo?

Nitric oxide : biology and chemistry / official journal of the Nitric Oxide Society, 2009.

21(2): p. 92-103.

508. Zimmermann, W.-H., I. Melnychenko, G. Wasmeier, M. Didie, H. Naito, U. Nixdorff, et

al., Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat

hearts. Nat Med, 2006. 12(4): p. 452-458.

509. Legant, W.R., A. Pathak, M.T. Yang, V.S. Deshpande, R.M. McMeeking and C.S. Chen,

Microfabricated tissue gauges to measure and manipulate forces from 3D microtissues.

Proceedings of the National Academy of Sciences of the United States of America, 2009.

106(25): p. 10097-102.

510. Kensah, G., I. Gruh, J. Viering, H. Schumann, J. Dahlmann, H. Meyer, et al., A novel

miniaturized multimodal bioreactor for continuous in situ assessment of bioartificial

cardiac tissue during stimulation and maturation. Tissue engineering. Part C, Methods,

2011. 17(4): p. 463-73.

511. Radisic, M., J. Malda, E. Epping, W. Geng, R. Langer and G. Vunjak-Novakovic,

Oxygen gradients correlate with cell density and cell viability in engineered cardiac tissue.

Biotechnology and bioengineering, 2006. 93(2): p. 332-43.

512. Onoe, H., T. Okitsu, A. Itou, M. Kato-Negishi, R. Gojo, D. Kiriya, et al., Metre-long cell-

laden microfibres exhibit tissue morphologies and functions. Nature materials, 2013.

12(6): p. 584-590.

Page 181: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

157

513. Wiedeman, M.P., Dimensions of blood vessels from distributing artery to collecting vein.

Circulation research, 1963. 12: p. 375-8.

514. Massion, P.B., O. Feron, C. Dessy and J.L. Balligand, Nitric oxide and cardiac function:

ten years after, and continuing. Circulation research, 2003. 93(5): p. 388-98.

515. Chouchani, E.T., C. Methner, S.M. Nadtochiy, A. Logan, V.R. Pell, S. Ding, et al.,

Cardioprotection by S-nitrosation of a cysteine switch on mitochondrial complex I.

Nature Medicine, 2013. 19(6): p. 753-759.

516. Miller, R.R., L.A. Vismara, R. Zelis, E.A. Amsterdam and D.T. Mason, Clinical use of

sodium nitroprusside in chronic ischemic heart disease. Effects on peripheral vascular

resistance and venous tone and on ventricular volume, pump and mechanical

performance. Circulation, 1975. 51(2): p. 328-36.

517. Mullens, W., Z. Abrahams, G.S. Francis, H.N. Skouri, R.C. Starling, J.B. Young, et al.,

Sodium nitroprusside for advanced low-output heart failure. J Am Coll Cardiol, 2008.

52(3): p. 200-7.

518. Ederli, L., L. Reale, L. Madeo, F. Ferranti, C. Gehring, M. Fornaciari, et al., NO release

by nitric oxide donors in vitro and in planta. Plant physiology and biochemistry, 2009.

47(1): p. 42-8.

519. Seddon, M., A.M. Shah and B. Casadei, Cardiomyocytes as effectors of nitric oxide

signalling. Cardiovasc Res, 2007. 75(2): p. 315-26.

520. Chiusa, M., F. Timolati, J.C. Perriard, T.M. Suter and C. Zuppinger, Sodium

nitroprusside induces cell death and cytoskeleton degradation in adult rat cardiomyocytes

in vitro: implications for anthracycline-induced cardiotoxicity. European journal of

histochemistry : EJH, 2012. 56(2): p. e15-e15.

521. Bhana, B., R.K. Iyer, W.L.K. Chen, R. Zhao, K.L. Sider, M. Likhitpanichkul, et al.,

Influence of substrate stiffness on the phenotype of heart cells. Biotechnology and

bioengineering, 2010. 105(6): p. 1148-60.

522. Radisic, M., H. Park, F. Chen, J.E. Salazar-Lazzaro, Y. Wang, R. Dennis, et al.,

Biomimetic approach to cardiac tissue engineering: oxygen carriers and channeled

scaffolds. Tissue Eng, 2006. 12(8): p. 2077-91.

