171
A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction by Lewis A. Reis A thesis submitted in conformity with the requirements for the degree of Doctor o Philosophy The Institute for Biomaterials and Biomedical Engineering University of Toronto © Copyright by Lewis Reis 2015

A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

  • Upload
    vandang

  • View
    214

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction

by

Lewis A. Reis

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

The Institute for Biomaterials and Biomedical Engineering

University of Toronto

© Copyright by Lewis Reis 2015

Page 2: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

ii

A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction

Lewis A. Reis

Doctor of Philosophy

The Institute of Biomaterials and Biomedical Engineering

University of Toronto

2015

Abstract

Myocardial infarction (MI) results in the death of cardiomyocytes (CM) followed by scar formation

and pathological remodeling of the heart. We postulate that immobilization of the pro-survival

angiopoietin-1-derived peptide, QHREDGS, to a chitosan-collagen hydrogel could produce a clinically

translatable thermo-responsive hydrogel to attenuate post-MI cardiac remodeling. Conjugation of

QHREDGS peptide to chitosan does not interfere with gelation, structure, or mechanical properties of

chitosan-collagen hydrogel blends. The storage modulus of 2.5mg/mL 1:1 mass:mass chitosan:collagen

was measured to be 54.9±9.1 Pa and loss modulus of 6.1±0.9 Pa. Dose response of the QHREDGS

peptide was assessed and it was found that CMs encapsulated in High peptide gel (651±8 nmol

peptide/mL-gel) showed improved morphology, viability, and metabolic activity in comparison to the

Low peptide (100±30 nmol peptide/mL-gel) and Control (No Peptide) groups. Construct (CMs in

hydrogel) functional properties were not significantly different between the groups, however success rate

of obtaining a beating construct was improved in the hydrogel with the High amount of QHREDGS

Page 3: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

iii

peptide immobilized compared to the Low and Control groups. An adult Lewis rat left anterior

descending coronary artery ligation procedure to mimic acute MI model was used to assess in vivo

hydrogel performance. QHREDGS-conjugated hydrogel (QHG213H), Control gel, or PBS was injected

into 3 locations in the MI zone. By in vivo tracking and chitosan staining, the hydrogel was demonstrated

to remain in situ for 2 weeks and cleared in about 3 weeks. By echocardiography and pressure-volume

analysis, the QHG213H hydrogel significantly improved cardiac function compared to the controls. Scar

thickness and scar area fraction were also significantly improved with QHG213H gel injection compared

to the controls. Mechanistically, there were significantly more cardiomyocytes, determined by cardiac

troponin-T staining, in the MI zone of the QHG213H hydrogel group. No significant difference in

inflammatory response between groups was observed as determined by gene regulation and cytokine

analysis of excised heart sections 24 hours after treatment. The interaction of CMs with QHREDGS was

found to be mediated by β1-integrins and to increase expression of the pro-survival effector MAPK.

Page 4: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

iv

Acknowledgements

First, I must thank my supervisor and mentor Dr. Milica Radisic for initially giving me the

opportunity to work in her lab, and for the years of support and guidance. I hope the successful

completion of this thesis and the project which we have worked on over the last number of years

is testament to that.

My committee members, Drs. Julie Audet and Molly Shoichet have scrutinized this work

many times and given invaluable advice on making it the best it can be, for which I am grateful.

The help, advice, and collaborative work from Dr. Ren-Ke Li and his research group,

specifically Dr. Jun Wu, have made a large part of this work possible and I must extend my

gratitude to them. Dr. Abdul Momen must also be acknowledged for his help in performing

much of the early in vivo studies.

All the past and present member of the Laboratory for Functional Tissue engineering have

given immeasurable support and help over the years and all have contributed to making this

project what it is. I must acknowledge Loraine Chiu who’s collaborative efforts undoubtedly

shortened my time here by many years, as well as the efforts of Nicole Feric, Carol Laschinger,

Jason Miklas, Kent Hyun, and Yan Liang for their contributions to this project and ensuring the

work was published.

Friends at U of T, Massey College, and those smart enough to not have gone into graduate

school, you all know who you are and I thank you for being there throughout.

To my family, none of this would have been possible without your love and support. Mom, if

it weren’t for you I never would have attempted this challenge nor seen it through to completion.

Emily, you came into my life at the beginning of this, changed my life forever, and have stood

by me throughout. Thank you and I love you.

Finally, I owe a debt of gratitude to all the animals who gave their lives in the name of science

and contributed to my work. Know that your sacrifice was not in vain.

Page 5: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

v

Declaration of Co-Authorship

The original scientific content of this thesis is comprised of two published, peer reviewed

articles in internationally recognized journals. These articles were primarily the work of Lewis

Reis. Introduction and literature review are from three published review papers, the transcribed

writing being that of Lewis Reis. The contributions of co-authors are stated in the thesis, in

conformity with the requirements for the degree of Doctor of Philosophy.

Page 6: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

vi

Permissions

Copyright © 2015 Wolters Kluwer Health. Contents of this thesis have been published in

Circulation: Heart Failure: Reis L.A., Chiu L.L.Y., Wu J., Feric N., Momen A., Li R.K., &

Radisic M. (2015). Hydrogels with integrin-binding angiopoietin-1-derived peptide, QHREDGS,

for treatment of acute myocardial infarction. Circulation: Heart Failure. DOI:

10.1161/CIRCHEARTFAILURE.114.001881. Reuse with permission from Wolters Kluwer

Health. A link to the published paper can be found at:

http://circheartfailure.ahajournals.org/content/early/2015/01/28/CIRCHEARTFAILURE.114.001

881.abstract

Copyright © 2014 John Wiley & Sons, Ltd. Contents of this thesis have been published in the

Journal of Tissue Engineering and Regenerative Medicine: Reis L.A., Chiu L.L.Y., Feric N., Fu

L., & Radisic M. (2014). Biomaterials in myocardial tissue engineering. Journal of Tissue

Engineering & Regenerative Medicine. DOI:10.1002/term.1944. Reuse with permission from

John Wiley & Sons, Ltd. A link to the published paper can be found at:

http://onlinelibrary.wiley.com/doi/10.1002/term.1944/abstract

Copyright © 2012 Frontiers in Bioscience. Contents of this thesis have been published in

Frontiers in Bioscience: Chiu LLY, Iyer RK, Reis LA, et al. (2012). Cardiac tissue engineering:

current state and perspectives. Frontiers in Bioscience. 17:1533-1550. Reuse with permission

from Frontiers in Bioscience. DOI: 10.2741/4002. A link to the published paper can be found at:

www.bioscience.org/2012/v17/af/4002/list.htm

Copyright © 2012 Elsevier. Contents of this thesis have been published in Acta Biomaterialia.

Reis LA et al. (2012). A peptide-modified chitosan-collagen hydrogel for cardiac cell culture

and delivery. Acta Biomaterilia. 8(3): 1022-36. Reuse with permission from Elsevier. DOI:

10.1016/j.actbio.2011.11.030. A link to the published paper can be found at:

www.sciencedirect.com/science/article/pii/S1742706111005332

Copyright © 2011 Elsevier. Contents of this thesis have been published in Current Opinion in

Biotechnology: Iyer RK, Chiu LLY, Reis LA, & Radisic M. (2011). Engineered cardiac tissues.

Page 7: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

vii

Current Opinion in Biotechnology. 22(5):706-714. Reuse with permission from Elsevier. DOI:

10.1016/j.copbio.2011.04.004. A link to the published paper can be found at:

www.sciencedirect.com/science/article/pii/S0958166911000668

Copyright © 2014 Elsevier. Contents of this thesis have been published in Cardiac

Regeneration & Repair (Vol. II, Ch. 3). Reis LA et al. (January 2014). Injectable biomaterials

for cardiac repair. In Li R.K. & Weisel R.D. (EDs), Cardiac Regeneration & Repair (Vol. II, Ch.

3). Cambridge: Woodhead Publishing. Reuse with permission from Elsevier. DOI:

10.1533/9780857096715.1.49. A link to the book chapter can be found at:

http://www.sciencedirect.com/science/article/pii/B9780857096593500037

Copyright © 2011 Springer. Contents of this thesis have been published in Biomaterials for

Tissue Engineering Applications: A review of past & future trends: Odedra D, Chiu LLY, Reis

LA, et al. (2011). Cardiac Tissue Engineering. In J. A. Burdick & R. L. Mauck (EDs.),

Biomaterials for Tissue Engineering Applications: A review of past & future trends (1st ed., p.

562). Vienna: Springer Vienna. Reuse with permission from Springer. DOI: 10.1007/978-3-

7091-0385-2_15. A link to the book chapter can be found at:

http://link.springer.com/chapter/10.1007/978-3-7091-0385-2_15/fulltext.html

Page 8: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

viii

Abstracts of Published Works Appearing in the Thesis

A peptide-modified chitosan-collagen hydrogel for cardiac cell culture and delivery

Reis, Lewis A; Chiu, Loraine L Y; Liang, Yan; Hyunh, Kent; Momen, Abdul; & Radisic, Milica.

Acta Biomaterialia. (2012). 8(6): 1022-1036.

Myocardial infarction (MI) results in the death of cardiomyocytes (CM) followed by scar

formation and pathological remodeling of the heart. We propose that chitosan conjugated with

the angiopoietin-1 derived peptide, QHREDGS, and mixed with collagen I forms a

thermoresponsive hydrogel better suited for the survival and maturation of transplanted

cardiomyocytes in vitro compared to collagen and chitosan-collagen hydrogels alone.

Conjugation of QHREDGS peptide to chitosan does not interfere with the gelation, structure or

mechanical properties of the hydrogel blends. The storage modulus of 2.5 mg ml(-1) 1:1

mass:mass (m:m) chitosan-collagen was measured to be 54.9 ± 9.1 Pa, and the loss modulus

6.1±0.9 Pa. The dose-response of the QHREDGS peptide was assessed and it was found that

CMs encapsulated in High-peptide gel (651 ± 8 nmol peptide ml-gel(-1)) showed improved

morphology, viability and metabolic activity in comparison to the Low-peptide (100 ± 30 nmol

peptide ml-gel(-1)) and Control (No Peptide) groups. Construct (CMs in hydrogel) functional

properties were not significantly different between the groups; however, the success rate of

obtaining a beating construct was improved in the hydrogel with the High amount of QHREDGS

peptide immobilized compared to the Low and Control groups. Subcutaneous injection of

hydrogel (Control, Low and High) with CMs in the back of Lewis rats illustrated its ability to

localize at the site of injection and retain cells, with CM contractile apparati identified after

seven days. The hydrogel was also able to successfully localize at the site of injection in a mouse

MI model.

Contributions: LAR-concept and design, performed all experiments and data analysis,

manuscript writing; LLYC- concept and design, assistance in surgical work, preparation of samples

for histology, data analysis; YL- image analysis for vasculature; KH- Pico green DNA analysis;

AM-performed surgical work; MR-concept, data interpretation, final approval of manuscript.

Page 9: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

ix

Hydrogels with integrin-binding angiopoietin-1-derived peptide, QHREDGS, for treatment

of acute myocardial infarction

Reis, Lewis A; Chiu, Loraine L Y; Wu, Jun; Feric, Nicole; Laschinger, Carol; Momen, Abdul;

Li, Ren-Ke; & Radisic, Milica. Circulation Heart Failure. (2015). DOI:

10.1161/CIRCHEARTFAILURE.114.001881.

Background: Hydrogels are being actively investigated for direct delivery of cells or

bioactive molecules to the heart post-myocardial infarction (MI) to prevent cardiac functional

loss. We postulate that immobilization of the pro-survival angiopoietin-1-derived peptide,

QHREDGS, to a chitosan-collagen hydrogel could produce a clinically translatable thermo-

responsive hydrogel to attenuate post-MI cardiac remodeling.

Methods & Results: In a rat MI model, QHREDGS-conjugated hydrogel (QHG213H),

Control gel, or PBS was injected into the peri-infarct/MI zone. By in vivo tracking and chitosan

staining, the hydrogel was demonstrated to remain in situ for 2 weeks and was cleared in ~3

weeks. By echocardiography and pressure-volume analysis, the QHG213H hydrogel

significantly improved cardiac function compared to the controls. Scar thickness and scar area

fraction were also significantly improved with QHG213H gel injection compared to the controls.

There were significantly more cardiomyocytes (CMs), determined by cardiac troponin-T

staining, in the MI zone of the QHG213H hydrogel group; and hydrogel injection did not induce

a significant inflammatory response as assessed by PCR and an inflammatory cytokine assay.

The interaction of CMs and cardiac fibroblasts with QHREDGS was found to be mediated by β1-

integrins.

Conclusions: We demonstrated for the first time that the QHG213H hydrogel can be injected

in the beating heart where it remains localized for a clinically effective period. Moreover, the

QHG213H hydrogel induced significant cardiac functional and morphological improvements

post-MI relative to the controls.

Contributions: LAR-concept and design, performed all experiments and data analysis,

manuscript writing; LLYC- concept and design, assistance in surgical work, preparation of samples

for histology, image & data analysis; JW- design, performed surgical work, preparation of samples

for histology, data analysis and interpretation, manuscript writing; NF- Attachment assay,

Page 10: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

x

manuscript writing; AM-performed surgical work; RKL & MR-concept, data interpretation, final

approval of manuscript.

Page 11: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

xi

Table of Contents

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

Acknowledgements ................................................................................................................................. iv

Declaration of Co-Authorship ................................................................................................................. v

Permissions ............................................................................................................................................. vi

Abstracts of Published Works Appearing in the Thesis ....................................................................... viii

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

1.1. Motivation ................................................................................................................................. 1

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

1.3. Specific aims ............................................................................................................................. 2

2. Literature Review .......................................................................................................................... 3

2.1. Cardiovascular Disease, Outcomes, and Treatment Options .................................................... 3

2.2. Cell sources, stem cells, & differentiation ................................................................................ 5

2.3. Design criteria for biomaterials in cardiac tissue engineering .................................................. 7

2.3.1. Biocompatibility ................................................................................................................ 7

2.3.2. Biodegradability ................................................................................................................ 7

2.3.3. Mechanical Support .......................................................................................................... 9

2.3.4. Injectability ..................................................................................................................... 11

2.3.5. Clinically Relevant Thickness......................................................................................... 11

2.3.6. Application Time ............................................................................................................ 11

2.4. Tissue engineered cardiac grafts (in vitro engineering) .......................................................... 12

2.5. Injection (in vivo engineering) ................................................................................................ 14

2.5.1. Cell only based therapies ................................................................................................ 15

2.5.2. Hydrogels to promote endogenous repair ....................................................................... 16

2.5.3. Hydrogels for the delivery of cells for regeneration ....................................................... 21

2.5.4. Hydrogels for the artificial maintenance of ventricle geometry and repair .................... 26

2.5.5. Clinical application ......................................................................................................... 29

2.6. Chitosan & collagen ................................................................................................................ 30

2.7. QHREDGS peptide ................................................................................................................. 33

2.8. Peptide modified chitosan-collagen hydrogel for cardiac tissue engineering ......................... 34

3. Hydrogel development, in vitro characterization & preliminary in vivo models ........................ 36

Page 12: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

xii

3.1. Materials & Methods .............................................................................................................. 36

3.1.1. Peptide modified chitosan (UP-G113-QHREDGS) ........................................................ 36

3.1.1.1. Conjugation .............................................................................................................. 36

3.1.1.2. Assessing conjugation efficiency ............................................................................. 36

3.1.2. Chitosan-collagen hydrogel ............................................................................................ 37

3.1.2.1. Hydrogel formulation ............................................................................................... 37

3.1.2.2. Scanning electron microscopy .................................................................................. 37

3.1.2.3. Hydrogel degradation ............................................................................................... 38

3.1.2.4. Rheological assessment of hydrogels ....................................................................... 38

3.1.3. In vitro cell culture .......................................................................................................... 38

3.1.3.1. CM isolation ............................................................................................................. 38

3.1.3.2. CM media ................................................................................................................. 39

3.1.3.3. CM encapsulation & culture ..................................................................................... 39

3.1.4. In vivo studies.................................................................................................................. 39

3.1.4.1. Subcutaneous injection ............................................................................................. 39

3.1.4.2. Mouse MI model ...................................................................................................... 40

3.1.5. Construct Characterization .............................................................................................. 40

3.1.5.1. Gel compaction ......................................................................................................... 40

3.1.5.2. Live/Dead staining .................................................................................................... 41

3.1.5.3. Functional testing ..................................................................................................... 41

3.1.5.4. XTT assay ................................................................................................................. 42

3.1.5.5. LDH assay ................................................................................................................ 42

3.1.5.6. PicoGreen DNA Assay ............................................................................................. 43

3.1.5.7. Histological & immunofluorescent staining ............................................................. 43

3.1.6. Statistical analysis ........................................................................................................... 45

3.2. Results & Discussion .............................................................................................................. 45

3.2.1. Hydrogel composition and characterization .................................................................... 45

3.2.1.1. Base collagen-chitosan hydrogel .............................................................................. 45

3.2.1.2. Collagen-chitosan-QHREDGS hydrogel .................................................................. 50

3.2.2. In vitro studies with cardiomyocytes in collagen-chitosan-QHREDGS hydrogels ........ 52

Page 13: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

xiii

3.2.2.1. Distribution, viability, and metabolism of CMs encapsulated in the hydrogels ....... 52

3.2.2.2. Construct functional & morphological properties .................................................... 57

3.2.3. In vivo studies.................................................................................................................. 59

3.2.3.1. Subcutaneous injection ............................................................................................. 59

3.2.3.2. Mouse MI model study ............................................................................................. 64

3.3. Summary ................................................................................................................................. 69

4. Utility of QH-G213-H hydrogel as a treatment for acute MI ..................................................... 70

4.1. Materials & Methods .............................................................................................................. 70

4.1.1. Experimental design overview ........................................................................................ 70

4.1.2. Peptide modified chitosan-collagen hydrogel (QHG213H) ............................................ 71

4.1.3. Peptide modified polyethylene glycol (PEG-QHREDGS) ............................................. 71

4.1.4. In vivo studies.................................................................................................................. 72

4.1.4.1. Assessment of cardiac function (6 Week time point) ............................................... 72

4.1.4.2. 24 hr time point ........................................................................................................ 73

4.1.4.3. In vivo hydrogel lifespan .......................................................................................... 74

4.1.5. Immunohistochemistry .................................................................................................... 74

4.1.6. Quantitative PCR ............................................................................................................ 75

4.1.7. Western blotting .............................................................................................................. 75

4.1.8. Image analysis techniques ............................................................................................... 76

4.1.9. Statistical analysis ........................................................................................................... 76

4.2. Results ..................................................................................................................................... 76

4.2.1. In vivo degradation (lifespan).......................................................................................... 76

4.2.2. Functional Data ............................................................................................................... 77

4.2.3. Gross morphology and histology .................................................................................... 81

4.2.4. Cardiomyocyte survival mechanism ............................................................................... 84

4.3. Discussion ............................................................................................................................... 86

4.4. Summary ................................................................................................................................. 94

5. Recommendations for future work ............................................................................................. 96

5.1. Specific Aim SA-2. i. Show ability to localize in vivo and quantify duration of its in vivo

presence. 96

5.2. Specific Aim SA-2. iii. Determine mechanism of action. ....................................................... 96

Page 14: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

xiv

6. Conclusions & Contributions to New Knowledge ...................................................................... 99

6.1. Publications & Contributions ................................................................................................ 101

7. References ................................................................................................................................. 105

Appendix A. Reaction schematics and chemical structures ........................................................... 133

Appendix B. In vitro construct CM distribution............................................................................. 136

Appendix C. Utility of QHG213H hydrogel for MI treatment published supplementary data ...... 139

Appendix D. Unpublished rat MI model supplementary material .................................................. 146

List of Tables

Table 3-1: Hydrogel rheological assessment ........................................................................... 48

Table 4-1: Comparative-analysis of reported MI studies ......................................................... 94

Table C-1: Genes and primers used in RT-qPCR .................................................................. 139

Table C-2: Description of studies considered for comparative-analysis .............................. 140

List of Figures

Figure 2-1: Histological stages of myocardial infarction ........................................................... 4

Figure 2-2: The structures of chitosan and chitin, and their proportions in 85% deacetylated

chitosan ......................................................................................................................................... 31

Figure 2-3: Molecular structure of Ang-1 derived peptide QHREDGS .................................. 34

Figure 2-4: QHREDGS peptide modified chitosan (QHG213H) ............................................ 35

Figure 3-1: Hydrogel Characterization .................................................................................... 49

Figure 3-2: Characterization of peptide conjugation and hydrogel morphology ..................... 52

Figure 3-3: Gel encapsulation results in uniform distribution of live cells.............................. 53

Figure 3-4: Metabolic activity and total number of cells encapsulated in 1:1 chitosan:collagen

hydrogels ....................................................................................................................................... 56

Figure 3-5: 1:1 Chitosan:collagen hydrogel with immobilized QHREDGS enables cultivation

of beating cardiac tissue ................................................................................................................ 58

Figure 3-6: Subcutaneous injection study ................................................................................ 61

Figure 3-7: Immunostaining for different cell populations in subcutaneously injected nodules

after 7 days in vivo ........................................................................................................................ 63

Page 15: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

xv

Figure 3-8: Chitosan:collagen gel is suitable for injection into the infarcted heart ................. 65

Figure 3-9: Vascularization and wall thickness in the mouse MI model ................................. 67

Figure 4-1: In vivo experimental design................................................................................... 71

Figure 4-2: In vivo lifespan of Dy-light 800 labelled chitosan-collagen gel............................ 77

Figure 4-3: Cardiac function post MI measured by echocardiography .................................... 79

Figure 4-4: Load dependent cardiac function and LV volumes at 6 weeks post MI ............... 80

Figure 4-5: Load independent pressure-volume analysis at 6 weeks post MI ......................... 81

Figure 4-6: Gross heart morphology at 6 weeks post MI......................................................... 82

Figure 4-7: Vascularization at 6 weeks post MI ...................................................................... 83

Figure 4-8: Apoptosis at 6 weeks post MI ............................................................................... 85

Figure 4-9: Cardiac cell attachment to PEG and PEG-QHREDGS gels ................................. 86

Figure 4-10: Proposed QHREDGS mediated mechanism ....................................................... 92

Figure A-1: Polyelectrolytic complexation of chitosan and collagen .................................... 133

Figure A-2: Modified chitosans used ..................................................................................... 134

Figure A-3: EDC/Sulfo-NHS reaction chemistry for conjugation of biomolecules .............. 134

Figure A-4: QHREDGS conjugation to PEG......................................................................... 135

Figure B-1: In vitro cell leakage and migration ..................................................................... 136

Figure B-2: Quantification of in vitro construct viability ...................................................... 137

Figure B-3: High QHREDGS hydrogel constructs contain elongated cardiomyocytes with

visible cross-striations ................................................................................................................. 138

Figure C-1: Quantification of hydrogel lifespan .................................................................... 141

Figure C-2: Histological staining 6 weeks post MI ............................................................... 142

Figure C-3: RT-qPCR of MI and border zone tissues from hearts excised 24 hrs post MI ... 143

Figure C-4: Western blot analysis of border zone tissue from hearts excised 24 hrs post MI144

Figure C-5: Reported cardiac morphological and functional data from studies used in meta-

analysis ........................................................................................................................................ 145

Figure D-1: Heart morphology & histology 3Wk post MI .................................................... 146

Figure D-2: Further histological staining 3Wks post MI ....................................................... 147

Figure D-3: 3Wk MI quantification measurements ............................................................... 149

Figure D-4: 3Wk MI scar extent assessment ......................................................................... 150

Figure D-5: 6Wk MI quantification measurements (from histology pictures) ...................... 151

Page 16: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

xvi

Figure D-6: Vascularization within the MI scar zone at 3Wks post infarct ........................... 152

Figure D-7: Apoptosis in the MI boundary zone at 3Wk ...................................................... 153

Figure D-8: 3Wk TUNEL/cTnT staining controls ................................................................. 154

Figure D-9: 6Wk TUNEL/cTnT staining raw counts ............................................................ 154

Figure D-10: 6Wk TUNEL/cTnT staining controls ............................................................... 155

Page 17: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

1

1. Introduction

Parts of the text used in this chapter has been published in Circulation Heart Failure (Reis et

al. 2015), and is used with permission from Wolters Kluwer Health.

1.1. Motivation

Cardiomyocyte (CM) death, myocardial remodeling, and scar tissue formation following

myocardial infarction (MI) results in severe cardiac dysfunction and serious health problems. If

not treated, and often with treatment, heart failure progresses. Direct transplantation of various

cell types and/or bioactive molecules has shown promise, however both strategies have been

hampered by low injection site retention and even lower long-term survival of the cells/bioactive

molecules (Mayfield et al. 2014; Nelson et al. 2012). To mitigate these problems these

treatments are combined with hydrogels to confine the cells/bioactive molecules to the site of

injection and promote their long-term survival therein.

The acute phase post-MI might be the most appropriate time to utilize a hydrogel-based

therapy because therein hydrogel injections can prevent cardiac remodeling, deliver cells to

replace the damaged tissue, and/or recruit endogenous stem cells (Menasché 2008). Chitosan and

collagen are natural, biodegradable, biocompatible polymers that have been explored for their

potential use in the treatment of cardiac dysfunction (Lu et al. 2009; Rask, Dallabrida, et al.

2010; Garbern et al. 2011; Suuronen et al. 2006; Y. Zhang et al. 2008; Wu et al. 2011).

Typically, collagen and chitosan, alone or in combination, are cross-linked using exogenous,

often toxic, chemical cross-linkers to improve the hydrogel mechanical properties (Lu et al.

2009; Deng et al. 2010; Fujita et al. 2005). We propose that chitosan-collagen composites can gel

naturally at physiological temperatures and pH to form mechanically stable hydrogels that are

appropriate for in vivo application without the need for exogenous cross-linkers. Furthermore,

the collagen-chitosan interaction within the gels resembles the collagen-glycosaminoglycan

interaction found in vivo in the extracellular matrix (ECM) (Tan, Krishnaraj, and Desai 2001).

Thus, chitosan-collagen may mediate physiological cell-matrix interactions.

The functional success of hydrogel-based cardiac and cell therapies can be improved by

modifying biomaterials with bioactive molecules because they (cytokines, growth factors, etc.)

have the potential to increase transplanted cell survival, reduce resident cell apoptosis, recruit

desired regenerative cells, and promote stem/progenitor cell differentiation (Chiu, Iyer, et al.

Page 18: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

2

2012; Reis et al. 2014a). One such bioactive molecule is the growth factor angiopoietin-1

(Ang1). In endothelial cells, Ang1 binds the Tie2 receptor (Hayes et al. 1999; I. Kim et al. 2000;

Papapetropoulos 2000) but in cells such as neonatal rat cardiomyocytes (NCMs) that lack the

Tie2 receptor, Ang1 binds to integrins (Carlson et al. 2001) and activates pro-survival pathways

(Dallabrida et al. 2005). We identified the short sequence QHREDGS as the integrin-binding

motif of Ang1, and the QHREDGS peptide was found to support CM attachment and survival

similar to full-length Ang1 (Rask, Dallabrida, et al. 2010; Rask, Mihic, et al. 2010). It is

therefore possible that the QHREDGS peptide could retain/restore cardiac contractile function

post-MI by promoting CM survival. Importantly, the QHREDGS peptide is water-soluble, stable,

fully-synthetic with a precisely defined composition, and does not require a specific orientation

to be functional. We therefore look to incorporate the pro-survival peptide QHREDGS into an

optimized chitosan-collagen hydrogel to produce a novel hydrogel-based cardiac regenerative

therapy. We chose to immobilize the peptide onto the hydrogel because the effects of

QHREDGS were found to be attachment-dependent, and immobilization would ensure sustained

localization at the injection site and requires a single low dose.

1.2. Hypothesis

Conjugation of Angiopoietin-1 derived peptide QHREDGS to chitosan, injected as a peptide-

modified chitosan-collagen gel, will enhance survival of cardiac muscle after myocardial

infarction in vivo and attenuate pathological remodeling.

1.3. Specific aims

SA-1. Develop a chitosan-collagen hydrogel that supports CM viability and phenotype.

Conjugate QHREDGS and demonstrate dose dependent improvement in CM phenotype and

function in vitro.

SA-2. Assess effect of the developed hydrogel in a rat MI model.

i. Show ability to localize in vivo and quantify duration of its in vivo presence.

ii. Demonstrate potential to attenuate cardiac remodeling post MI.

iii. Determine mechanism of action.

Page 19: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

3

2. Literature Review

Contents of this chapter have been published in (Iyer et al. 2011; Odedra et al. 2011; Chiu,

Iyer, et al. 2012; Reis et al. 2012; Reis et al. 2014b; Reis et al. 2015) and are used with

permission from respective publishers.

2.1. Cardiovascular Disease, Outcomes, and Treatment Options

Cardiovascular disease (CVD) is currently the leading cause of death in the world and it is

projected that it will remain as such throughout the next decade (World Health Organization

2011a; World Health Organization 2011b; Mathers and Loncar 2006). In 2008 alone, CVD

accounted for 1 in 3 deaths in the US (811,940 of 2,471,984) and of those, half (approximately 1

in 6 American deaths) were attributable to coronary heart disease (Roger et al. 2012).

Furthermore it is estimated that each year 785,000 Americans have a new coronary attack, about

470,000 have a recurrent attack and a further 195,000 suffer their first silent myocardial

infarction (Roger et al. 2012). While the disorders categorized as CVDs have such divergent

causes as atherosclerosis, rheumatic fever, congenital malformations and thrombosis, they all

converge to cause damage to the heart muscle. Unfortunately, the damage is irreversible because

the heart muscles cells, cardiomyocytes, are thought to be terminally differentiated and non-

proliferative (Sutton and Sharpe, 2000; Laflamme and Murry, 2005), which necessarily limits the

regenerative potential of the heart.

Among CVDs, ischemic heart disease is the most prevalent (World Health Organization

2011b; Roger et al. 2012). It has therefore been the subject of intensive research. The partial or

complete blockage of a coronary artery reduces/prevents blood supply to the downstream heart

muscle and the affected tissue becomes severely nutrient and oxygen deprived, inducing

cardiomyocyte death: an event known as a myocardial infarction (MI). Myocardium undergoes

irreversible damage within 20 minutes of MI and a subsequent wave-front of cell death sweeps

over the area of ischemia over a three to six hour period finally resulting in the death of up to a

billion cells (M. A. Laflamme and Murry 2005). In the weeks that follow, cardiac function is

greatly reduced due to the invasion of leukocytes into the infarct area, removal of dead tissue,

and deposition of granulation tissue. Occurring in as little as two months the ventricular

remodeling results in the formation of tough, rigid, collagenous fibrotic scar tissue at the site of

(and surrounding the) infarct, and thinning of the ventricular wall; both leading to increased

Page 20: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

4

ventricular wall stress and volume, and decreased ejection fraction and contraction force; see

Figure 2-1(Sutton and Sharpe 2000; M. A. Laflamme and Murry 2005). In the end, without

intervention and in most cases even with intervention, a myriad of health complications manifest,

ultimately leading to irreversible heart failure and premature death.

Figure 2-1: Histological stages of myocardial infarction (Reproduced with permission from Nature Publishing Group, Laflamme and Murry 2005)

Even with treatment of the underlying cause of heart disease, approximately 50% of patients

will experience heart failure within five years of an acute MI (Pantilat and Steimle 2004; Go et

al. 2013). At present, the gold standard treatment for those who have reached end-stage heart

failure is a heart transplant; but the insufficiency of donors combined with the need for patient-

donor matched organs severely limits the number of patients that can be treated (Zammaretti and

Jaconi 2004; Menasché 2008). In 2010, there were 2,333 heart transplants performed in the US

(Roger et al. 2012). In 2011, greater than 3,000 people were on the waiting list for a heart

transplant and thus living with end-stage heart failure (Roger et al. 2012).

Over the last few years left ventricular assist devices (LVADs) have shown great

improvement in safety and efficacy. As such, LVADs have been increasingly used as a bridge to

transplantation, giving those awaiting a transplant a credible life-saving therapy option by

supporting the failing heart, with the consequence of reducing transplant waitlist deaths (Wilson

et al. 2009; Kirklin et al. 2013). Additionally, after months to a year on an LVAD, many patients

experience improvement in the global contractile properties of the heart (e. g. ejection fraction)

allowing their removal from the device. More recently, some VADs have been approved as a

destination therapy, providing individuals ineligible for a heart transplant with a long-term

therapy option (Wilson et al. 2009; Kirklin et al. 2013). However, there remains a subset of

Page 21: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

5

patients that do not experience a significant and/or long-lasting improvement on an LVAD.

Moreover, there is a great need to develop therapies that prevent end-stage heart failure.

Consequently, the field of cardiac tissue engineering emerged as a means of developing

alternative sources of cardiac tissue and methods for replacing tissue damaged by CVD.

Biomaterials have featured prominently in cardiac regenerative therapy and can be divided

among two main strategies for inducing functional repair to the heart muscle post-injury: (i) the

production of functional cardiac patches in vitro that can be implanted onto the damaged area

and thereby directly replace the non-viable portion of a damaged heart, and (ii) injectable

biomaterials (hydrogels) to help prevent cardiac remodeling, deliver cells to replace the damaged

tissue, and recruit endogenous cell types in an attempt to restore functionality (Menasché 2008).

In considering possible delivery of cells to the heart for tissue repair, the first issue that arises is

that of cell source.

2.2. Cell sources, stem cells, & differentiation

One of the biggest issues with cardiac repair is that adult cardiac cells do not proliferate, so

damaged tissue cannot repair itself. One cannot take a heart tissue biopsy, expand the cells to a

sufficient number, and re-introduce them back into the patient. This has led to the need to find a

cell source, ideally autologous, capable of differentiating into functional cardiomyocytes for use

in cardiac tissue engineering. Many cell types have been assessed or used as potential

cardiomyocyte replacements post infarction (Q. Z. Chen et al. 2008). The use of various adult

cell types including skeletal myoblasts and mesenchymal and hematopoietic stem cells have all

been assessed and although advantageous as they can come from autologous sources, results

have been varied, as will be discussed in coming sections. Neonatal cardiomyocytes are a

common cell source for they retain some proliferative capacity and have shown ability to couple

with adult myocytes, however their clinical use in humans is not feasible (Reinecke et al. 1999;

Reffelmann and Kloner 2003). They do however remain a good model cell source in developing

methods for regeneration as they provide a model of what can be achieved (Reffelmann and

Kloner 2003).

Embryonic and induced pluripotent stem cells (ESC and iPS cells, respectfully) are at the

moment the most promising cell sources for true myocardial repair. Studies from a number of

groups have shown that it is possible to generate CMs from mouse and human ESCs, and human

iPS cells (Kattman, Huber, and Keller 2006; Yang et al. 2008; M. Zhang et al. 2001).

Page 22: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

6

Flk1+ cells derived from ESCs and cultivated as embryonic bodies (EBs) were identified as

multipotent cardiovascular progenitors, and it was further demonstrated that Flk1 expression can

be used to enrich for cardiac specific progenitors (Kouskoff et al. 2005). When isolated from the

differentiated EBs, on day four of culture, and cultured as a monolayer, these progenitors

generate cultures highly enriched for contracting CMs. As these progenitors differentiate they

progress through the developmental stages thought to be involved in the establishment of the

cardiovascular lineages in vivo and the presence of specific cytokines is required. A combination

of activin-A and bone morphogenetic protein-4 over the first four days of EB differentiation

induces the initial stages of specification and subsequent exposure to dickkopf homolog 1

(DKK1) and vascular endothelial growth factor (VEGF) significantly enhances the

differentiation of Flk1+ progenitors into CMs (Kattman, Huber, and Keller 2006; Yang et al.

2008). The method reported shows the potential of the indentified progenitor to successfully

differentiate into the three cell types found in healthy adult tissue and thus is of great interest as a

cell source. There does remain some concern in that ESCs are associated with teratoma

formation due to their proliferative capacity and as they are not an autologous source there is

immune response issues, as well as the ethical issues of using ESCs (Vunjak-Novakovic et al.

2010).

A method for producing iPS cells from mouse, and subsequently human, somatic cells was

originally reported by Yamanaka and colleagues, and is significant as iPS cells present a much

needed autologous cell source for transplantation into the heart and differentiation into CMs and

vasculature (Takahashi and Yamanaka 2006; Takahashi et al. 2007). Problems with the reported

method, however, are low success rate at producing iPS cells (1/10000) and the use of a viral

vector to deliver the four factors needed to induce pluripotency. The latter is disconcerting as it

entails modification of the cells genome and thus raises questions of long-term effects. A new

method recently reported by Warren et al. (2010) uses mRNA to deliver the same transcription

factors as Yamanaka but with much higher efficiency and success rate at producing iPS cells

very close in phenotype to true ESCs. Furthermore, addition of further mRNA with

differentiation factors showed success at directing iPS cell fate (Warren et al. 2010). Advantages

of the newly reported method are that cell reprogramming to a pluripotent state and subsequent

differentiation can be achieved without compromising genetic integrity (Warren et al. 2010). As

advances in iPS cell production continues they remain a clear favorite in terms of a bench mark

Page 23: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

7

cell source for transplantation, however the concern of teratoma formation upon transplantation,

as with ESCs, remains.