523. Radisic, M., L. Yang, J. Boublik, R.J. Cohen, R. Langer, L.E. Freed, et al., Medium

perfusion enables engineering of compact and contractile cardiac tissue. American

journal of physiology. Heart and circulatory physiology, 2004. 286(2): p. H507-16.

524. Agarwal, A., J.A. Goss, A. Cho, M.L. McCain and K.K. Parker, Microfluidic heart on a

chip for higher throughput pharmacological studies. Lab on a chip, 2013. 13(ii): p. 3599-

3608.

525. Maidhof, R., N. Tandon, E.J. Lee, J. Luo, Y. Duan, K. Yeager, et al., Biomimetic

perfusion and electrical stimulation applied in concert improved the assembly of

engineered cardiac tissue. Journal of tissue engineering and regenerative medicine, 2012.

6(10): p. e12-23.

Page 182: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

158

526. Barash, Y., T. Dvir, P. Tandeitnik, E. Ruvinov, H. Guterman and S. Cohen, Electric field

stimulation integrated into perfusion bioreactor for cardiac tissue engineering. Tissue

engineering. Part C, Methods, 2010. 16(6): p. 1417-26.

527. Kaneko, T., K. Kojima and K. Yasuda, An on-chip cardiomyocyte cell network assay for

stable drug screening regarding community effect of cell network size. The Analyst, 2007.

132(9): p. 892-8.

528. Lee, K., E.A. Silva and D.J. Mooney, Growth factor delivery-based tissue engineering:

general approaches and a review of recent developments. J R Soc Interface, 2011. 8(55):

p. 153-70.

529. Simmons, C.A., E. Alsberg, S. Hsiong, W.J. Kim and D.J. Mooney, Dual growth factor

delivery and controlled scaffold degradation enhance in vivo bone formation by

transplanted bone marrow stromal cells. Bone, 2004. 35(2): p. 562-9.

530. Chen, R.R., E.A. Silva, W.W. Yuen and D.J. Mooney, Spatio-temporal VEGF and PDGF

delivery patterns blood vessel formation and maturation. Pharm Res, 2007. 24(2): p. 258-

64.

531. Silva, E.A. and D.J. Mooney, Spatiotemporal control of vascular endothelial growth

factor delivery from injectable hydrogels enhances angiogenesis. J Thromb Haemost,

2007. 5(3): p. 590-8.

532. Wang, X., E. Wenk, X. Zhang, L. Meinel, G. Vunjak-Novakovic and D.L. Kaplan,

Growth factor gradients via microsphere delivery in biopolymer scaffolds for

osteochondral tissue engineering. J Control Release, 2009. 134(2): p. 81-90.

533. Lu, S., J. Lam, J.E. Trachtenberg, E.J. Lee, H. Seyednejad, J.J. van den Beucken, et al.,

Dual growth factor delivery from bilayered, biodegradable hydrogel composites for

spatially-guided osteochondral tissue repair. Biomaterials, 2014. 35(31): p. 8829-39.

534. Borselli, C., H. Storrie, F. Benesch-Lee, D. Shvartsman, C. Cezar, J.W. Lichtman, et al.,

Functional muscle regeneration with combined delivery of angiogenesis and myogenesis

factors. Proc Natl Acad Sci U S A, 2010. 107(8): p. 3287-92.

535. Wylie, R.G., S. Ahsan, Y. Aizawa, K.L. Maxwell, C.M. Morshead and M.S. Shoichet,

Spatially controlled simultaneous patterning of multiple growth factors in three-

dimensional hydrogels. Nat Mater, 2011. 10(10): p. 799-806.

536. Wang, Y., M.J. Cooke, N. Sachewsky, C.M. Morshead and M.S. Shoichet, Bioengineered

sequential growth factor delivery stimulates brain tissue regeneration after stroke. J

Control Release, 2013. 172(1): p. 1-11.

537. Sun, Y.B., K.M.A. Yong, L.G. Villa-Diaz, X.L. Zhang, W.Q. Chen, R. Philson, et al.,

Hippo/YAP-mediated rigidity-dependent motor neuron differentiation of human

pluripotent stem cells. Nature Materials, 2014. 13(6): p. 599-604.