2.3. Design criteria for biomaterials in cardiac tissue engineering

The complexity of the native heart environment and pathophysiology of post-MI remodeling

challenges the development of strategies for the treatment and design of biomaterials. An

important aspect of biomaterial design is the consideration of the objective of the biomaterial

application. Design criteria will be different whether the biomaterial is a vehicle for cell delivery,

a functionalized material aimed at artificially retaining normal ventricular geometry, or a

scaffold to generate tissue patches. However, there are some common design criteria that must

first be addressed: (i) biocompatibility, (ii) biodegradability, (iii) mechanical support, (iv)

injectability (in the case of hydrogels), (v) clinically relevant thickness (in the case cardiac

patches), and finally (vi) envisioned application time post-infarct (Leor, Landa, and Cohen 2006;

Q. Z. Chen et al. 2008; Vunjak-Novakovic et al. 2010; Bouten et al. 2011).

2.3.1. Biocompatibility Biocompatibility is generally defined as the “ability of a material to perform with an

appropriate host response in a specific application” (Williams 1987). In the context of cardiac

tissue engineering this encompasses the need for the material to function without initiating a

significant foreign body response in vivo, while retaining the ability to support both

cardiomyocyte survival in vitro and in vivo readily without cytotoxicity, as well as the contractile

function of the myocardium (Leor, Landa, and Cohen 2006; Q. Z. Chen et al. 2008; Vunjak-

Novakovic et al. 2010). This does not preclude the activation of the host inflammatory and

immune response but rather focuses on mitigating and controlling the type of response in order

to prevent further injury to the heart and not impede its function. Specifically, a biocompatible

material should be resistant to blood clotting and bacterial colonization, and if immunogenic

should not recruit cell types that can exacerbate the remodeling process. For example, activation

of the host immune response such that there is preferential recruitment of reparative M2

macrophages over cytotoxic M1 macrophages is generally considered to be a beneficial trait for

biomaterials.

2.3.2. Biodegradability

Page 24: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

8

Biodegradability refers to the mechanism through which an implanted material breaks down

and the inherent life-span of the material. A thorough discussion of the various mechanisms and

subsequent definitions are covered in detail elsewhere (Treiser et al. 2010). However, in brief,

there are three modes to consider: (i) bioerosion is degradation of a material through hydrolytic

mechanisms (covers both surface and bulk erosion); (ii) bioresorption is degradation through

cellular activity; and (iii) biodegradation is degradation through enzymatic activity. In the

context of cardiac tissue engineering, a biomaterial is considered to be biodegradable if

degradation occurs through disintegration, a hydrolytic mechanism or by enzymatic activity that

the biomaterial will encounter in vivo; and that the degradation products similarly conform to the

requirements of both biocompatibility and biodegradability (Leor, Landa, and Cohen 2006; Q. Z.

Chen et al. 2008; Vunjak-Novakovic et al. 2010; Bouten et al. 2011). While biocompatibility and

biodegradability are distinct concepts, they are often considered in tandem during biomaterial

design as there is little use in designing a biocompatible material that degrades into toxic

components.

Many physiological extracellular matrix (ECM)-based biomaterials readily fit these dual

criteria (e. g. fibronectin and collagen) as they inherently contain the correct molecular

composition required for cell attachment and survival, and they are readily degraded in vitro and

in vivo within days to weeks by enzymes secreted by cells into biocompatible and biodegradable

degradation products (Q. Z. Chen et al. 2008). Notably, cells can turnover these ECM

biomaterials and replace them with their own ECM components, thereby remodeling their

environment as necessary (Z. Li and Guan 2011). However, sourcing these materials can be

complicated by the fact that they may retain many of their surface antigens and may elicit an

immune response if used in xeno-transplantation. Despite this limitation there are sources of the

ECM-biomaterials that have been approved for human use, including fibrin that can be isolated

from a patient’s own blood (Odedra et al. 2011).

Conversely, synthetic materials have been developed such as polyethylene glycol (PEG), poly

(glycerol-sebacate) (PGS), and poly (tetrafluoroethylene) (PTFE) with defined chemical

compositions and designed to have no foreign body response. Modification of the chemical

composition can permit the selection of degradation rates in the range of a few weeks to years.

However there is the concern as to whether the degradation products are truly being removed

from the body or rather accumulate, the long-term effects of which are unknown (Q. Z. Chen et

Page 25: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

9

al. 2008; Z. Li and Guan 2011). An additional limitation is that these synthetic biomaterials often

do not support cell adhesion and survival, and therefore need to be modified with appropriate

bioactive molecules (Vunjak-Novakovic et al. 2010).

Another important consideration in terms of biodegradability is the issue of how quickly the

material should be removed in order to properly execute its desired function when applied for

cardiac ventricular repair. Biomaterials designed for cell delivery, recruitment, and survival

(anti-apoptotic, pro-angiogenic) should survive in vivo at least one week, based on the fact that

most cell death occurs within the first few days post-MI; and should be fully degraded in 6-8

weeks (in animal models) recognizing that pathological remodeling is complete by

approximately 6 weeks after MI (Patten et al. 1998; Krzemiński et al. 2008; Z. Li and Guan

2011). Thus, the biomaterial should remain long enough to have the desired effect but no longer

than necessary as it may become a hindrance to repair. For example, improved cardiac function

post treatment was demonstrated with fibrin glue used to transplant skeletal myoblasts into

ischemic myocardium wherein it degraded in 7-10 days (Christman, Vardanian, et al. 2004).

Scaffold degradation is likely to require a similar timeframe. Most scaffolds are designed to be

quickly replaced by new ECM secreted from the seeded cells, with the engineered tissues that are

implanted containing very little of the original scaffold material. For scaffolds that are present at

the time of implantation, in vivo degradation should not exceed weeks to months and should be

quickly replaced by functional tissues. Zimmermann et al. have spent considerable time

developing functional cardiac tissue constructs in vitro demonstrating that their collagen-based

scaffold can be remodeled and replaced by maturing cardiomyocytes resulting in a scaffold-free

transplantable engineered heart tissue construct (Eschenhagen et al. 1997; W.-H. Zimmermann

2001). In the case of biomaterials designed to provide support to the failing ventricle, they

should have relatively slow degradation rates on the order of months to years and while

controversy persists as to the timeframe required, it may be desirable for such materials to

remain for the very long-term (see Section 2.5.4 for examples and further discussion) (Nelson et

al. 2011). It is important to note however that there is still much debate as to the mechanical

properties and degradation requirements that are necessary for certain outcomes due to the large

disparity between studies (Nelson et al. 2011). These topics therefore continue to be active areas

of investigation.

2.3.3. Mechanical Support

Page 26: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

10

The third criteria of mechanical support requires some forethought as to the envisioned

application with consideration of whether the biomaterial can withstand the mechanical demands

that will be placed upon it after ventricular application and also whether it is likely to interfere

with the normal mechanical functioning of the surrounding tissue. The in vivo model system for

the biomaterial should be considered with respect to species-specific mechanical demands. This

is because the mechanical forces placed upon a biomaterial by the human heart will vastly differ

from the forces exerted by a small rodent heart in an animal application. Specifically, the human

myocardium ranges in stiffness from 20kPa (end of diastole) to 500kPa (end of systole), whereas

rat myocardium ranges from 0. 1 to 140kPa (Q. Z. Chen et al. 2008; Vunjak-Novakovic et al.

2010; Bouten et al. 2011; Venugopal et al. 2012). A material envisioned to artificially thicken the

ventricle wall and maintain ventricular geometry during remodeling should have a stiffness in

the high end of the range characteristic for the native ventricle; whereas a material designed to be

injected, to act as a temporary matrix for transplanted cells and/or to recruit endogenous cells can

have a low-end stiffness so long as it is sufficiently stiff to withstand the contraction/dilation of

the heart. Biomaterials intended for in vitro applications can have a very low stiffness, as long as

the cells seeded into it (hydrogels or scaffold) are able to remodel it into a material that as a final

product is mechanically similar to the native myocardium. This is especially evident in the case

of cardiac tissue grafts for repair of full thickness defects, wherein mismatching the mechanical

properties can result in the grave consequences of inducing undue strain on the injured heart if

the graft is too stiff or graft failure due to the stresses experienced in vivo if the graft is

insufficiently stiff (Ozawa et al. 2002). Furthermore, issues such as burst pressure and suture

retention must be considered as the graft has no time to integrate with the host tissue since it

experiences full cardiac load immediately upon implantation. Results from tissue engineering of

arterial grafts provide useful benchmarks for these properties that are achievable using current

tissue engineering methods e.g. burst pressure of over 3000mmHg and suture retention strength

of over 160g (L’Heureux et al. 2006; Dahl et al. 2011). Motivated by these challenges, Lang et al

have been developing a surgical glue to improve cardiac grafts and achieved success in repairing

a full thickness defect (N. Lang et al. 2014).

In general, naturally-derived biomaterials have weak mechanical properties, with moduli in

the tens of Pa to tens of kPa range (Q. Z. Chen et al. 2008). Moreover, there is a batch-to-batch

and source-to-source variability in the physical properties of these biomaterials. As a

Page 27: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

11

consequence, biomaterials made purely of naturally-derived components are limited as to their

mechanical support applications. Synthetic materials, on the other hand, are more consistent in

their composition between batches and have mechanical properties such as stiffness, elasticity,

and porosity that can be precisely controlled (Q. Z. Chen et al. 2008).

2.3.4. Injectability A hydrogel that can pass through a fine gauge needle (~27G) is described as injectable as it is

possible to safely administer it into the heart in a minimally invasive manner. Injectability can be

achieved by two approaches wherein gelation of the hydrogel (by temperature, chemical, light-

induced cross linking (Yeo et al. 2007; Habib et al. 2011), etc.) is: (i) initiated but not completed

prior to the hydrogel passing through the needle, and (ii) initiated after delivery to the desired

site. Importantly, the polymerization time should be in the order of minutes to tens of minutes to

ensure the hydrogel is delivered and successfully localized at the site of injection and not

completely washed out (Vunjak-Novakovic et al. 2010). This is because for polymerization that

requires tens of minutes to hours to complete there is enough time for the biomaterial to be

subjected to the contraction of the heart and to be carried away in the blood stream, rather than

gel properly in the ventricle wall.

2.3.5. Clinically Relevant Thickness Biomaterials used for tissue engineering strategies where cells are cultured with the

biomaterial in vitro then implanted in vivo have their own distinct requirement in that they must

be capable of supporting the cultivation of tissues of clinically relevant thickness: up to ~10 mm

for full thickness cardiac grafts, whereas tissue patches can be thinner (Chiu and Radisic 2010).

The limits of oxygen diffusion within a metabolically active tissue of high cell density (e.g.

108cells/cm3) restricts tissue thickness to approximately 200µm, thus scaffolds often require that

a primitive vascular network or a channel array for culture medium perfusion be incorporated

into the design to allow sufficient nutrient exchange to the centre of the tissues during in vitro

cultivation (Radisic et al. 2006). This is a major issue in producing in vitro cardiac patches that

will be discussed in greater detail in Section 2.4.

2.3.6. Application Time An additional consideration that can influence biomaterial design for cardiac regeneration

therapy is the time post-infarction at which the biomaterial is to be applied since new and old

infarcts present their own unique challenges. The rapid cell death that results from nutrient and

Page 28: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

12

oxygen deprivation downstream of a coronary artery blockage suggests that a cell injection-

based strategy should be most effective if applied shortly after an MI. The application of a

biomaterial modified with both cells and growth factors within hours or days after an MI may

promote directed wound repair such that the scar tissue formed would be minimized, the

contractile function maintained in the border zone, the ischemic area reduced, and consequently

pathological remodeling attenuated. Recent work has shown that early application of a collagen

based hydrogel 3 hours after MI significantly improved pathological remodeling when measured

4 weeks after treatment in comparison to application of gel 7 or 14 days after infarct (Blackburn

et al. 2014). Injection 7 days after MI did show significantly improved cardiac function in

comparison to injection 14 days after, which showed no improvement over PBS injection,

however not to the same level injection 3 hours after infarct imparted, thus indicating the earlier

the application the better the outcomes (Blackburn et al. 2014). Notably, incorrectly timed

administration of the therapy could potentially exacerbate the problem. While early delivery of

cells might conceptually be more effective by initiating early re-vascularization and contributing

to the protection of the spared myocardium, it may at the same time expose the delivered cells to

a very hostile environment because of the significant immune response, the presence of cell

death-associated cytokines and the by-products of dead and dying cells in the infarcted area soon

after insult, which can compromise the viability of the cells. The correct time to deliver cells has

been investigated in the clinic with both the TIME and SWISS-AMI trials showing that injection

of cells from 3 days to 4 weeks after infarct did not improve LV cardiac functional or

pathological outcomes (Traverse 2011; Traverse 2012; Sürder et al. 2013). A scaffold-based

contractile tissue engineering strategy while applicable in the acute phase may have a more

significant effect if implanted after scar formation. Similarly, larger areas of damaged cardiac

muscle evident in chronic cases might benefit from a scaffold-based regenerative therapy

approach. Therefore, choosing the right time point post-MI for an intervention is a challenge and

no clear consensus has been reached.

2.4. Tissue engineered cardiac grafts (in vitro engineering)

In vitro tissue engineering approaches involve pre-organizing cells seeded into scaffolds or on

their own into a functional tissue (i.e. one capable of contracting and propagating electrical

signals). To be clinically relevant any engineered cardiac tissue must have properties (functional

and morphological) similar to that of native heart tissue and remain viable after implantation

Page 29: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

13

(Wolfram-Hubertus Zimmermann, Melnychenko, and Eschenhagen 2004). Challenges in the

development of such in vitro tissues is that oxygen diffusion limits tissue sizes to 100-200 µm,

thus vascularization is required, and generating tissues that can electromechanically couple with

the native myocardium (and generate significant contractile force) (Vunjak-Novakovic et al.

2010). A number of different groups have tried to tackle these problems using different

approaches ranging from using decellularized tissue as a scaffold, using mechanical/electrical

stimulation of cells seeded in hydrogel scaffolds, to layering individual cell sheets.

In an effort to improve the properties of engineered heart tissue (EHT) Eschenhagen et al.

(1997) seeded a mix of collagen I, extracellular matrix proteins (Matrigel), and neonatal rat

cardiomyocytes into lattice or circular molds and saw spontaneous remodeling of the liquid

reconstitution mixture and a development of spontaneously and synchronously contracting solid

EHTs after 5-7 days (Eschenhagen et al. 1997). Subsequent culture of EHTs with cyclic

mechanical strain improved morphological, functional, and mechanical properties of the EHT,

and by stacking a number of these constructs together formed spontaneously contracting tissue 1-

4 mm thick that could be implanted at the site of infarct in a rat MI model. One month later it

was observed that the constructs showed un-delayed electrical coupling with the native tissue

and improved diastolic and systolic function of rats who received the patches in comparison to

sham-operated rats (W. H. Zimmermann et al. 2006).

Another approach to mechanical stimulation for achieving functional improvement of EHT

was noted by Radisic et al. (2004). It was demonstrated that neonatal cardiomyocytes seeded on

collagen sponges with Matrigel and exposed to physiologically relevant electric field stimulation

during culture induced the formation of mature myocardium with elongated, viable cells aligned

in parallel. Ultra-structural organization in stimulated cardiac constructs was remarkably similar

to that present in the native myocardium, with hallmarks of electromechanical cell coupling

including gap junctions, intercalated disks and sarcomeres all markedly more frequent than in

non-stimulated constructs (Radisic et al. 2004).

A pioneering study done by Ott et al. (2008) looked to tackle the larger problem of whole-

organ transplant, while at the same time illustrating an approach for answering the questions of

oxygen transport and tissue thickness in engineered heart tissues. By decellularizing whole,

adult, cadaveric Fisher rat hearts by coronary artery perfusion of detergents they were able to

obtain whole heart scaffolds. The process preserved the underlying extracellular matrix (ECM),

Page 30: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

14

and produced an acellular scaffold with perfusable vascular architecture, competent acellular

valves and intact chamber geometry. Reseeding the scaffolds with neonatal rat CMs or

endothelial cells and culturing by perfusion in a bioreactor mimicking cardiac physiology they

observed macroscopic contractions by day four. At day eight they were able to show a small, but

significant pump function comparable to about 2% of the adult heart when the engineered hearts

were exposed to physiological loads and electrical stimulation (Ott et al. 2008).

A method for developing functional EHT was also accomplished using no scaffold at all, but

stacking sheets of neonatal rat cardiomyocytes cultured on the surface of Poly(N-

isopropylacrylamide)-grafted polystyrene dishes. The treated dishes allow for cell attachment at

37°C, but culturing at 20°C changes the properties of the surface causing cell layers to detach, at

which point individual layers can be stacked, and these stacked sheets looked like homogeneous

heart-like tissue (Miyagawa et al. 2005). When these scaffold free cell stacks were transplanted

onto infarcted rat hearts they became attached to the infarcted myocardium, showed

angiogenesis, expressed connexin-43 (a gap junction protein associated with functionally

coupled cells), appeared as a homogeneous tissue in the myocardium, and significantly improved

cardiac performance (determined by echocardiography) in comparison to those that did not

receive the treatment (Miyagawa et al. 2005). Furthermore, connexin-43 staining and

transmission electron microscopy (TEM) showed the existence of gap junctions and intercalated

disks (associated with coordinated cardiac muscle contraction) between implanted and host

tissue. One limitation of this technique is that it is still limited to the hundreds of µm in thickness

unless methods for oxygen transport or vascularization are introduced.

Many of the issues with engineering tissue in vitro are the same as those for direct in vivo

application (injection methods), including appropriate biomaterial (scaffold) choice, cell source,

and survival/integration once transplanted, thus the two methods are not mutually exclusive.

Injection of cells/hydrogels into heart tissue shortly after MI has the potential to minimize the

formation of scar tissue and attenuate the pathological remodeling process whereas tissue

engineered cardiac patches may be more useful in later stages of remodeling such as the

complete replacement of non-contractile areas. In vitro tissue engineering may also provide

living patches for repair of congenital malformations that cannot be addressed through injectable

methods (Soonpaa and Field 1998).

2.5. Injection (in vivo engineering)

Page 31: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

15

The first type of injection that must be discussed is that of direct cell transplantation. Based on

the successes and limitations from such injections the use of biomaterials was developed to aid in

the successful transplantation of cells. In general, injectable hydrogels for cardiac regeneration

and repair can be divided into three categories according to their state of development and

current application. The first group of hydrogels is designed to promote endogenous repair

through maintaining cell survival, recruiting endogenous cells or inducing neovascularization.

The second group acts a temporary matrix for cell transplantation and exogenous repair. The

final group consists of injectable hydrogels that act as bulking material to support the failing left

ventricle, in turn maintaining or restoring normal geometry of the heart, and improving cardiac

function. It should be noted that there is a significant overlap between the categories, and most

hydrogels fit into more than one category.

2.5.1. Cell only based therapies The first type used in clinical studies for cell based cardiac repair was skeletal myoblasts and

results were varied. Skeletal myoblasts were initially chosen due to their relative ease of isolation

and culture (expansion) from small muscle biopsies, pre-clinical efficacy, non-immunogenicity,

and ability to cope well with ischemic environments (Menasché et al. 2003). Injection showed

reduced infarct size, survival of transplanted cells, and slight improvement of cardiac function,

however it was found that most cells did not trans-differentiate into cardiomyocytes (as hoped,

becoming skeletal muscle) and therefore lack the proteins needed to electromechanically couple

with the native myocardium and it was reported that the grafts did not beat in synchrony with the

surrounding tissue (Reinecke, Poppa, and Murry 2002; Reinecke et al. 2000). Even so, Phase I

trials were deemed successful and Phase II trials are currently underway.

Neonatal cardiomyocytes retain the ability to proliferate and have received much attention as

a potential cell source. Injection of CMs isolated from neonatal rats and transplanted to the site

of infarct in adults have shown attenuation of pathological remodeling leading to improved

function, and even integration of cells into the host myocardium through gap junctions and

intercalated disks (Reinecke et al. 1999; Muller-Ehmsen 2002; R. R. K. Li et al. 1996). Further

studies, however, show that direct injection of these cells results in survival of only ~50% of

cells immediately after injection, loss of up to ~90% of cells due to extrusion, and only ~10%

survival of the remaining cells after as little as one week (Müller-Ehmsen et al. 2002; M. Zhang

et al. 2001).

Page 32: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

16

Mesenchymal and hematopoietic stem cells have also been looked at as potential cell sources

for direct injection and have had some promising results. They are advantageous for the two cell

lines have multi-lineage differentiation potential, and in vitro studies showed promise in the two

cell type’s ability to become cardiomyocytes. Direct injection of these cell types into MI areas

showed incorporation into host myocardium, reduced infarct size, and even showed functional

improvements, however it has been noted that fusion events are rare and no significant muscle

mass has been generated in vitro (Bittira et al. 2002; Makino et al. 1999; Shake et al. 2002; Orlic,

Kajstura, Chimenti, Jakoniuk, et al. 2001; Murry et al. 2004).

Perhaps the most interesting observation about cell transplantation is that regardless of cell

type, developmental stage, or origin most studies showed some improvement in cardiac function

post injection; yet all seem to have the same issues with incorporation into the native tissue,

survival, and retention (W. H. Zimmermann et al. 2006). Meta-analysis of clinical trial results

demonstrated a significant, albeit low 3%, increase in left ventricular ejection fraction (LVEF) as

well as a significant reduction in infarct size (-5.6%) and end systolic volume (-7.4ml) in patients

treated by intracoronary cell injection after acute MI (Lipinski et al. 2007). Dose-response

between injected cell volume and LVEF change was reported (Lipinski et al. 2007). Although

these studies are encouraging, modest improvements motivate investigation of new cell sources

and methods that increase survival and retention of injected cells.

It has also been suggested that the improvements seen with cell transplantation may be due to

release of cardio-protective and angiogenic cytokines from transplanted cells that modulate

remodeling rather than integration of the cells into the host myocardium (Menasché 2008). As

such, groups have also looked at identifying specific cytokines and directly injecting them in

place of cells. While the delivery of cells to the injury site is a major aspect of cardiac

regeneration therapy and the cell types used are many and varied, the topic is discussed in further

detail elsewhere (Shiba et al., 2009; Hilfiker et al., 2011; Martinez and Kofidis, 2011; Liau et al.,

2012).

2.5.2. Hydrogels to promote endogenous repair The group of injectable hydrogels that promote endogenous repair works based on either the

bioactivity of the base biomaterial or the incorporation of bioactive molecules that are anti-

apoptotic or cardioprotective (i.e. QHREDGS, Tβ4, insulin-like growth factor-1), angiogenic

(i.e. Tβ4, bFGF, VEGF, PDGF, hepatocyte growth factor, pleiotrophin plasmid), or chemotactic

Page 33: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

17

(i.e. stromal cell-derived factor-1). The incorporation of bioactive molecules into injectable

hydrogels is important for the successful delivery of the biomolecules and in turn cardiac

regeneration, since biomolecules in the soluble form are rapidly diffused from the injection site

and proteolytically degraded in the in vivo environment.

The existence of different cardiac progenitor cell types has been reported in native heart

tissue, and it was demonstrated that these progenitors can be induced to migrate to areas of

infarct with the addition of specific peptides or cytokines (Beltrami et al. 2003; Oh et al. 2003;

Martin et al. 2004; Cai et al. 2003). One such peptide is thymosin β-4 (Tβ4) which has been

shown to promote migration and survival of CMs, endothelial cells, smooth muscles cells, and

vascular progenitors from cardiac explants in vitro and has garnered attention for clinical trials as

a drug for patients with acute MI (Smart et al. 2007; Bock-Marquette et al. 2004; Crockford

2007).

A temperature-responsive hydrogel made of chitosan and glycerol phosphate was used to

encapsulate bFGF, which was released through hydrogel degradation (H. Wang et al. 2010).

After 4 weeks of injection in a rat MI model, the bFGF-hydrogel improved cardiac function (i.e.

decreased LV end-diastolic diameter and LV end-systolic diameter, as well as increased LV

ejection fraction and LV fractional shortening), as compared to PBS control, bFGF in PBS, and

chitosan hydrogel alone (H. Wang et al. 2010). This may be due to the increase in arteriole

density and in turn decreased infarct size in the bFGF-hydrogel group compared to all other

groups. The chitosan hydrogel alone also increased arteriole density and decreased infarct size

compared to PBS control and bFGF in PBS (H. Wang et al. 2010). This shows that the presence

of the injectable hydrogel was necessary for mechanical support and sustaining the release of

encapsulated bFGF, while the incorporation of bioactive molecules such as bFGF further

improved endogenous cardiac repair through promoting arteriogenesis. By contrast, treatment

with soluble bFGF showed no improvement compared to PBS control due to the short half life of

the growth factor and its high diffusibility from the injection site (H. Wang et al. 2010). Other

groups also investigated the incorporation and release of bFGF from various hydrogel systems,

including encapsulation into biodegradable gelatin hydrogel microspheres (Iwakura et al. 2003;

Yamamoto et al. 2001; Y. Liu et al. 2006), UV crosslinkable chitosan hydrogels (Fujita et al.

2005), and synthetic random copolymer poly(N-isopropylacrylamide-co-propylacrylic acid-co-

butyl acrylate) (Garbern et al. 2011). These studies all showed increased vascular density with

Page 34: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

18

the injection of bFGF hydrogels, and in turn improved cardiac function after myocardium

infarction in rats, rabbits, or dogs.

Conjugation of VEGF to a temperature-sensitive, aliphatic polyester hydrogel, poly (δ-

valerolactone)-block-poly (ethylene glycol)-block-poly (δ-valerolactone) (PVL-b-PEG-b-PVL),

showed that the hydrogel provided localized and sustained VEGF activity, which increased

blood vessel density (Wu et al. 2011). While the hydrogel alone maintained thicker ventricular

wall and improved cardiac function (i.e. fractional shortening, ventricular volumes, preload

recruitable stroke work, end-systolic elastance) after MI in rats, the conjugation of VEGF to the

hydrogel further enhanced cardiac repair (Wu et al. 2011).

While alginate, a bio-inert natural material, has been shown to prevent cardiac remodeling and

dysfunction in the rat MI model, it has also been modified with peptides and growth factors to

create a bioactive injectable hydrogel for endogenous cardiac repair (Landa et al. 2008). Alginate

that was modified with RGD conjugation led to increased angiogenic response compared to non-

modified alginate when injected into the infarct area in rats 5 weeks post-MI (Yu, Gu, et al.

2009).

In addition, alginate hydrogel was found to be an effective injectable delivery system for

cardiac repair post-MI, since it could sequentially deliver VEGF and PDGF to induce formation

of mature blood vessels and in turn improve cardiac function (Hao et al. 2007). In this alginate

system, PDGF was released more slowly than VEGF likely due to the difference in the affinities

of VEGF and PDGF to alginate. As such, VEGF induced angiogenesis, while PDGF was present

in the later time to mature the newly formed capillaries. This sequential delivery approach

improved vascularization and cardiac repair compared to the delivery of VEGF or PDGF

individually (Hao et al. 2007).

Injectable affinity-binding alginate hydrogel microbeads were also used to sequentially

release cytoprotective IGF-1 and pro-angiogenic HGF to increase angiogenesis and formation of

mature blood vessels, as well as to prevent infarct expansion and scar fibrosis after 4 weeks in a

rat acute MI model (Ruvinov, Leor, and Cohen 2011). The injection of these hydrogel

microbeads also reduced cell apoptosis, induced cell cycle re-entry of CMs and enhanced

presence of GATA-4 positive cells, indicating an improved endogenous cardiac regeneration

(Ruvinov, Leor, and Cohen 2011).

Page 35: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

19

Hydrogels have also been used to enhance gene therapy. The human VEGF plasmid was

injected in a rat MI model with an amphiphilic, thermo-responsive polymer synthesized by

alternatively cross-linking poloxamer and di-(ethylene glycol) divinyl ether (Kwon et al. 2009).

The sustained human VEGF expression within the infarct area increased capillary density and

formation of larger blood vessels. In a separate study, the pleiotrophin plasmid was shown to

induce neovasculature formation in the ischemic rat myocardium without angioma formation, but

the injection of the plasmid in saline resulted in low transfection efficiency due to limited

exposure of cells to the plasmid (Christman et al. 2005). The incorporation of pleiotrophin

plasmid in fibrin glue created a gene-activated matrix that increased neovasculature formation

compared to injection of the plasmid in saline.

To deliver stromal cell-derived factor-1 (SDF-1) as a chemokine to attract stem cells for

cardiac regeneration, a modified chemokine S4V was designed to retain chemotactic bioactivity

while being resistant to cleavage by matrix-metalloproteinase-2 and exopeptidase (Segers et al.

2007). S4V was then tethered to self-assembling peptides that formed nanofibers suitable for

local delivery. The intra-myocardial delivery of S4V-nanofibers post-MI in rats led to the

recruitment of c-kit positive stem cells and increased capillary density, as well as improved

cardiac function (i.e. increase in ejection fraction) (Segers et al. 2007).

Hydrogels derived from the native cardiac extracellular matrix (ECM) retain the complexity

of the extracellular environment normally found in vivo and were demonstrated to improve

maturation of cardiomyocytes derived from human embryonic stem cells (DeQuach et al. 2010),

thus they constitute a promising approach for myocardial repair (Duan et al. 2011). ECM

components have been isolated from the healthy myocardium and used as injectable hydrogels

for endogenous cardiac repair. Cardiac ECM-derived hydrogels are prepared by decellularizing

heart tissues with sodium dodecyl sulfate, followed by lyophilization and milling into a fine

powder that is then solubilized by enzymatic digestion using pepsin and HCl and neutralized

using NaOH (Singelyn et al. 2009; Seif-Naraghi et al. 2010; Singelyn et al. 2012). Myocardial

matrix yielded from the decellularization of porcine myocardial tissue gelled at 37°C and showed

collagen and glycosaminoglycan content (Singelyn et al. 2009). In addition, the matrix induced

the homing of endothelial and smooth muscle cells, as shown by cell migration towards the

matrix in vitro, as well as cell infiltration into the gel and increased arteriole formation in vivo 11

days post-injection within the rat myocardium. While the solubilized form of the decellularized

Page 36: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

20

porcine pericardium was a stronger chemo-attractant for endothelial and smooth muscle cells,

both decellularized porcine and human pericardium were shown to retain the native protein and

glycosaminoglycan and induce neovascularization when injected into the left ventricular free

wall of rats (Seif-Naraghi et al. 2010). Moreover, a low number of c-kit+ cells were found within

the injection site, indicating an increase in endogenous CMs with the injection of the material

(Seif-Naraghi et al. 2010; Singelyn et al. 2012). As such, the use of decellularized cardiac matrix

as injectable hydrogels can promote endogenous homing and in turn cardiac-specific tissue

formation, as mediated through the presence of cardiac-specific cues. In addition, cardiac

function was maintained without arrhythmias in the rat MI model, when treated with the ECM

hydrogel (Singelyn et al. 2012). Besides demonstrating the use of decellularized ECM in small

animal models, it has also been shown that hydrogels made from decellularized porcine

ventricular ECM can be delivered via percutaneous, transendocardial catheter injection in both

healthy and infarcted porcine myocardium (Singelyn et al. 2012). The injection was successful,

with no catheter clogging. While there was some leakage of the injectate into the ventricle, it was

confirmed that a hydrogel was formed within the porcine myocardium without any hydrogel

found in other organs, since any leaked matrix would be rapidly diluted in the blood and

prevented from gelation. This study shows the feasibility of delivering in situ gelling materials

via transendocardial injection in a large animal model and motivates the translation of injectable

materials for MI treatment in humans (Singelyn et al. 2012).

Keeping with ECM derived materials, gels derived from the small intestinal submucosa

extracellular matrix were found to be a suitable injectable material for cardiac repair, due to the

presence of bFGF (Okada et al. 2010). The gel induced angiogenesis and reduced infarct size

compared to saline control in the murine MI model (Okada et al. 2010). In another study, the

injection of small intestine ECM emulsion into the myocardium recruited c-kit positive cells,

myofibroblasts and macrophages after rat MI (Zhao et al. 2010). It also increased VEGF levels,

enhanced angiogenesis and improved cardiac function (i.e. fractional shortening, ejection

fraction, stroke volume) compared to saline control, likely mediated by the cell recruitment

(Zhao et al. 2010).

Similarly, fibrinogen, the precursor to fibrin, has also been used in biomaterial strategies.

Rufaihah et al have demonstrated the applicability of a photocrosslinkable semi-synthetic PEG-

fibrinogen hydrogel loaded with VEGF in rat models of MI. They showed that a PEG-fibrinogen

Page 37: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

21

hydrogel was able to store and release VEGF in a sustained and controlled fashion, as well as

significantly improve arteriogenesis and cardiac performance after infarct (Rufaihah et al. 2013).

With tunable mechanical properties they have also shown the ability of the PEG-fibrinogen

hydrogel on its own to mechanically support the failing LV and to improve cell transplantation,

giving the developed hydrogel multiple courses of action as well as promise as a clinically

relevant biomaterial (Habib et al. 2011; Rufaihah et al. 2013).

G-CSF has also been studied in depth but has seen varied results in experiments with various

animal species (Orlic, Kajstura, Chimenti, Limana, et al. 2001; Iwanaga et al. 2004; Deten et al.

2005). While it undoubtedly promotes migration of progenitors to the site of infarct the

population is most likely non-cardiac lineage. It has been suggested, and to some extent shown,

by a number of groups that G-CSF may not induce cardiac regeneration but mitigate the damage

done during MI and influence cardiac repair (Nygren et al. 2004; Minatoguchi et al. 2004). One

group showed in mice that G-CSF treatment reduced final infarct size and resident cell apoptosis,

and increased vascular density and ventricular function in comparison to mice who did not

receive the treatment (Harada et al. 2005). Despite some conflicting studies and varied results G-

CSF has already entered clinical trials being administered to patients with recent MI, and has

shown some promising results (H.-J. Kang et al. 2004; H.-J. Kang et al. 2007; J. Kang et al.

2014).

2.5.3. Hydrogels for the delivery of cells for regeneration Early studies in cell transplantation for cardiac repair lacked control over cell retention,

survival, and function after injection of the cells into the heart. Later studies used natural and

synthetic injectable hydrogels that polymerized in situ as a cell delivery vehicle to overcome

such limitations. Some hydrogels used in cell delivery for cardiac regeneration include fibrin,

chitosan, collagen, Matrigel™, and various synthetic polymers.

Fibrin was one of the earliest biomaterials studied for use in the heart and for cell

transplantation. It exhibits several advantages: it is naturally occurring and could be patient-

specific; it is biocompatible, biodegradable, and pro-angiogenic. Furthermore, fibrin glue has

ideal properties for cell delivery in cardiac repair due to its binding domains for growth factors

and receptors. A three-dimensional fibrin hydrogel is formed upon injection with a dual-barreled

syringe, which contains fibrinogen and fibrinolysis inhibitor aprotinin in the first barrel and

thrombin and CaCl2 in the second barrel (Christman, Fok, et al. 2004). The cross-linking

Page 38: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

22

mechanism of the fibrin hydrogel is similar to how clotting occurs in vivo. Skeletal myoblasts

that were injected with fibrin glue into the myocardium post-MI were found in both the border

zone and within the infarct, while myoblasts were found only in the border zone when injected in

bovine serum albumin (Christman, Fok, et al. 2004). Fibrin gel provided a temporary scaffold for

transplanted cells and contained RGD motifs that bind to cell receptors, thus entrapping the cells

upon injection and improving their survival (Christman, Vardanian, et al. 2004). The surviving

myoblasts at five weeks post-injection into the myocardium were located in clumps around

arterioles (Christman, Vardanian, et al. 2004). The injection of fibrin glue with or without

skeletal myoblasts led to smaller infarcts compared to injection of bovine serum albumin or

myoblasts in bovine serum albumin (Christman, Vardanian, et al. 2004). There was also

increased arteriole density in the infarct area when skeletal myoblasts were delivered in the fibrin

glue compared to delivery in bovine serum albumin (Christman, Vardanian, et al. 2004).

Although fibrin seemed to significantly decrease infarct size and increase arteriole formation

without the presence of transplanted cells, it could be seen that transplantation of cells without an

appropriate injectable hydrogel was not capable of appreciable cardiac repair (Christman, Fok, et

al. 2004; Christman, Vardanian, et al. 2004).

In a separate study, rat adipose-derived stem cells isolated from subcutaneous adipose tissues

were injected in fibrin glue into the rat left ventricular wall post-MI (X. Zhang et al. 2010). At 4

weeks after injection, the co-injection of cells and hydrogel showed greater graft size, better

cardiac function (decreased LV end-diastolic diameter and LV end-systolic diameter, as well as

increased fractional shortening and ejection fraction), and increased arteriole density compared

to injection of cells alone (X. Zhang et al. 2010). In addition, another study was conducted to

deliver human bone marrow-derived mesenchymal stem cells in fibrin glue in a nude rat MI

model using percutaneous injection catheters (Martens et al. 2009). Fibrin glue increased cell

retention and survival. More importantly, this study determined the viscosity limits for the

delivery of the cell-hydrogel suspension using several commercially available catheters. By

controlling the composition of fibrinogen and thrombin, the fibrin hydrogel could be tailored to

be compatible with the catheters. This also shows the potential of using catheter delivery for cell

transplantation therapies involving other cell types and hydrogels (Martens et al. 2009). While

fibrin-based biomaterials show promise and have the additional benefit of fibrin being FDA

approved for human use, these biomaterials may only provide short-term cardiac functional

Page 39: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

23

benefits that decrease with time (Yu, Christman, et al. 2009; Nelson et al. 2011; Segers and Lee

2011).

Another natural hydrogel used for cell transplantation is a temperature responsive hydrogel

made of chitosan and glycerol phosphate similar to that described by Wang et al above (Lu et al.

2009). This hydrogel gels in approximately 10 to 15 minutes at physiological temperatures.