538. Kim, H.N., A. Jiao, N.S. Hwang, M.S. Kim, D.H. Kang, D.H. Kim, et al.,

Nanotopography-guided tissue engineering and regenerative medicine. Advanced Drug

Delivery Reviews, 2013. 65(4): p. 536-558.

Page 183: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

159

539. Lau, K., H. Tao, H. Liu, J. Wen, K. Sturgeon, N. Sorfazlian, et al., Anisotropic stress

orients remodelling of mammalian limb bud ectoderm. Nat Cell Biol, 2015. 17(5): p. 569-

79.

540. Huh, D., B.D. Matthews, A. Mammoto, M. Montoya-Zavala, H.Y. Hsin and D.E. Ingber,

Reconstituting Organ-Level Lung Functions on a Chip. Science, 2010. 328(5986): p.

1662-1668.

541. Khetani, S.R. and S.N. Bhatia, Microscale culture of human liver cells for drug

development. Nat Biotechnol, 2008. 26(1): p. 120-6.

542. Tostoes, R.M., S.B. Leite, M. Serra, J. Jensen, P. Bjorquist, M.J. Carrondo, et al., Human

liver cell spheroids in extended perfusion bioreactor culture for repeated-dose drug

testing. Hepatology, 2012. 55(4): p. 1227-36.

543. Schaaf, S., A. Shibamiya, M. Mewe, A. Eder, A. Stohr, M.N. Hirt, et al., Human

engineered heart tissue as a versatile tool in basic research and preclinical toxicology.

PLoS One, 2011. 6(10): p. e26397.

544. Jung, Y., H. Ji, Z. Chen, H. Fai Chan, L. Atchison, B. Klitzman, et al., Scaffold-free,

Human Mesenchymal Stem Cell-Based Tissue Engineered Blood Vessels. Sci Rep, 2015.

5: p. 15116.

545. Jang, K.J., H.S. Cho, H. Kang do, W.G. Bae, T.H. Kwon and K.Y. Suh, Fluid-shear-

stress-induced translocation of aquaporin-2 and reorganization of actin cytoskeleton in

renal tubular epithelial cells. Integr Biol (Camb), 2011. 3(2): p. 134-41.

546. Jang, K.J., A.P. Mehr, G.A. Hamilton, L.A. McPartlin, S. Chung, K.Y. Suh, et al.,

Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity

assessment. Integr Biol (Camb), 2013. 5(9): p. 1119-29.

547. Kim, H.J., D. Huh, G. Hamilton and D.E. Ingber, Human gut-on-a-chip inhabited by

microbial flora that experiences intestinal peristalsis-like motions and flow. Lab on a

Chip, 2012. 12(12): p. 2165-2174.

548. Mahler, G.J., M.B. Esch, R.P. Glahn and M.L. Shuler, Characterization of a

Gastrointestinal Tract Microscale Cell Culture Analog Used to Predict Drug Toxicity.

Biotechnology and Bioengineering, 2009. 104(1): p. 193-205.

549. Gautrot, J.E., C. Wang, X. Liu, S.J. Goldie, B. Trappmann, W.T. Huck, et al., Mimicking

normal tissue architecture and perturbation in cancer with engineered micro-epidermis.

Biomaterials, 2012. 33(21): p. 5221-9.

550. Sriwiriyanont, P., K.A. Lynch, E.A. Maier, J.M. Hahn, D.M. Supp and S.T. Boyce,

Morphogenesis of chimeric hair follicles in engineered skin substitutes with human

keratinocytes and murine dermal papilla cells. Exp Dermatol, 2012. 21(10): p. 783-5.

551. Wagner, I., E.M. Materne, S. Brincker, U. Sussbier, C. Fradrich, M. Busek, et al., A

dynamic multi-organ-chip for long-term cultivation and substance testing proven by 3D

human liver and skin tissue co-culture. Lab Chip, 2013. 13(18): p. 3538-47.

552. Vunjak-Novakovic, G., S. Bhatia, C. Chen and K. Hirschi, HeLiVa platform: integrated

heart-liver-vascular systems for drug testing in human health and disease. Stem cell

research & therapy, 2013. 4 Suppl 1(Suppl 1): p. S8-S8.

Page 184: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

160

553. Sung, J.H., M.B. Esch, J.M. Prot, C.J. Long, A. Smith, J.J. Hickman, et al.,

Microfabricated mammalian organ systems and their integration into models of whole

animals and humans. Lab on a Chip, 2013. 13(7): p. 1201-1212.

554. Bhise, N.S., J. Ribas, V. Manoharan, Y.S. Zhang, A. Polini, S. Massa, et al., Organ-on-a-

chip platforms for studying drug delivery systems. J Control Release, 2014. 190: p. 82-93.

555. Caplin, J.D., N.G. Granados, M.R. James, R. Montazami and N. Hashemi, Microfluidic

Organ-on-a-Chip Technology for Advancement of Drug Development and Toxicology.

Advanced Healthcare Materials, 2015. 4(10): p. 1426-1450.

556. Jain, A., M. Betancur, G.D. Patel, C.M. Valmikinathan, V.J. Mukhatyar, A. Vakharia, et

al., Guiding intracortical brain tumour cells to an extracortical cytotoxic hydrogel using

aligned polymeric nanofibres. Nature Materials, 2014. 13(3): p. 309-U118.

557. Perin, E.C., J.T. Willerson, C.J. Pepine, T.D. Henry, S.G. Ellis, D.X. Zhao, et al., Effect

of transendocardial delivery of autologous bone marrow mononuclear cells on functional

capacity, left ventricular function, and perfusion in chronic heart failure: the FOCUS-

CCTRN trial. JAMA, 2012. 307(16): p. 1717-26.

558. Zhang, B., M. Montgomery, L. Davenport-Huyer, A. Korolj and M. Radisic, Platform

technology for scalable assembly of instantaneously functional mosaic tissues. Sci Adv,

2015. 1(7): p. e1500423.

Page 185: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

161

Appendices List of publications and contributions

Journal articles:

1. Xiao Y, Radisic M. Instructive matrix design for wound healing applications. In preparation.

2. Xiao Y, Feric N, Knee EJ, Gu J, Cao S, Laschinger CA, Londono C, McGuigan AP, Radisic

M. Diabetic wound regeneration using peptide-modified hydrogels targeting the epithelium.

Submitted to Proceedings of the National Academy of Sciences.

3. Xiao Y, Reis L, Zhao Y, Radisic M. Modifications of biomaterials with immobilized growth

factors or peptides for tissue engineering applications. Methods 2015;84:44-52. DOI:

10.1016/j.ymeth.2015.04.025

4. Davenport Huyer L, Montgomery M, Zhao Y, Xiao Y, Conant G, Korolj A, Radisic M.

Biomaterial based cardiac tissue engineering and its applications. Biomedical Materials

2015;10:034004. DOI: 10.1088/1748-6041/10/3/034004

5. Miklas JW, Nunes SS, Sofla A, Reis L a, Pahnke A, Xiao Y, Laschinger C, Radisic M.

Bioreactor for modulation of cardiac microtissue phenotype by combined static stretch and

electrical stimulation. Biofabrication 2014;6:024113. DOI: 10.1088/1758-5082/6/2/024113

6. Liu H, Wen J, Xiao Y, Liu J, Hopyan S, Radisic M, Simmons C, Sun Y. In situ mechanical

characterization of the cell nucleus by atomic force microscopy. ACS Nano 2014;8:3821–8.

DOI: 10.1021/nn500553z

7. Xiao Y, Zhang B, Liu H, Miklas JW, Gagliardi M, Pahnke AQ, Thavandiran N, Sun Y,

Simmons C, Keller G, Radisic M. Microfabricated perfusable cardiac biowire: a platform that

mimics native cardiac bundle. Lab Chip 2014;14:869–82. (Front cover; Top 10%; Hot

Article) DOI: 10.1039/c3lc51123e

8. Thavandiran N, Nunes SS, Xiao Y, Radisic M. Topological and electrical control of cardiac

differentiation and assembly. Stem Cell Res Ther 2013;4:14. DOI: 10.1186/scrt162

9. Nunes SS, Miklas JW, Liu J, Aschar-Sobbi R, Xiao Y, Zhang B, Jiang J, Masse S, Gagliardi

Page 186: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

162

M, Hsieh A, Thavandiran N, Laflamme MA, Nanthakumar K, Gross GJ, Backx P, Keller G,