Embryonic stem cells were injected into the infarcted heart wall of rats in the chitosan hydrogel,

showing better cell retention at 24 hours and increased graft size, improved cardiac function,

thicker ventricular wall and greater micro-vessel density at 4 weeks as compared to injection of

the cells in phosphate buffered saline (PBS) (Lu et al. 2009). Histological staining of excised

heart sections showed a significant presence of chitosan at 24 hours after injection, while sparse

positive chitosan staining was found at 4 weeks. No trace of the hydrogel was found at 6 weeks

post-injection, showing complete degradation of the hydrogel in vivo (Lu et al. 2009). Chitosan

has also been explored for the delivery of adipose-derived mesenchymal stem cells into the

infarcted heart (Z. Liu et al. 2012). The chitosan hydrogel improved MI microenvironment by

recruiting SDF-1, which is a key chemokine for homing of stem cells, and scavenging reactive

oxygen species generated by ischemia that would otherwise impair adhesion molecules of the

delivered stem cells (Z. Liu et al. 2012).

Pre-treatment of animals with gelatin hydrogel microspheres incorporating bFGF was used to

improve transplantation of fetal rat CMs in rat MI models (Sakakibara et al. 2002). It has been

shown that bFGF can enhance viability of CMs through neovascularization and also directly

affect CMs by stimulating DNA synthesis, myocyte proliferation and differentiation during

development. In the experimental groups with bFGF microspheres alone and with bFGF

microspheres followed by CM transplantation, there was neovascularization in the scar tissue

after 1 week. Importantly, more transplanted cells survived in the scar area with the pretreatment

with bFGF microspheres compared to no pretreatment, in which transplanted cells only survived

in the peri-infarct regions (Sakakibara et al. 2002).

In another study, human embryonic stem cell (hESC)-derived CMs were delivered to the heart

within Matrigel™ using a cocktail of pro-survival factors (M. a Laflamme et al. 2007). The pro-

survival cocktail resulted in a 4-fold improvement in myocardial graft size relative to hESC-CMs

injected with Matrigel™ alone after 1 week. After 4 weeks, increased LV wall thickening and

reduced ventricular dilation, improved global function (fractional shortening and ejection

Page 40: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

24

fraction), and attenuated changes in LV chamber dimensions were observed in hearts injected

with hESC-CMs along with the pro-survival cocktail relative to pro-survival cocktail-only,

buffer-only, or non-cardiac cell controls (M. a Laflamme et al. 2007).

An example of a synthetic hydrogel aimed at improving cell transplantation therapies to

reduce LV remodeling post-MI is an injectable α-cyclodextrin/poly(ethylene glycol)–b-

polycaprolactone-(dodecanedioic acid)-polycaprolactone–poly(ethylene glycol) (MPEG–PCL–

MPEG) hydrogel (T. Wang, Jiang, et al. 2009). Culturing bone marrow-derived stem cells

(BMSCs) in the hydrogel showed it was non-toxic and allowed for the maintenance of cell

morphology in vitro. In a rabbit LV MI model co-injection of α-cyclodextrin solution with

BMSCs and MPEG–PCL–MPEG solution in the infarct area 7 days post-MI showed immediate

gelation and localization (T. Wang, Jiang, et al. 2009). Histological analysis of excised heart

cross sections 4 weeks after injection showed the hydrogel was absorbed (degraded), and that the

hydrogel significantly increased BMSC retention and vessel density around the infarct compared

to injecting BMSCs alone. Functional echocardiography studies demonstrated an increased LV

ejection fraction (~77% increase) and attenuated left ventricular dilatation with hydrogel-BMSC

co-injection compared to PBS and BMSC only controls (T. Wang, Jiang, et al. 2009).

A synthetic injectable hydrogel made of oligo[poly(ethylene glycol) fumarate] (OPF) was

developed to deliver mouse embryonic stem cells into the LV wall of a rat MI model (H. Wang

et al. 2012). The hydrogel improved cell retention compared to delivery in PBS, and was

biodegradable with complete degradation by 6 weeks after injection in the infarcted heart. The

hydrogel alone significantly reduced the infarct size and improved cardiac function at 4 weeks

after injection (H. Wang et al. 2012). However, the combination of cells and hydrogel further

increased revascularization, improved cardiac function, decreased infarct size and fibrotic area,

as compared to hydrogel alone and cells injected in PBS. Importantly, the transplanted

embryonic stem cells expressed cardiovascular markers, such as cardiac troponin-T, von

Willebrand factor and α-smooth muscle actin. As such, the OPF hydrogel not only supported cell

transplantation by improving cell retention and survival, but the stem cells also underwent

differentiation into the cardiac linage, thus showing potential of this delivery method for heart

regeneration (H. Wang et al. 2012).

Synthetic hydrogels have also been modified by incorporation of bioactive peptides and

growth factors to improve their use for cell delivery. A synthetic PEG-based hydrogel modified

Page 41: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

25

with matrix metalloproteinase (MMP) cleavable peptide domains was developed for the co-

delivery of Tβ4 and vascular cells (Kraehenbuehl et al. 2009). Tβ4 was physically entrapped in

the hydrogel, which gels in approximately 30 minutes at physiological conditions. Due to the

MMP cleavable peptide domains, the hydrogel was prone to cell-mediated proteolytic

degradation and the encapsulated Tβ4 could be released due to the MMP secretion of

encapsulated human umbilical vein endothelial cells in vitro. At the same time, the presence of

Tβ4 led to improved adhesion, survival, migration and organization of the cells (Kraehenbuehl et

al. 2009). In vivo, the MMP-responsive PEG hydrogel was used to co-deliver Tβ4 and hESC-

derived vascular cells into the infarcted myocardium of rats (Kraehenbuehl et al. 2011). By 6

weeks after MI, there was improved cardiac function (i.e. decreased end-systolic volume and

increased ejection fraction) in the group with vascular cells injected in Tβ4-encapsulated

hydrogel, as compared to treatment with PBS control, Tβ4-free hydrogel and Tβ4-encapsulated

hydrogel. This improvement was due to the better organization of native CMs and endothelial

cells in the combined Tβ4 and cell delivery, as supported by the presence of Tβ4 and the

secretion of human vascular cytokines (i.e. VEGF, EGF, HGF) and pro-survival factors (i.e.

survivin) by the transplanted vascular cells (Kraehenbuehl et al. 2011). They hypothesized that

the gel provided temporary support and pro-survival factors as it substituted for the degrading

endogenous matrix, and hESC-derived vascular cells contributed to formation of capillary-like

vessels, stabilization of host vessels, secretion of paracrine factors, or induction of paracrine

factor secretion from native rat cells (Kraehenbuehl et al. 2011). The injection of cell-free PEG-

hydrogel with Tβ4 improved cardiac function compared to PBS control, showing that the

injection of the bioactive hydrogel alone could also be explored for endogenous cardiac repair

(Kraehenbuehl et al. 2011).

Injection of a synthetic self-assembling hydrogel RAD16-II (initially developed by Zhang et

al. 1993) formed nanofiber microenvironments in the host myocardium and promoted

recruitment of endogenous ECs and vascular smooth muscle cells, which appeared to form

functional vascular structures, necessary for improving blood flow and improving the general

environment for CM survival. Furthermore, co-injection of the self-assembling peptide with

CMs showed survival (albeit low) of the transplanted cells, but significant increase in

recruitment of endogenous progenitors (M. E. Davis et al. 2005). The addition of insulin-like

growth factor-1 (IGF-1, a CM growth and differentiation factor) proved to be anti-apoptotic and

Page 42: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

26

increased cell growth in transplanted (injected) CMs. It showed sustained, controlled, and

targeted release of IGF-1 from the nanofibers for 28 days. Improved systolic function was

observed when CMs were injected with the new functionalized hydrogel in a rat MI study (M. E.

Davis et al. 2006).

2.5.4. Hydrogels for the artificial maintenance of ventricle geometry and repair The third approach for preservation of cardiac function is the injection of a bulking material

into the ventricular wall to prevent progressive adverse remodeling due to high wall stresses that

develops after MI. Theoretical modeling suggests that hydrogels change the geometry and

dilation mechanics of the ventricle in such a way as to reduce the elevated local wall stresses,

and further that the mechanical support provided by the hydrogels improves the ejection fraction

and the stroke volume/end-diastolic volume relationship (Z. Li and Guan 2011; Wall et al. 2006).

To this end Fujimoto et al developed an N-isopropylacrylamide (NIPAAm), acrylic acid (AAc),

and hydroxyethyl methacrylate-poly(trimethylene carbonate) (HEMAPTMC) based synthetic

hydrogel (Fujimoto et al. 2009). It was found that a feed ratio of 86/4/10 poly(NIPAAm-co-AAc-

co-HEMAPTMC) monomers formed a hydrogel at 37°C, which gradually became soluble over a

5 month period, and furthermore that no degradation product cytotoxicity was observed in vitro

(Fujimoto et al. 2009). The hydrogel or PBS was injected into the MI zone two weeks after MI in

a rat model, and analysis was done 8 weeks after injection. In the PBS group, LV cavity area

increased and contractility decreased at 8wk post-MI, while in the hydrogel group both

parameters were preserved during this period, and furthermore the hydrogel was still present at 8

weeks (Fujimoto et al. 2009). Echocardiography was used to measure heart function and it was

reported that the hydrogel maintained fractional area change (contractility) at the same level

measured just prior to injection, and was ~55% better than the PBS group at 8 weeks post

injection (10 weeks post MI). Tissue in-growth was observed in the hydrogel injected area and a

thicker LV wall (~100% thicker) and higher capillary densities were found for the hydrogel

versus PBS group (Fujimoto et al. 2009).

Thermo-responsive materials that transform from solution to gel near body temperature are

frequently employed for tissue engineering because their transitional properties make them easy

to deliver and manipulate. T. Wang et al developed a series of hydrogels by introducing

hydrophobic PCL (polycaprolactam)-grafted polysaccharide chains into the thermo-responsive

poly(N-isopropylacrylamide) (PNIPAAm) network (T. Wang, Wu, et al. 2009). The resulting

Page 43: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

27

hydrogel (Dex-PCL-HEMA/PNIPAAm) transitions from solution to gel within 30s at 37°C and

is reversible within the same time frame. To test the reparative effect of this hydrogel to

damaged cardiac function, 200µL was injected four days after MI in a rabbit model. One month

after the treatment, histological staining of heart sections showed a significant increase in scar

thickness and reduction in infarct size, compared with the control group (T. Wang, Wu, et al.

2009). Moreover, LV end systolic pressure was increased and end diastolic pressure reduced in

comparison with the MI control group. It was also observed that hydrogel implantation partially

reduced the dilatation of the LV, as shown by a significant increase in ejection fraction and a

significant decrease in LV end diastolic and systolic diameters (T. Wang, Wu, et al. 2009). These

results suggest that this hydrogel could serve as an injectable biomaterial that prevents LV

remodeling and dilation for the treatment of MI.

Similarly, Wu et al developed a temperature-sensitive, aliphatic polyester hydrogel (HG)

conjugated with VEGF and looked at its effects on cardiac recovery after MI (Wu et al. 2011). It

was shown that the hydrogel gelled in ~10 minutes at physiological conditions, and that in vitro

it was stable for up to 5 weeks, but completely degraded by 6 weeks when injected

subcutaneously. Injection around the infarct site was done seven days after coronary artery

ligation and rats were monitored up to five weeks post-injection (six weeks post-MI). Compared

with outcomes in the PBS group, ventricular volumes, preload recruitable stroke work, and end-

systolic elastance were all significantly preserved with HG-VEGF, and furthermore the VEGF

conjugated hydrogel led to improved blood vessel density in the infarct area (Wu et al. 2011).

Functionally, ejection fraction was ~60% higher, fractional shortening ~37% better with the HG-

VEGF group over the PBS injected group, and while the infarct thinned and dilated after PBS

injection it was ~30% smaller and 50% thicker in hearts treated with HG-VEGF (all measured at

5 weeks post injection) (Wu et al. 2011). They did note that most of these effects could be seen

with injection of hydrogel (no VEGF), or hydrogel with VEGF mixed in (not conjugated).

However, none of the effects were as significant as those observed with their VEGF conjugated

polyester hydrogel leading to the conclusion that conjugation provided sustained, localized

VEGF function promoting regeneration (Wu et al. 2011).

Furthermore, an important relationship on how hydrogel mechanical properties correlate their

therapeutic outcomes has been brought up and studied by Ifkovits et al (Ifkovits et al. 2010).

They compared two injectable methacrylated hyaluronic acid (MeHA) formulations that exhibit

Page 44: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

28

similar degradation and tissue distribution upon injection but have differential moduli, using

infarct only as a control group. The MeHA or control hydrogels were injected 30 min after the

infarction and the result showed both treatments significantly increased the wall thickness in the

apex (>200% increase) and basal (>40% increase) infarct regions compared with the control

infarct (Ifkovits et al. 2010). Specifically, the treatment with the higher modulus (MeHA high)

group had significantly smaller infarct area (approximately 16% smaller) compared with the

control infarct group. Moreover, the normalized end-diastolic and end-systolic volumes were

reduced (though not significantly) for the MeHA High group (1.7 and 1.9, respectively)

compared to control (2.1 and 2.5, respectively) (Ifkovits et al. 2010). Functionally, real time 3D

echocardiography was used to assess the LV dimension and cardiac function for each animal

before and immediately after infarction, suggesting that MeHA group also tended to have a

better cardiac output and higher ejection fraction (again however, not significant) than the low-

modulus (MeHA Low) and control infarct groups (Ifkovits et al. 2010).

Some care must be taken in interpreting the results of bulking material strategies as recent

work suggest passive wall support on its own does not prevent LV remodeling and preservation

of cardiac function. Rane et al looked to decouple biomaterial effects from mechanical effects of

such treatments by injecting bio-inert, non-degradable PEG hydrogel into a rat MI model nine

days after infarction (Rane et al. 2011). While infarct wall thickness was significantly increased

in the injected group verses saline injected control animals, there was no difference in cardiac

function (ejection fraction, end diastolic and systolic volumes) between groups. Confirmation of

the decoupling between material bioactivity and mechanical effects was done by comparing

cellular response between groups and showing no difference, thus leading to the conclusion that

benefits reported from other studies are likely due to differences in inflammatory and cellular

response to the material and not due to the bulking effect of the material itself (Rane et al. 2011).

For an injected hydrogel to act as a long term mechanical support, it would be assumed its

presence long-term (slow or no degradation) is required. To investigate this, a non-degradable, in

situ polymerizable PEG hydrogel was assessed for its effect on short and long term cardiac repair

after MI. Chosen for its degradation properties (i.e. lack thereof) as well as its very high

mechanical stiffness and inertness, gel components were injected into the infarcted region and

polymerized in situ following permanent ligation of the left anterior descending artery in male

Wistar rats (Dobner et al. 2009). Benefits were seen after 4 weeks with increased wall thickness

Page 45: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

29

(100%) and modestly attenuated dilation (43%) compared to a saline treated control group.

However, morphological improvements were only seen during the first 4 weeks as by 3 months

the hydrogel injection group had regressed to a cardiac geometry and function similar to the

control group (Dobner et al. 2009). The results suggest that while maintaining ventricle geometry

using stiff biomaterials is beneficial in the short term, successful long term application will most

probably require substantial further research toward developing a material with the necessary

characteristics and a greater understanding of the degree of stress relief needed to halt

pathological remodeling (Dobner et al. 2009).

As is evident, complex synthetic polymer hydrogels are generally the choice for ventricle

geometry preservation strategies due the high tunability of mechanical properties to match the

needs of the failing LV. This does not exclude groups from using natural polymers as well,

however. Dai et al investigated whether commercially available collagen injection could thicken

the infarcted LV wall and improve function by preventing systolic bulging in Fischer rats (Dai et

al. 2005). 100μL of collagen or saline was injected into the scar area of rats with week old MIs,

and then cardiac morphology and function was monitored a further six weeks. The results

showed significantly increased scar thickness in the collagen injected group (719±26μm)

compared with the saline-treated group (440±34μm), and also that injected collagen remained

present in the scar at six weeks. Furthermore, stoke volume was significantly larger in the

collagen-treated group (163±8μL) than in the saline-treated group (129±6μL), and LV ejection

fraction was greater in the collagen-treated group (48.4±1.8%) than in the saline-treated group

(40.7±1.0%) (Dai et al. 2005).

2.5.5. Clinical application The true implication of any of these injectable biomaterial strategies can only really be

assessed in a clinical setting. If the hydrogels are inert like alginate, they could be regulated as

devices, leading to a shorter time to reach the clinic. Two groups have developed alginate based

hydrogels currently in Phase II clinical trials (NCT01226563, & NCT01311791). The calcium-

cross-linked alginate hydrogel developed by Leor et al. was first tested in a swine anterior MI

model (Leor et al. 2009). Animals were monitored up to 60 days (56 days post-injection) and

control animals (intracoronary saline injection) showed an increase in LV diastolic area by 44%,

LV systolic area by 45%, and LV mass by 35%, whereas intracoronary hydrogel injection (2mL)

prevented and even significantly reversed LV enlargement by greater than 100% compared to

Page 46: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

30

control for all measures. Furthermore scar thickness was 53% greater with 2mL hydrogel

injection compared to the control, and further proved that their developed biomaterial was

feasible, safe, and effective (Leor et al. 2009). Dubbed IK-5001, the hydrogel has been approved

for Phase II clinical trials and they are currently recruiting patients to test the safety and

effectiveness of the device. Intracoronary injection of 4mL of IK-5001 into the blocked artery

(after successful stent placement) will be done with the goal of prevention of ventricular

remodeling and congestive heart failure when administered after a recent acute MI

(NCT01226563).

By contrast, the hydrogel developed by Lee et al is designed for direct injection into the

infarcted wall for restoration of LV geometry. Called Algisyl-LVR™, it is another calcium-

cross-linked alginate biomaterial that gels in 3-4 minutes and achieves a material strength of 3-

5kPa (Lee et al. 2012). An initial pilot study in 6 patients suffering from dilated cardiomyopathy

demonstrated sustained improvements in LV size and function that were accompanied by

statistically significant improvements in clinical status and quality of life 3 months post-

injection, with no implant related complications (Lee et al. 2012). The approved Phase II clinical

trial will evaluate the concept that direct mid LV intramyocardial injections of the biomaterial

into the free wall of the failing LV of patients with dilated cardiomyopathy will reduce LV size,

restore LV shape, lower LV wall stress and improve global LV function (NCT01311791). The

goal is to show significantly improved peak maximum oxygen uptake 6 months after treatment

between Algisyl-LVR™ treated patients versus those receiving medical management.

2.6. Chitosan & collagen

Chitosan is a naturally occurring linear polysaccharide, composed of N-acetyl-D-glucosamine

and D-glucosamine monomers linked by β(1-4) glycosidic bonds, as seen in Figure 2-2. It is

derived from the deacetylation of chitin, the most abundant organic molecule and main

component of the exoskeleton of crustaceans, molluscs, and insects. It is biocompatible, and its

degradation products are nontoxic and non-immunogenic (Prasitsilp et al. 2000; Muzzarelli

1993). Its biodegradation products are lower molecular weight chitosans, chito-oligomers, and

the monomers (N-acetyl-D-glucosamine, itself an anti-inflammatory drug, and D-glucosamine).