Radisic M. Biowire: a platform for maturation of human pluripotent stem cell–derived

cardiomyocytes. Nat Methods 2013;10:781–7. DOI: 10.1038/nmeth.2524

10. Kang K, Sun L, Xiao Y, Li SH, Wu J, Guo J, Jiang S, Yang L, Yao TM, Weisel RD, Radisic

M, Li RK. Aged human cells rejuvenated by cytokine enhancement of biomaterials for

surgical ventricular restoration. J Am Coll Cardiol 2012;60:2237–49. DOI:

10.1016/j.jacc.2012.08.985

11. Zhang B, Xiao Y, Hsieh A, Thavandiran N, Radisic M. Micro- and nanotechnology in

cardiovascular tissue engineering. Nanotechnology 2011;22:494003. DOI: 10.1088/0957-

4484/22/49/494003

Book chapters:

1. Chiu LLY, Zhang B, Xiao Y, Radisic M. Cardiac Tissue Regeneration in Bioreactors.

Biomaterials and Regenerative Medicine, Cambridge University Press; 2014, p. 640-668.

DOI: http://dx.doi.org/10.1017/CBO9780511997839.042

2. Xiao Y, Zhang B, Hsieh A, Thavandiran N, Martin C, Radisic M. Microfluidic Cell Culture

Techniques. Microfluidic Cell Culture Systems, Elsevier; 2013, p. 303–21. DOI:

10.1016/B978-1-4377-3459-1.00012-0

Oral presentations:

1. Xiao Y, Zhang B, Liu H, Sun Y, Simmons C, Radisic M. Microfabricated Perfusable

Cardiac Biowire: A Platform That Mimics Native Cardiac Bundle. Tissue Engineering and

Regenerative Medicine International Society (TERMIS) AM Annual Conference,

Washington, D.C. Dec. 13-16, 2014.

2. Xiao Y, Zhang B, Liu H, Sun Y, Simmons C, Radisic M. Developing cardiac biofibers with

microfabricated devices. MATCH/Ontario-On-A-Chip Symposium 2013, Toronto, Canada,

May 23-24, 2013

Page 187: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

163

Poster presentations:

1. Xiao Y, Radisic M. Chitosan-Collagen Hydrogel Modified with QHREDGS Peptide for

Wound Healing. 32nd Annual Meeting of the Canadian Biomaterials Society, Toronto,

Canada, May 28 - 30, 2015

2. Xiao Y, Radisic M. Chitosan-Collagen Hydrogel Modified with QHREDGS Peptide for

Wound Healing. Tissue Engineering and Regenerative Medicine International Society

(TERMIS) AM Annual Conference, Washington, D.C. Dec. 13-16, 2014.

3. Xiao Y, Zhang B, Liu H, Sun Y, Simmons C, Radisic M. Microfabricated Perfusable

Cardiac Biowire: A Platform That Mimics Native Cardiac Bundle. MATCH/Ontario-On-A-

Chip Symposium 2014, Toronto, Canada, May 29-30, 2014.

4. Xiao Y, Zhang B, Liu H, Sun Y, Simmons C, Radisic M. Microfabricated Perfusable

Cardiac Biowire: A Platform That Mimics Native Cardiac Bundle. 30th Annual Meeting of

the Canadian Biomaterials Society, Ottawa, Canada, May 29 - Jun 1, 2013.

5. Xiao Y, Zhang B, Liu H, Sun Y, Simmons C, Radisic M. Developing cardiac biofibers with

microfabricated devices. Launch of Boundless Campaign for Faculty of Applied Science &

Engineering, University of Toronto, Sept 18, 2012.

6. Xiao Y, Zhang B, Liu H, Sun Y, Simmons C, Radisic M. Developing cardiac biofibers with

microfabricated devices. 9th World Biomaterials Congress, Chengdu, China, Jun 1-5, 2012.

7. Xiao Y, Zhang B, Liu H, Sun Y, Simmons C, Radisic M. Developing cardiac biofibers with

microfabricated devices. MATCH/Ontario-On-A-Chip Symposium 2012, Toronto, Canada,

May 17-18, 2012.