Additionally, the degradation of chitosan through enzymatic hydrolysis by lysozyme can be

controlled by modifying the degree of deacetylation (I. Y. Kim et al. 2008).

Page 47: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

31

Figure 2-2: The structures of chitosan and chitin, and their proportions in 85% deacetylated chitosan

The presence of amino groups on the polysaccharide backbone of chitosan allows for

conjugation with various bioactive molecules (Figure 2-2) (Shi et al. 2006; I. Y. Kim et al.

2008). Furthermore, the amino groups make chitosan poly-cationic, meaning it can adhere more

easily to negatively-charged tissue (Chenite et al. 2000), and can be used for the controlled

release of negatively-charged molecules (Fujita et al. 2004). However, chitosan suffers from

mechanical weakness and instability, as well as an inability to maintain predefined shapes (Puppi

et al. 2010). Chitosan is not a novel material but thanks to the diversity of potential uses it has

been well studied and utilized in many biomedical applications including wound repair, tissue

engineering, and drug delivery (as discussed above and reviewed extensively in Q Li et al. 1992;

Majeti and Kumar 2000; IY Kim et al. 2008).

Collagen I is a poly-anionic natural protein polymer and is a major constituent of the cardiac

extracellular matrix. Currently, collagen based materials are one of the most common ECM

materials used for culturing cells in vitro, and form the backbone of many hydrogels used in vivo

(as described earlier) as it is the most characterized and abundant component of the ECM

(Johnson, Lin, and Christman 2011). Native fibrous collagen can be processed into an injectable

solubilized form, which can self-assemble under specified conditions into a hydrogel with

nanoscale fibrous structure (Johnson, Lin, and Christman 2011). Collagen molecules assemble to

form microfibrils, which, in turn, form fibrils to create collagen fibers. Collagen can be

reconstituted to form hydrated gels that are similar to loose connective tissue in vivo, with cell

behavior in three dimensional hydrated collagen gels being more typical of in vivo behavior than

those grown on two-dimensional plastic culture surfaces. The material properties—material and

gelation kinetics, stiffness, nanoscale fibrous architecture—and cell-matrix interactions of

Page 48: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

32

collagen gels have been well characterized and can be manipulated by changes in temperature,

ion concentration, phosphate content, and pH to specify the properties of the hydrogel. It is

desirable as a biomaterial since it is biodegradable, has attachment sites for many cell types, and

has a mild inflammatory response when implanted (Pulapura and Kohn 1992).

Limitations in its use for cardiac tissue engineering, however, are its mechanical properties

and suitability for culturing cardiomyocytes, as well as its rapid biodegradation (Puppi et al.

2010). Numerous methods for improving the mechanical properties of collagen gels have been

explored including addition of chemical cross-linkers, but the effect of the by-products of these

chemical reactions can face scrutiny from regulatory and clinical bodies (Nicodemus and Bryant

2008). In contrast, in the natural ECM the structural properties of collagen are known to be

modified by glucosaminoglycans. The role they (glucosaminoglycans) play in modifying

collagen fibril formation is by intertwining the microfibrils, forming thicker fibrils, and thus

improving structural properties and increasing pore size. The structural characteristics of

chitosan make it very similar to glucosaminoglycans, thus it was proposed and shown that

addition of poly-cationic chitosan improves the mechanical stability and compressive strength of

the highly poly-anionic collagen based gels through ionic interactions between the two

components (Tan, Krishnaraj, and Desai 2001). Furthermore, the addition of chitosan to collagen

gels has been shown to reduce the rate of degradation by collagenase (H. Chen et al. 2008). The

interactions between collagen and chitosan in polyelectrolyte complexed hydrogels is illustrated

in Figure A-1 (Appendix A).

Collagen-chitosan hydrogel mixes have shown positive results by other groups as well. The

work of Lu et al suggested a thermo-responsive chitosan based hydrogel increased

vasculogenesis in rat infarcts when co-injected with ESCs in comparison to phosphate buffered

saline (PBS) control (Lu et al. 2009). Based on this study, chitosan was added to an already

established collagen hydrogel from another group, forming a chemically cross-linked collagen-

chitosan hydrogel aimed at improving recruitment, differentiation and survival of vascular

progenitors (Suuronen et al. 2006; Deng et al. 2010). While Lu et al were focused primarily on

CM regeneration (they were successful in improving ESC survival and differentiation), the work

by Deng et al showed that addition of chitosan stabilized the collagen hydrogel, was better suited

for the maturation of endothelial cells (in vitro), and promoted greater vascular growth and

recruited more endothelial/angiogenic cells than collagen alone (Lu et al. 2009; Deng et al.

Page 49: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

33

2010). Addition of the oligosaccharide sialyl LewisX (sLeX, which binds to the adhesion

molecule L-selectin) to the hydrogel improved mobilization and recruitment of endogenous and

transplanted circulating progenitor cells. Furthermore it enhanced neovascularization, and

improved tissue perfusion in a rat hind-limb ischemia model (Suuronen et al. 2009).

2.7. QHREDGS peptide

The angiopoietins are a family of growth factors that are known to bind to the receptor

tyrosine kinase Tie2 acting as both agonists and context-dependent antagonists (S. Davis et al.

1996; Maisonpierre 1997). In endothelial cells, angiopoietin 1 (Ang1) was found to act as a

survival factor by activating the phosphoinositide-3-kinase (PI3K)/Akt anti-apoptotic pathway

(Hayes et al. 1999; I. Kim et al. 2000; Papapetropoulos 2000). It was later found that fibroblasts

lacking the Tie2 receptor could still bind to Ang1 and that the resultant adhesion could be

inhibited by blocking integrin receptors (Carlson et al. 2001). This led to the discovery that the

αV integrin subunit, and more specifically the integrin receptor αVβ3, was responsible for

angiopoietin-mediated attachment (independent of Tie2), which induced endothelial cell

adhesion, migration, and activated characteristic integrin signal transduction pathways

(Camenisch et al. 2002). It has also been reported that while neonatal rat cardiomyocytes (NCM)

do not express the Tie2 receptor, they could still adhere to Ang1; adhesion to Ang1 conferred a

significant survival advantage to NCMs; Akt and mitogen-activated protein kinase (MAPK)

pathways were involved in the Ang1-mediated survival of CMs; and finally adhesion to Ang1

was integrin-dependent (Dallabrida et al. 2005). Further study identified the short sequence

QHREDGS, conserved among mouse, rat, human, and other species, as the integrin-binding

motif in the fibrinogen-like domain of Ang1, and it was demonstrated that the peptide

QHREDGS was capable of supporting CM attachment and survival similar to full-length Ang1

(Figure 2-3) (Rask, Mihic, et al. 2010; Rask, Dallabrida, et al. 2010). While chitosan on its own

does not promote cell attachment previous work has shown the ability of the peptide QHREDGS

to promote CM attachment and growth, as well as survival and maturation when covalently

attached to a photo-crosslinkable Az-chitosan hydrogel (Rask 2009; Rask, Mihic, et al. 2010;

Rask, Dallabrida, et al. 2010). Although a well known integrin ligand, RGD, also promotes

attachment of CMs, it prevents caspase-3 activation, it decreases the force of contraction of

papillary muscle, and leads to pathological hypertrophy of CMs (Boateng et al. 2005;

Page 50: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

34

Balasubramanian and Kuppuswamy 2003). The work previously described with Az-chitosan also

showed the superiority of QHREDGS to an RGD based peptide, specifically RGDS.

Figure 2-3: Molecular structure of Ang-1 derived peptide QHREDGS

2.8. Peptide modified chitosan-collagen hydrogel for cardiac tissue engineering

As it is known chitosan can interact with collagen to improve the mechanical stiffness of

collagen based gels it is proposed that a mixture of the two components can form a hydrogel

suitable for the culture of CMs in vitro as well as be mechanically stable enough for injection

into the heart for treatment of acute MI. The QHREDGS peptide sequence has been

demonstrated to promote the attachment and survival of CMs, suggesting it might have the

therapeutic potential to help restore cardiac contractile function post MI. Moreover, since the

beneficial effects of the QHREDGS peptide were found to be attachment dependent, we

postulated that immobilizing the peptide onto a hydrogel would promote its pro-survival activity

in addition to ensuring its sustained localization at the site of injection. Importantly, as a peptide,

QHREDGS has an advantage over the protein-based therapies in that it is water-soluble, very

stable, fully-synthetic with a precisely defined composition, it can be produced in a cost-effective

and facile manner, and does not require a specific orientation to be functional.

As discussed earlier, the presence of amino groups on the backbone of chitosan allows for its

conjugation with the carboxyls of bioactive molecules. Conjugation can be achieved through

many routes; however one of the most common methods for stable cross-linking of molecules is

Page 51: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

35

the use of carbodiimide compounds as they provide the most popular and versatile method for

labeling or cross-linking to carboxylic acids. The most readily available and commonly used

water soluble carbodiimide is 1-ethyl-3-(-3-dimethylaminopropyl) carbodiimide hydrochloride

(EDC) (Hayworth 2014). EDC reacts with carboxylic acid groups to form an active, unstable O-

acylisourea intermediate that is easily displaced by nucleophilic attack from primary amino

groups in the reaction mixture. The primary amine forms an amide bond with the original

carboxyl group, and a soluble urea derivative is released as the EDC by-product. Failure of the

intermediate to quickly react with an amine results in hydrolysis of the intermediate, regeneration

of the carboxyls, and the release of an N-unsubstituted urea (Hayworth 2014). The efficiency of

the reaction can be greatly improved with the addition of sulfo-N-hydroxysuccinimide (Sulfo-

NHS). Sulfo-NHS replaces EDC during the reaction to form a Sulfo-NHS ester that is

considerably more stable than the O-acylisourea intermediate, yet is still highly reactive with

primary amines at physiologic pH. The Sulfo-NHS is regenerated upon successful conjugation of

the carboxyl and amine, and it along with other reaction byproducts can subsequently be

removed and the products purified through methods such as dialysis or chromatography. The

modified chitosan-QHREDGS product is illustrated below (Figure 2-4), with further

modifications used seen in Figure A-2 (Appendix A). A full schematic of the reaction and

involved components can be seen in Figure A-3 (Appendix A).

Figure 2-4: QHREDGS peptide modified chitosan (QHG213H)

Page 52: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

36

3. Hydrogel development, in vitro characterization & preliminary in vivo

models

The original work described in this chapter was published in (Reis et al. 2012), and is used

with permission from Elsevier.

3.1. Materials & Methods

All animal experimental procedures were approved by the Animal Care Committee of the

Toronto General Research Institute and the University of Toronto Committee on Animal Care,

according to the Guide for the Care and Use of Laboratory Animals.

3.1.1. Peptide modified chitosan (UP-G113-QHREDGS)

3.1.1.1. Conjugation

QHREDGS peptide was conjugated to chitosan using EDC chemistry in a manner similar to

that previously described for conjugation to Az-chitosan (Rask, Dallabrida, et al. 2010). To

assess the dose response of the peptide two levels were used, a Low and a High concentration of

the peptide in proportion to the amount of chitosan present. Briefly, chitosan (UP-G113,

Novamatrix) was dissolved at 20 mg/mL in 0.9% normal saline and peptide at 10 mg/mL in

phosphate buffered saline (PBS, Lonza). These were then mixed with EDC and N-

hydroxysulfosuccinimide (S-NHS) dissolved in PBS to obtain final reaction solutions of 5

mg/mL chitosan and 0.5 mg/mL (Low) or 3 mg/mL (High) peptide with the ratio of

[EDC]/[peptide] and [S-NHS]/[EDC] kept constant in the two reaction mixtures at 0.8 and 2.75,

respectively.

The reaction solution (1.5 mL) was then left on a vortex mixer (VWR) at 650 RPM for 3

hours. The solutions were diluted 4X with PBS and dialysed (using a dialysis membrane,

Spectra/POR MWCO 3500, Spectrum Labs) against two 2 L changes of distilled water for ~24

hours.

Sterilization of the dialyzed reaction mixture was done by passing the solution through a 0.2

µm syringe filter (Progene) and then the material was recovered through lyophilisation for

approximately 48 hours. The recovered material was stored at -20°C until use.

3.1.1.2. Assessing conjugation efficiency

Page 53: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

37

Fluorescently labelled peptide, Fitc-Ahx-QHREDGS was used to assess the conjugation

efficiency and true final concentration of peptide attached to chitosan post dialysis. Fitc-labelled

QHREDGS (Biomatik) was substituted for regular peptide in the protocol above (3.1.1.1) and all

steps were protected from light. As the molecular weight cut-off of the dialysis membrane is at

most a tenth that of chitosan it can be safely assumed that all of the chitosan is retained and

recovered, and the peptide present is only that which was successfully attached to the chitosan.

Standards of the FITC-Ahx-QHREDGS in PBS was made ranging from 0.0005 to 0.01

mg/mL. For both the standards and the reaction solutions recovered post dialysis, the pH was

adjusted to 7, as fluorescence is greatly affected by pH (Graber et al. 1986). The samples (Low &

High) were diluted a further 1:10 and 1:100 and then run, with the standards, through a

fluorometer (Spectra Max Geminin EM, Molecular Devices) at an excitation wavelength of 490

nm and emission of 520 nm, with all samples run in triplicate. The true final concentrations of

peptide and conjugation efficiency were calculated by comparing the fluorescence of the samples

to the standards, correcting for the dilution factor and for the volume recovered post dialysis.

3.1.2. Chitosan-collagen hydrogel

3.1.2.1. Hydrogel formulation

Chitosan-collagen hydrogels were made through modification of the standard protocol for

collagen gelation from BD Biosciences. Briefly, pure or peptide-modified chitosan was dissolved

in sterile 0.9% normal saline and mixed with 10X PBS, collagen (BD Biosciences), and 1N

NaOH on ice, in that order. The final solution consisted of 2.5 mg/mL chitosan, 2.5 mg/mL

collagen, 10% of the final (solution) volume of 10X PBS (V10XPBS=0.1*VTotal), and 2.3% the

volume of collagen added of 1N NaOH (VNaOH=0.023*VCollagen). The final hydrogel solution was

mixed thoroughly and kept on ice until needed. An appropriate amount of hydrogel solution was

pipetted onto a tissue culture plate bottom, or mixed with cells first, and allowed to gel for 30

minutes in a humidified 5% CO2 37°C incubator. To make 0:1 or 3:1 mass:mass (m:m)

chitosan:collagen hydrogels the final solution was the same as above, but with 0 mg/mL or 7.5

mg/mL chitosan, respectively.

3.1.2.2. Scanning electron microscopy

Samples were imaged using environmental scanning electron microscopy (Hitachi S-3400 N).

Hydrogel samples (N=2/group) were allowed to gel for 30 min and individually placed into the

Page 54: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

38

specimen chamber. A filter paper was used to gently remove the excess water from the sample.

The chamber was closed and the temperature of the chamber was decreased to -20°C. The

samples were imaged under variable pressure mode at 70 Pa and 15 kV.

3.1.2.3. Hydrogel degradation

To assess the in vitro degradation of the hydrogel 2.0 mL Eppendorf tubes were weighed after

having 5 holes punched in the caps with an 18G1/2 needle. 500 μL of 2.5 mg/mL 1:1 m:m

chitosan-collagen hydrogel was then added to each tube and allowed to gel as previously

described. Culture media (1 mL) was added to each tube and incubated for different amounts of

time, with media changes (1 mL removed and replaced) every 48 hours. Gels were left for 1, 72,

120, or 240 hours (N=3/time point) when 1 mL of media was removed and the tubes and gel

frozen at -80°C. All groups were lyophilized together for 72 hours, and then weighed again.

Degradation was assessed by comparison of the lyophilized material weight to that of the 1 hour

group.

3.1.2.4. Rheological assessment of hydrogels

To assess differences in the mechanical stability of hydrogels with varying ratios of chitosan

present, as well as the effect of conjugating QHREDGS peptide to chitosan, a rheological

assessment measuring the loss and storage moduli of varying hydrogel compositions was

performed. Rheology was performed using a TA Instruments AR1000 rheometer with a 6 cm

acrylic cone and plate geometry and calibrated as per the manufacturer’s instructions prior to

seeding approximately 1 mL of appropriate hydrogel (0:1, 1:1, 3:1, or 1:1 with conjugated

QHREDGS peptide). Temperature was controlled using an integrated Pelletier plate and

equilibrated to 4°C and prior to hydrogel seeding. To determine the Linear Visco-elastic Range

(LVR), and show gel transition, a strain sweep was done at 4°C from 0-5% strain and frequency

of 1Hz, and after allowing the hydrogel to equilibrate at 37°C for 30 minutes. Second, from the

data collected from the strain sweeps a frequency sweep was performed on samples from all

formulations at 37°C. The frequency sweeps were performed by seeding hydrogel onto the

rheometer at 4°C, raising the temperature of the plate to 37°C and gelling for 30 minutes. A total

of N=3 samples per group per analysis (strain or frequency sweep) were performed.

3.1.3. In vitro cell culture

3.1.3.1. CM isolation

Page 55: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

39

Cardiomyocytes used for in vitro work were isolated from neonatal (1-2 day old) Sprague-

Dawley rat hearts as previously described (Radisic et al. 2003; Naito et al. 2006; Rask,

Dallabrida, et al. 2010). Briefly, rats were euthanized; hearts were removed, quartered and then

digested overnight with trypsin followed by five rounds of collagenase digests. Cells were then

pre-plated for 1hr to enrich for CMs, and pre-plate supernatant was collected. CM number in the

pre-plate supernatant was determined using a haemocytometer, and then cells were used for

experiments. Quantification of the cell population before and after pre-plating to enhance the CM

fraction has been performed previously, and it is reported after pre-plating the cell fractions are

63±2% CM, 33±3% cardiac fibroblasts, 3-4% smooth muscle cells, and 2-3% endothelial cells

(Naito et al. 2006). A second pre-plate can be performed to improve the CM fraction to ~80% if

desired (Iyer, Chui, and Radisic 2009).

3.1.3.2. CM media

The cardiomyocyte culture media was comprised of 10% (v/v) FBS, 1% Hepes, 100 U/mL

penicillin-streptomycin, 0.02 U/mL Insulin, 5 μg/mL vitamin C, and the remainder Dulbeco’s

Modified Eagle medium (10% F.I.V. media). Cells were always re-suspended prior to

encapsulation in a serum rich media, specifically 30% F.I.V (30% FBS).

3.1.3.3. CM encapsulation & culture

Unless otherwise specified each sample prepared for in vitro experiments consisted of 5x105

CMs re-suspended in 2.5 μL 30% F.I.V. media and encapsulated in 12.5 μL of appropriate gel

prepared as described in 3.1.2.1. The gel, media, and CM mix was pipetted onto the surface of a

24-well tissue culture plate and put in an incubator for 30 minutes to allow for gelation, after

which samples were supplemented with 500 μL warm 10% F.I.V. media and cultured for 120

hours (five days). Media was aspirated, or collected, and 100% changed every 48hrs (two days).

3.1.4. In vivo studies

3.1.4.1. Subcutaneous injection

CMs from neonatal Lewis rat pups were collected in the same manner as described in 3.1.3.1,

spun down and 2 million CMs were re-suspended in 10μL 30% F.I.V. media first, followed by

the addition of 100μL appropriate gel (Control (No Peptide), Low, and High) in Eppendorf tubes.

The cell/gel suspension was then randomly injected subcutaneously (100 μL) with a 23G1/2

needle into the back of adult female Lewis rats (four injections per rat). The area where samples

Page 56: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

40

were injected had been shaved and rough injection sites were marked with permanent marker in

order to locate the injected material later. Samples (N=6/group) were left for seven days and then

the animals were sacrificed, the skin surrounding the area where injections were made was

removed and the material (hydrogel with cells) recovered and fixed with 10% formalin at 4°C for

approximately 12hrs. Recovered samples were then transferred to PBS and sent to the Pathology

Research Program (PRP) histology lab at University Health Network for paraffin-embedding and

sectioning.

3.1.4.2. Mouse MI model

To assess the potential of the developed hydrogel to be injected into the infracted heart, a

mouse MI model was used. Adult male C57 Black-6 mice were subjected to a left ventricular

anterior descending coronary artery ligation procedure (LAD procedure) to mimic severe MI

with the help of Dr. A. Momen from the University Health Network. The mouse MI model has

become important for studying myocardial regeneration following interventions such as cell

based therapies, and studies have shown the LAD procedure gives rise to MIs of reproducible

size and severity as well as an immediate surgical survival of 60%, and 2 week survival of ~83%

(or 50% overall survival) (Kumar et al. 2005). Animals were divided into three groups: Sham,

MI Only, and MI+Control gel. Briefly, animals were initially anaesthetized with 5% isoflurane

and maintained with 2-2.5% isoflurane during surgery. Animals were intubated and ventilated

using a Minivent ventilator (Harvard Apparatus, March-Hugstetten, Germany) at ~200

breaths/min. A left thoracotomy was performed, and the left coronary artery was ligated using a

7-0 silk suture (Ethicon) passed with a reverse cutting needle. Sham mice did not receive the

LAD ligation and the chest was closed immediately. MI+Control gel mice had 50µL of

2.5mg/mL 1:1 m:m chitosan:collagen Control (No Peptide) gel injected with a 27G1/2 needle

into the area immediately below the ligation suture. The hydrogel was warmed to ~37°C for

~10min prior to injection to allow gelling to initiate. The animals were removed from the

ventilator and allowed to recover below a heat lamp for approximately 30 minutes. (N=3 animals

for MI, N=3 for MI+Control Gel, N=1 for Sham).

3.1.5. Construct Characterization

3.1.5.1. Gel compaction

Page 57: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

41

To assess the chitosan-collagen ratio that was most appropriate for formation of beating

cardiac constructs, CMs were encapsulated in the same manner as described in 3.1.3.3, with

groups consisting of CMs in 0.5:1, 1:1, or 3:1 m:m chitosan-collagen hydrogel, with the

concentration of collagen kept at 2.5 mg/mL. The cell/hydrogel mix was pipetted onto 12mm

circular glass cover-slips located in the wells of a 12-well tissue culture plate. This was done to

obtain uniform initial construct sizes and prevent cell/gel attachment to the bottom of the plates,

which would confound results. After 144 hours (6 days) of incubation (with media changes every

48 hours) the media was removed and pictures of the hydrogel constructs taken (N=4/group).

Adobe Photoshop CS3 was then used to measure the area of each hydrogel sample, and mean

values compared between groups.

3.1.5.2. Live/Dead staining

Live/dead staining was performed according to the manufacturer’s protocol (Invitrogen) using

5-carboxyfluorescein diacetate acetoxymethyl ester (CFDA, green for live cells) and propidium

iodide (PI, red for dead cells) on N=2 constructs/group. CFDA is a non-fluorescent compound

that can penetrate intact cell membranes wherein it is cleaved by non-specific esterases to release

polar fluorescent fluorescein which is retained inside viable cells, whereas PI is a nucleic acid

probe that is unable to cross intact cell membranes but penetrates the membranes of dead cells.

Quantification was performed using ImageJ as previously described (Iyer, Chiu, and Radisic

2009). To show live cell distribution confocal images (z-stacks) were taken at the edge and

center of cell/gel constructs.

3.1.5.3. Functional testing

Two measurements were taken to assess the electrical function of encapsulated

cardiomyocytes in chitosan-collagen hydrogels (No peptide, Low, and High peptide) cultured for

five days as described in 3.1.3.3. Excitation threshold (ET) is the minimum voltage required to

pace most of the cells in a construct and the maximum capture rate (MCR) is the maximum

frequency at which the encapsulated CMs could be induced to beat simultaneously (Radisic et al.

2004). N=11 constructs/group were cultured as described above and then ET/MCR were

measured using bi-phasic electrical pulses as previously reported (Rask, Mihic, et al. 2010).

Briefly, media was removed, a pair of parallel carbon electrodes were placed surrounding the

gels, and the system was immersed in warm Tyrode’s solution (Sigma, pH≈7.3). All

Page 58: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

42

measurements were done in an environmental chamber at 37°C and ET was measured by

increasing the voltage until ~90% of cells in the field of view were seen to be beating

synchronously with square biphasic pulses of 2 ms and frequency of 1 Hz. MCR was measured

by setting the voltage at 12 V increasing the frequency until most of the cells were no longer

beating synchronously with the driving signal.

Success rate was measured as the percentage of constructs for which ET/MCR measurements

were able to be taken (i.e. constructs with significant beating portions) to the total number of

constructs seeded for that group. Chi-square and Fisher’s Exact tests were performed to analyze

the association between groups.

3.1.5.4. XTT assay

CMs were cultured for five days as per 3.1.3.3 at which time media was removed and

replaced with fresh 10% F.I.V. media and XTT assay solution as per the manufacturer’s protocol

(XTT cell viability assay kit, Biotium, Inc.). Samples were incubated for a further three hours

with the mix and then three 100 μL samples of mix were taken from each hydrogel sample and

put in corresponding wells of a 96 well plate. The absorbance of wells was read using an

absorbance meter (Apollo LB911, Berthold Technologies) at a wavelength of 450 nm and a

reference of 620 nm. The triplicate readings per hydrogel were averaged to get a value for the

sample, and then all sample values were averaged to get a value for the group. A total of N=6 per

group were tested. In addition, N=2 per group of no-cell control gels were used. The mean

absorbance reading of the no-cell controls was subtracted from all test groups. A standard curve

was also made to show that XTT is indeed a good indicator of cell metabolism and indirectly cell

number by encapsulating varying numbers of CMs in the same volumes of serum rich media and

No-QHREDGS Control hydrogel and incubating for 1 or 120 hours. Relative absorbance is

reported, with mean absorbance of samples relative to that of constructs with no CMs. Higher

relative absorbance indicates a higher level of viability/metabolic activity.

3.1.5.5. LDH assay

The LDH cytotoxicity assay (Cayman Chemical Company) was used to assess cell death at

varying time points. It was performed as per the manufacturer’s protocol on media samples taken

every 24 hours from samples prepared as per 3.1.3.3. Briefly, 50 μL media samples (in triplicate)

were put into the wells of a 96 well plate and an equivalent volume of LDH reagent was added to

Page 59: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

43

each well. The plate was placed on an orbital shaker for 30 minutes at 600 rpm protected from

light and then the absorbance of each well read using an absorbance meter (Apollo LB911,

Berthold Technologies) at a wavelength of 492 nm and reference of 620 nm. A total of N=6

gels/group plus 2 gels/group of no-cell controls were assessed. The absorbance of the no-cell

controls was subtracted from the readings of CM samples to get a more accurate value. A cell

standard was made in the same manner as for the XTT assay, and the LDH assay performed on

media samples from the standard (N=4/group) as well as the LDH standard.

3.1.5.6. PicoGreen DNA Assay

Constructs cultured as per 3.1.3.3 for four days were subsequently immersed in 1 mL of lysis

buffer (0.2% Triton X-100, 200 mM Tris-HCl, 20 mM EDTA, pH 7.5) and transferred to 2 mL

Eppendorf tubes. Several autoclaved 1 mm diameter silica beads were added to each vial, and the

constructs were homogenized using a Mini-Beadbeater-16 (Biospec), via six cycles of ten

seconds each. 500 μL of each homogenate was then transferred to separate wells of a 24-well

plate, and the Picogreen assay was performed as directed by Invitrogen. Briefly, 500 μL of

Picogreen reagent was added to each well, and incubated in the dark for 3-5 minutes, after which

fluorescence was read using a fluorescence micro-plate reader (Spectra Max Gemini EM,

Molecular Devices; excitation 480 nm, emission 520 nm). A separate assay, which was done on

gels lysed immediately after encapsulation with a known quantity of cells, served as a control to

determine the cell number in gels at the end of the experiment.

3.1.5.7. Histological & immunofluorescent staining

Samples for immunohistochemical and immunofluorescent staining were fixed at 4°C with

10% neutral buffered formalin (Sigma, HT501129) for approximately 12 hrs, and then

transferred to PBS. For paraffin embedding and sectioning, as well as hematoxylin and eosin

(H&E), Mason’s trichrome, smooth muscle actin (SMA), Factor VIII (F8), and CD31 staining,

the fixed samples were sent to the Pathology Research Program (PRP) at the University Health

Network. Chitosan staining was performed using Cibacron Brilliant Red-3BA (CBR-3BA,

Sigma Aldrich) and Weigert’s Iron Hematoxylin as described (Rossomacha, Hoemanni, and

Shive 2004). Paraffin sections were also stained for cardiac troponin T (cTnT, mouse

monoclonal antibody, Thermo Scientific MS-295-P). Deparaffinised slides were blocked using

Normal Horse Serum (Gibco 16050-122) for 40 minutes and then primary antibody (cTnT) was

Page 60: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

44

applied at a dilution factor of 1:200 for at least 12 hrs at 4°C. Secondary antibody solution

(Alexafluor 488 donkey anti-mouse IgG (H+L), Invitrogen A21202) was applied at a dilution of

1:200 with DAPI (Sigma) nuclear stain at 1:200 dilution for ~40 min at room temperature. The

slides were mounted using Fluoromount aqueous mounting medium (Sigma-Aldrich, F4680),

covered with cover-slips (VWR, 22x50 mm) and imaged.

Sections stained for CD3 and vimentin were deparaffinised and rehydrated with subsequent

baths (3 of each) of 100% xylene, 100% EtOH, 95% EtOH, 75% EtOH, and distilled water for 3

minutes per change. Antigen retrieval was performed by microwaving slides immersed in TRIS-

EDTA buffer for 5 min, and blocked using Dako Serum Free Protein Block (Dako X0909) for 30

min at room temperature. Primary antibody (poly rabbit anti-human CD3 diluted 1:200, Dako

A0452 or mouse monoclonal anti-vimentin diluted 1:400, Sigma V6630) was applied to sections

for 2 hrs at room temperature followed by secondary antibodies (goat anti-rabbit FITC IgG

(H+L), Jackson ImmunoResearch 111-095-144 or Alexafluor 488 donkey anti-mouse IgG

(H+L), Invitrogen A21202) at 1:400 dilution for 1 hr at room temperature. DAPI (Sigma) nuclear

stain was applied at 1:1000 dilution for 10 minutes and finally slides were mounted using Dako

Fluorescence Mounting Medium (Dako, S3023) and imaged.

Quantification of area covered by positively stained cells in immunostained sections was

performed using Adobe Photoshop CS3 and ImageJ updated from a previously reported method

(Chiu and Radisic 2010). Up to five distinct images per section per sample were gathered. The

colour profiles were saved such that the same colour selection profile could be applied

automatically to every image to select for positive staining (removing subjective human staining

selection). Unselected image areas were deleted and the image was converted to black and white

and saved as a JPEG. Adjusted images were then opened in ImageJ, thresholded to remove

potential background, and then analyzed to count particles. The resultant output was Area

Fraction of black particles (positive stain) in relation to the whole image which gives a relative

measure of positive immunostaining. The algorithm was tested on images from every group to

ensure consistency, colour profile refined as necessary, and then applied to all images. The five

images per section were averaged to obtain a % Positive Area measure for the sample, and then

sample values averaged to obtain Mean±SD for each group.

The number of F8+ vessels and their diameters were determined using ImageJ according to a

previously published method (Chiu and Radisic 2010). Five randomly selected 300x300pixels

Page 61: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

45

(131x131mm2) areas were cropped and magnified from each original F8 image. The numbers of

F8+ vessels were manually counted from the cropped images and normalized to the area of the

image to determine the vessel density. The diameters of the vessels were evaluated by drawing a

line across the short axis of the vessel and then measuring the length of the line in ImageJ.

3.1.6. Statistical analysis Statistical analysis was performed using SPSS Statistics 17.0 and GraphPad Prism 5.0.

Differences between experimental groups were analyzed by using one-way ANOVA with post-

hoc Tukey tests or two-way ANOVA with Bonferroni post-tests. Categorical data was compared

using Chi-square or Fisher’s Exact tests. P<0.05 was considered significant for all statistical

tests. Results were plotted with GraphPad Prism 5.0, with all data being reported as mean ±

standard deviation (Mean±SD).

3.2. Results & Discussion

3.2.1. Hydrogel composition and characterization

3.2.1.1. Base collagen-chitosan hydrogel

The first obstacle in designing a hydrogel as a vehicle for cell transplantation to the heart is

demonstrating its suitability for cardiomyocyte culture as well as an appropriate gelation time to

retain transplanted cells/material in the site of injection. Using previous work conducted in

optimizing a chitosan-collagen hydrogel for a hematopoietic cell line, similar concentrations and

ratios of the two components were chosen as starting points (Tan, Krishnaraj, and Desai 2001).

Various initial concentrations of collagen varying from 1.5-3 mg/mL were qualitatively

assessed for structural stability once gelled as per the manufacturer’s protocol (BD Biosciences).

It was determined that concentrations below 2 mg/mL were not “gelled” enough, whereas high

concentrations of around 3 mg/mL provided a much more stable gel it severely limited the stock

concentration of chitosan required to get the necessary ratios (needed to be on the order of 100

mg/mL, which is unfeasible). As such, a collagen concentration of 2.5 mg/mL was chosen and

ratios of 0:1, 1:1, and 3:1 chitosan:collagen m:m gels were made to assess differences in gelling

time and remodelling when cardiomyocytes were encapsulated (see 3.1.2.1 & 3.1.3.3).

No apparent differences in gelling time between the three groups (0:1, 1:1, & 3:1) were

observed, all forming nice, stable gels in 30 min at 37°C, however the differences in remodelling

that took place during cell culture were marked, as seen in Figure 3-1 A. The initial size of all

Page 62: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

46

gels used for gel compaction studies was 9 mm. Assuming gels formed a uniform cylinder with a

diameter of 9 mm and volume of 12.5 μL then the theoretical average initial height of the

constructs was 197 μm. The 0:1 group compacted almost six times more than the 3:1 group after

6 days of culture (which remained significantly unchanged from its initial size, data not shown),

and almost four times more than the 1:1 gel group (P<0.001 between all groups). The 3:1 group

gels, however where very brittle and broke apart easily with any outside disturbance, while both

the 0:1 and 1:1 group maintained their form after handling.

Cardiomyocytes require a matrix soft enough that they can remodel, yet stiff enough to resist

compaction from the remodelling as well as contraction of the cells. Our previous studies

demonstrated that substrates of stiffness comparable to the native heart maintains the

differentiated phenotype of cardiomyocytes and prevents fibroblast overgrowth in monolayer

cultures of heart cells (Bhana et al. 2010). If a matrix is not stiff enough to maintain its shape and

fully compacts, as seen in the 0:1 group, then proper elongation and maturation of the CMs is not

possible. Consistently, beating constructs were observed in the 1:1 group, not at all in the 0:1

group, and only in small portions of the 3:1 group (Supplemental Movie 1&2, from Reis et al.

2012).

The elongation and ability of the construct to contract in the 1:1 group but not in 0:1 or 3:1

groups after 6 days of culture (both spontaneous and induced) point toward the health and

performance of the cells (images and videos not shown).

Confirmation of the results seen in the in vitro cell culture work performed was obtained

through rheological assessment of the varying hydrogel formulations (seen in Figure 3-1 C-E).

At 4 °C all hydrogels are in a liquid state, as seen in Figure 3-1 C with the loss modulus (G11) of

all groups being greater than that of the storage modulus (G1) for all groups throughout a strain

sweep from 0-100%. All formulations show gel-like behavior after 30 minutes at 37°C (Figure

3-1 D), with a good LVR up to strains of at least 5%. The differences in the magnitude of loss

and storage moduli can be seen between groups, with the 1:1 group showing the highest storage

modulus and 3:1 the lowest. This observation is even more apparent in the results from the

frequency sweeps performed on samples of all formulations with the same trend of 1:1 having

the highest storage modulus and the 3:1 group the lowest, and similarly seen with the loss

modulus although the differences are not as pronounced (Figure 3-1 E). It should be noted that in

many instances at 4°C moduli values could not be obtained at low strains due to the sensitivity of

Page 63: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

47

the rheometer, that is why there are clusters of readings some points. Likewise, due to the brittle

nature of the 3:1 gel the rheometer had difficulty measuring the loss and storage moduli at 37°C

for some samples. It should also be noted that time varying viscosity rheology measurements

were performed, however it was found that in initiating gelation through increasing temperature

from 4-37°C caused an immediate change over from liquid (G11> G1) to gel (G1>G11)which is

not a practical measure for our application. Furthermore, during the elastic storage modulus (G1)

never equilibrated during these experiments to give a meaningful measure of time to gelation.

Mechanical properties of collagen-chitosan hydrogels were studied previously (L. Wang and

Stegemann 2011; Tan, Krishnaraj, and Desai 2001). The initial increase in storage modulus and

subsequent decrease with further addition of chitosan is likely due to the marked changes in the

gel microstructure (Figure 3-1 D&E). The resultant structure of collagen only gels is a highly

interconnected fibrous network with fibers of fairly uniform size. Addition of chitosan does not

seem to affect fiber formation, however they interfere with the resultant network by sequestering

fibers together and thus forming thicker “struts”, yet retaining a high level of interconnectivity in

the structure (i.e. 1:1 gels). By bringing together many collagen fibers along a chitosan

backbone, formation of these thicker struts results in an increase in mechanical integrity.