8. Xiao Y, Radisic M. Engineering Cardiac Purkinje Fibers with Microfabricated Devices.

2011 Annual University of Toronto Institute of Biomaterials & Biomedical Engineering

Scientific Day, Toronto, Canada, May 19, 2011.

Page 188: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

164

Other conference proceedings:

1. Nunes SS, Miklas JW, Xiao Y, Zhang B, Radisic M. Stem cell-derived cardiomyocyte

maturation by biomimetic topographical and electrical cues. North American Vascular

Biology Organization (NAVBO), Hyannis, Massachusetts, Oct. 20-24, 2013.

2. Sun L, Kang K, Xiao Y, Li SH, Wu J, Guo J, Jiang S, Yang L, Yao TM, Weisel RD, Radisic

M, Li RK. Aged human cells rejuvenated by cytokine enhancement of biomaterials for

surgical ventricular restoration. American Heart Association 2012 Scientific Sessions, Los

Angeles, California, Nov. 3-7, 2012.

3. Xiao Y, Thavandiran N, Au H, Radisic M. Microbioreactors for cardiac tissue engineering.

Tissue Engineering and Regenerative Medicine International Society (TERMIS) EU Annual

Conference, Granada, Spain, Jun. 7-10, 2011.

Page 189: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

165

Copyright Acknowledgements

Copyright © 2012 Elsevier. Contents of this chapter have been published in J Am Coll

Cardiol: Kang K, Sun L, Xiao Y, Li SH, Wu J, Yao TM, Weisel RD, Radisic M, Li RK. Aged

human cells rejuvenated by cytokine enhancement of biomaterials for surgical ventricular

restoration. J Am Coll Cardiol. 2012;60:2237–2249. Reuse with permission from Elsevier. A

link to the published paper can be found at:

http://www.sciencedirect.com/science/article/pii/S0735109712043677

Copyright © 2013 BioMed Central. Contents of this thesis have been published in Stem Cell

Res Ther: Thavandiran N, Nunes SS, Xiao Y, Radisic M. Topological and electrical control of

cardiac differentiation and assembly. Stem Cell Res Ther. 2013;4:14. Reuse with permission

from BioMed Central. A link to the published paper can be found at:

http://www.stemcellres.com/content/4/1/14

Copyright © 2014 Cambridge University Press. Contents of this thesis have been published in:

Chiu LLY, Zhang B, Xiao Y, Radisic M. Cardiac tissue regeneration in bioreactors. Biomaterials

and Regenerative Medicine, Cambridge University Press. 2014;640-668. Reuse with permission

from Cambridge University Press. A link to the published chapter can be found at:

http://ebooks.cambridge.org/chapter.jsf?bid=CBO9780511997839&cid=CBO9780511997839A0

46

Copyright © 2014 Royal Society of Chemistry. Contents of this chapter have been published

in Lab Chip: Xiao Y, Zhang B, Liu H, Miklas JW, Gagliardi M, Pahnke A, Thavandiran N, Sun

Y, Simmons C, Keller G, Radisic M. Microfabricated perfusable cardiac biowire: a platform that

mimics native cardiac bundle. Lab Chip. 2014; 14:869–82. Reuse with permission from Royal

Society of Chemistry. A link to the published paper can be found at:

http://pubs.rsc.org/en/content/articlelanding/2014/lc/c3lc51123e

Copyright © 2015 Elsevier. Contents of this thesis have been published in Methods: Xiao Y,

Reis LA, Zhao Y, Radisic M. Modifications of collagen-based biomaterials with immobilized

growth factors or peptides. Methods. 2015;84:44–52. Reuse with permission from Elsevier. A

Page 190: Instructive Matrix Cues for Tissue Engineering Applications · Chapter 2 introduces the key topics on designing instructive biomaterials for tissue engineering applications, with

166

link to the published paper can be found at:

http://www.sciencedirect.com/science/article/pii/S1046202315001723

Copyright © 2015 IOP Publishing. Contents of this chapter have been published in

Biomedical Materials: Davenport Huyer L, Montgomery M, Zhao Y, Xiao Y, Conant G, Korolj

A, Radisic M. Biomedical Materials. 2015;10:034004. Reuse with permission from IOP

Publishing. A link to the published chapter can be found at:

http://iopscience.iop.org/article/10.1088/1748-6041/10/3/034004