Increasing the fraction of chitosan (i.e. 3:1 chitosan:collagen gels) causes a further increase in

collagen fiber sequestration (thus thicker struts), however the degree of connectivity between

resultant fibers is greatly diminished (see Appendix A, Figure A-1). Furthermore, Tan et al

(2001) found previously that in the case of a human hematopoietic cell line, changes in cell

growth were determined by chitosan:collagen ratio only, and that increasing or decreasing the

total protein concentration (while keeping the ratio constant) had no effect on cell proliferation

(Tan, Krishnaraj, and Desai 2001).

Protein based gels are commonly tested at strain and frequency values on the order of 1% and

1 Hz respectively (L. Wang and Stegemann 2011; Ikeda and Foegeding 2003; Whittingstall

2003). As these values fall in the LVR for all chitosan:collagen formulations tested here they

were chosen as a basis to compare the loss and storage moduli between gel formulations. The

values are summarized in Table 3-1, below, and as expected confirm that a 1:1 collagen:chitosan

gel formulation provides the most mechanically stable gel in terms of storage modulus, but also

shows the greatest difference (separation) between loss and storage moduli (P<0.0001).

Page 64: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

48

Based on these findings it was determined that a 2.5 mg/mL 1:1 m:m chitosan:collagen gel

formulation provided the most suitable environment of the three tested for cardiomyocyte

culture, and all further work done used this formulation.

Table 3-1: Hydrogel rheological assessment

Hydrogel Storage

Modulus±SD (Pa) Loss

Modulus±SD (Pa)

ANOVA Multiple Comparisons (Sig.)

0:1 1:1 3:1 1:1-P

0:1 (Collagen Only)

32.32±7.55 4.20±1.16 <0.001 <0.001 0.014

1:1 (Control (No Peptide))

54.87±9.05 6.05±0.90 0.006 <0.001 0.119

3:1 11.63±3.46 2.45±0.66 0.010 <0.001 <0.001

1:1-HP (High Peptide)

45.37±4.29 5.53±0.45 0.073 1.000 <0.001

Page 65: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

49

Figure 3-1: Hydrogel Characterization (A) Chitosan:collagen blends resist gel compaction by the heart cells significantly better than collagen alone

after 6 days in culture. (2.5 mg/mL collagen concentration, 1:1 or 3:1 mass ratio of chitosan:collagen). Images show cell/gel constructs after 6 days in culture. Gel sizes in the graph are normalized to the 0:1 group. N=4/group. (B) 2.5 mg/mL 1:1 chitosan-collagen m:m ratio shows no significant degradation over 10 days in vitro. N=3/group. (C-E) Rheological analysis of various ratios confirms 1:1 has mechanical properties suitable for

Page 66: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

50

CM culture. (C) All gel formulations were liquid at 4°C. (D) All gel formulations exhibit linear viscoelastic behaviour gel properties between 0.05 and 5% strain after 30 minutes at 37°C. (E) 1:1 chitosan-collagen mixture has the highest storage modulus (G’) at all frequencies (with constant strain at 1%) after gelling for 30 minutes at 37°C. (C-E) Conjugation of QHREDGS peptide to chitosan does not alter the rheological properties of the 1:1 gel formulation. N=3/group C-E. All data presented as Mean±SD; ***P<0.001.

A study of the degradation of the hydrogel over the time course of in vitro experiments was

done to assess how much the material degraded on its own (hydrolytic and in response to the

components of culture media) as described in 3.1.2.3. The results are seen in Figure 3-1 B and

show no significant difference in dry material weight at all time points up to 10 days of exposure

to culture medium. A control of just Eppendorf tubes with culture media and no hydrogel was

used to demonstrate there was no inherent changes in dry tube mass over the course of the

experiment, as they could have an effect on the very small material masses measured (only 6-8

mg). It should be noted that the 1 hr group shows a dry weight close to the expected 5 mg total

for 1 mL of hydrogel but this increases slightly, not significantly, over the course of the

experiment and may be attributed to absorption of media components by the hydrogel.

3.2.1.2. Collagen-chitosan-QHREDGS hydrogel

The amount of QHREDGS peptide successfully bound to chitosan and the efficiency of the

process was evaluated using fluorescently labelled peptide, Fitc-Ahx-QHREDGS, as described in

3.1.1.2. The efficiency of peptide conjugation is affected by the ratios of EDC and S-NHS to

peptide in the reaction mixture, and as this study has been performed on Az-chitosan the ratios

used for the protocol described in 3.1.1.1 were kept the same as those previously reported (Rask,

Mihic, et al. 2010; Rask, Dallabrida, et al. 2010). Furthermore, the minimum level of peptide

needed to elicit a cellular response was characterized in the same study, thus target amounts of

QHREDGS peptide to be conjugated were kept in the same range. The results of the current

study are seen in Figure 3-2 A. The differences in conjugation efficiency of both the Low and

High amounts of peptide were not statistically significant and average to be 55%, the same as

that found in Rask, Mihic, et al. (2010). It follows that there was statistically significant

(P=0.002) differences in the concentration of peptide successfully conjugated, with the Low

group being 0.053±0.016 mg/mL and the High 0.346±0.004 mg/mL in post dialysis solution (or

100±30 nmol/mL-gel and 651±8 nmol/mL-gel for the Low and High final gel formulations

respectively), as seen in Figure 3-2 A.

SEM images of 2.5 mg/mL 1:1 hydrogels with varying levels of QHREDGS peptide are seen

in Figure 3-2 B and were done to identify differences in hydrogel morphology with the addition

Page 67: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

51

of peptide. The images show that the gels have a porous structure, which is desirable for

encapsulation systems to allow for nutrient exchange and that the addition of peptide causes no

apparent differences in structure. This indicates, qualitatively at least, that addition of

QHREDGS to chitosan does not affect its structure, or ability to interact with collagen and form

a stable hydrogel.

Quantitative assessment of the effect peptide conjugation has on the hydrogel was done using

rheology as in 3.1.2.4. The results of strain sweeps at 4°C and 37°C and frequency sweeps at

37°C, seen in Figure 3-1 , illustrate transition from liquid to gel after 30 minutes at 37°C and no

significant difference in the loss or storage moduli of peptide conjugated hydrogel to Control (no

peptide) 1:1 collagen:chitosan hydrogel at constant 1% strain and frequency of 1Hz (see Table

3-1). The 1:1 chitosan:collagen blend with and without the peptide had a storage and loss

modulus similar to the blend of 25% cardiac ECM and collagen I used in previous studies to

increase maturation of cardiomyocytes derived from human embryonic stem cells (Duan et al.

2011).

Therefore it can be concluded that peptide conjugation does not affect the morphology nor

mechanical properties of the hydrogel and thus any differences in cell survival, maturation, and

performance within the gel should be due entirely to the varying amounts of QHREDGS peptide

present.

It should be noted that the current hydrogel preparation does not allow us to decouple

mechanical properties of the hydrogel from the composition. There are other more elegant

chemistries that allow for independent control of mechanical properties and concentration of

presented ligands (Deforest, Sims, and Anseth 2010). In addition, using identical EDC chemistry

and covalent immobilization of VEGF to collagen, we previously demonstrated that the

biomolecule was stably immobilized to the substrate with negligible biomolecule release in PBS

over 28 days (Chiu and Radisic 2010; Miyagi et al. 2011). Thus it can be expected that the

QHREDGS peptide will also be stably retained within the hydrogel during culture.

Page 68: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

52

Figure 3-2: Characterization of peptide conjugation and hydrogel morphology (A) Fluorescent analysis using Fitc-QHREDGS shows no difference in conjugation efficiency of peptide to

chitosan and significant difference in the amount of peptide between Low and High groups. (B) Representative SEM images show no visible differences in landscape between groups. Results are shown for 2.5mg/mL 1:1 chitosan:collagen gel (Control) and 1:1 chitosan-QHREDGS:collagen with Low and High amount of the peptide immobilized.

3.2.2. In vitro studies with cardiomyocytes in collagen-chitosan-QHREDGS hydrogels

3.2.2.1. Distribution, viability, and metabolism of CMs encapsulated in the hydrogels

For in vitro studies the same amount of cell/gel mixture as above (12.5l) was pipetted onto

the surface of 12 well TC plates and spread to form uniformly sized constructs of 6.2mm±0.3mm

in diameter (N=4/group, p=0.07 between groups). These gels had a theoretical height of

413μm±43μm. The encapsulated cells were distributed evenly in three dimensions in the cross-

section immediately after gelation and at 5 days (120 hrs) of culture in the hydrogels (Figure

Page 69: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

53

3-3). There was very minimal leakage of cells during gelation, and very little migration of cells

out of the gels after 5 days (Figure B-1, found in Appendix B). It was therefore assumed that

effects of cells outside of the gels could be ignored and all assays were performed in the wells.

Figure 3-3: Gel encapsulation results in uniform distribution of live cells Confocal cross-sectional images of CFDA stained (green) cell/gel constructs (A) 1 hr post seeding (30

minutes post gelation) and (B) 120 hrs post seeding. All scale bars=50 µm.

Page 70: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

54

The XTT assay is a measure of cell viability based on metabolic activity and was assessed for

cells encapsulated in the various gels (Control (no peptide), Low, and High peptide). The results

are seen in Figure 3-4 A&B. Figure 3-4 A shows the applicability of XTT in measuring cell

viability/activity when encapsulated in the hydrogel (Control gel), indicating a low amount of

metabolic activity 1 hr after encapsulation and increased, significantly different activities

between varying groups (numbers of cells encapsulated) after 5 days in culture. When comparing

the three test groups (Control, Low, & High) directly as seen in Figure 3-4 B cell metabolism is

significantly higher (P<0.01) in the High QHREDGS group in comparison to the Control, but not

the Low group. There is an overall trend however pointing toward increased metabolic activity

with increased peptide concentration in the gel. A question arises in interpreting these results as

directly pointing toward increased viability in the High group as the cell population used to seed

the hydrogels is not a pure (non-proliferative) CM population, but contains fibroblasts and

endothelial cells as well. While there is no doubt that metabolic activity is indeed higher in the

High group it is possible that this is due to an increased level of fibroblast and endothelial cell

proliferation, and not CM activity.

Cell death at varying time points during culture of encapsulated CMs was assessed using an

LDH assay which measures cytotoxicity based on the activity of lactate dehydrogenase (LDH)

released from damaged cells. The assay was performed, as described in 3.1.5.5, to determine if

there was any difference in the survival/apoptosis of cells in the three gel test groups, and also to

give an indication of whether the results of the XTT assay could be due to differences in cell

number. Figure 3-4 C shows that discernable differences in LDH activity could be measured

from varying numbers of cells encapsulated in the hydrogels, and therefore the LDH assay could

be used to compare the Control, Low and High gel groups. The results of directly comparing

groups (Figure 3-4 D) show a general trend toward lower cell death (LDH activity, Control to

Low to High) at all time points measured, and there is a statistically significant difference in cell

death between Control and High groups over the course of the experiment (two-way ANOVA,

P<0.05). It should be noted that data was collected up to 120 hrs (5 days) however there was

negligible cell death in all groups after 72 hrs. This is consistent with previous reports on the

cultivation of heart tissues in well plates showing the majority of cell death occurs early on in

culture, within the first 24 hr (Radisic et al. 2003). Collectively, these data show the effect of

Page 71: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

55

QHREDGS peptide in improving cell survival, and correlate with the results of the XTT assay as

lower cell death leads to higher cell metabolic activity and viability.

Finally, total cell number at the end of five days in culture was assessed using the Picogreen

DNA assay which uses an ultra-sensitive fluorescent nucleic acid stain to quantify double-

stranded DNA. Performed as described in 3.1.5.6, the strong correlation between initial cell

number encapsulated and fluorescence (total DNA) shows the hydrogel does not affect the assay

(Figure 3-4 E) and that the assay can be used to assess the differences in cell number between

test groups. The results of comparing the three groups (Figure 3-4 F) show no significant

differences in total cell number initially (data not shown) and after 120 hrs in culture. These data

confirm that the results seen in both the XTT and LDH assays were not due to differences in cell

number encapsulated (higher or lower respectively), that cell proliferation does not play a major

role, and thus point toward the improved health of CMs encapsulated in gels with QHREDGS

and specifically with the High concentration.

When the three assays, LDH, DNA and XTT are taken as a whole it is clear that there is no

appreciable proliferation of non-myocytes in this system. Higher XTT activity in the high

QHREDGS group indicates improved metabolic activity of all cells.

Page 72: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

56

Figure 3-4: Metabolic activity and total number of cells encapsulated in 1:1 chitosan:collagen hydrogels

(A) Cell metabolic activity is significantly lower at 1Hr post encapsulation compared to 120Hrs after. (N=4/group for 1hr, N=3/group for 120hrs). (B) Cell metabolism in the Control (No QHREDGS) gels is significantly lower than in the High (3mg/mL QHREDGS) hydrogel group but not in the Low (0.5mg/mL QHREDGS) hydrogel group based on XTT assay (N=6/group). Data is relative to control gels with no cells. (C) LDH assay shows correlation between final cell number and cell death at all time points (N=3/group for ½ hr group, N=4/group for the remaining time points). (D) LDH assay shows significantly lower cell death in the High (3 mg/mL QHREDGS) compared to Control (No QHREDGS), but not in the Low (0.5 mg/ml QHREDGS)

Page 73: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

57

hydrogel group (N=6/group/time point). (E) Picogreen DNA assay shows strong correlation between initial total cell number encapsulated and fluorescence (N=3/group). (F) Picogreen DNA assay shows no difference in total cell number between all groups after 120 hrs in culture (N=4 Low & High, N=3 Control; initial cell number=5·105 CMs). All data presented as Mean±SD, * P<0.05, ** P<0.01, *** P<0.001.

3.2.2.2. Construct functional & morphological properties

Native heart tissue responds and paces to electrical stimulation and this ability is usually

associated with healthy tissue. Therefore CMs encapsulated in the three hydrogel groups were

tested to determine their excitation threshold (ET) and maximum capture rate (MCR), as

described in 3.1.5.3. The results of measuring the ET/MCR of cells encapsulated for 120 hrs are

illustrated in Figure 3-5 A and it is seen that there is no significant difference between the

functional properties between groups. There does appear to be a trend toward lower ET and

higher MCR when comparing the Control and High peptide gels, however. The ET and MCR

values of healthy neonatal rat heart tissue were previously measured and are 3.2±1.5 V and

5.1±1.1 Hz respectively (Dengler 2009). While ET of all groups was about double that for native

tissue, the MCR was very close to what is seen in the native heart.

There was a significant difference in the success rate for obtaining beating constructs Figure

3-5 B. Spontaneous beating was consistently observed throughout the entire gel in the High

QHREDGS peptide group, while whole gel synchronous beating was only observed in the

Control group ~50% of time. These observations are consistent with the ~100% success rate for

inducing synchronous contractions in the High QHREDGS group compared to ~55% in the

Control (Figure 3-5 B, P<0.05). When whole gel synchronous beating was not observed there

were no significant local colonies of beating cells (please see L. A. Reis et al. (2011)

Supplementary Movie 1 & Supplementary Movie 2).

It should be noted that there are tissue engineering methods (e.g. electrical/mechanical

stimulation during cultivation) that could potentially improve the functionality of the cultivated

constructs, however since the end goal of this work is to design a hydrogel for cell delivery and

not creating a cardiac patch only these basic assessments of functionality were performed here

(Radisic et al. 2004; Wolfram-Hubertus Zimmermann, Melnychenko, and Eschenhagen 2004).

Page 74: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

58

Figure 3-5: 1:1 Chitosan:collagen hydrogel with immobilized QHREDGS enables cultivation of beating cardiac tissue

(A) ET and MCR show no significant difference between groups after 120 hrs of cultivation (N=11/group, data presented as Mean±SD). (B) There is a statistically significant association between success rate and group, with the High group generating a significantly higher percentage of beating constructs than the Control (*P<0.05). (C) Representative images of paraffin-sectioned constructs with CMs encapsulated in gels and grown in vitro (N=4/group). Live/Dead (PI, red/CFDA, green); Hematoxylin & Eosin staining; Immunostaining for CD31 (brown), Vimentin (green) and cTnT (green) with DAPI counterstained nuclei shown (blue). Scale bars: 20µm H&E, CD31 and vimentin; 0.1mm cTnT.

Samples of constructs grown in vitro for five days were fixed in 10% formalin and sent to be

paraffin-embedded, sectioned, and stained for either Hematoxylin & Eosin (H&E) or cardiac

troponin T (cTnT) (see 3.1.5.7). Live/dead staining was also performed on other samples of

healthy constructs and imaged, as per 3.1.5.2. The results of all the staining are seen in Figure

3-5 C. There were no apparent differences in the Live/Dead (Figure 3-5 C & Figure B-2 A,

Appendix B) or H&E stained sections amongst the groups (Figure 3-5 C), and quantitative image

analysis confirmed no differences in cell viability as assessed by CFDA/PI staining (Figure B-2,

Appendix B). However, the viability was significantly lower in the middle of the hydrogel

compared to the edges (Figure B-2, Appendix B) when hydrogels were cultivated in the tissue

Page 75: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

59

culture plates. This observation is consistent with oxygen diffusional limitations (Radisic et al.

2003). Definitive quantitative conclusions about viability and cell number were made from the

XTT, LDH, and Picogreen assays as due to the 3D nature of the constructs there was

considerable fluorescent background in the Live/Dead images, thereby possibly skewing results

(3.2.2.1).

The presence of endothelial cells was confirmed by CD31+ immunostaining in all groups

(Figure 3-5 C). While ECs were rounded in the Control gel and the Low peptide group, they

exhibited clearly elongated morphology in the High peptide group (Figure 5C). Immunostaining

for vimentin, which indentifies all non-myocytes (i.e. endothelial cells and fibroblasts together)

did not indicate appreciable differences in the presence of these non-myocytes in different

groups (Figure 3-5 C). Cardiac troponin T is a thin filament protein which takes part in muscle

contraction and is expressed and forms myofibrils in healthy cardiomyocytes. In sample images

taken for cTnT staining (Figure 3-5 C) there were larger areas of cTnT positive, elongated cells

forming contractile units in the High and Low groups, and much less in the Control group. High

magnification images indicated the presence of cross-striated cardiomyocytes in the High

QHREDGS group (Figure B-3, Appendix B).

3.2.3. In vivo studies

3.2.3.1. Subcutaneous injection

Subcutaneous injection of CMs and hydrogel was performed as an initial experiment to gauge

the immune response towards the injected biomaterial as well as determine how well the

hydrogel would localize at the site of injection. Prior to in vivo injection, the collagen:chitosan

mixture was pre-gelled for 10 min. In this state the gel was still injectable through a syringe and

needle thus it could be delivered either subcutaneously or into the heart and remain capable of

gelling and localizing at the delivery site. While photocrosslinkable gelation is faster, the low

penetration depth of UV light (~1.5mm) limits applicability of this gelation mechanism in larger

animals (Elisseeff et al. 1999). The recovery rate of the material injected under the skin in the

backs of the adult female Lewis rats was 50% after one week in vivo; five samples were

recovered from the Control group, three from the Low group, and four from the High group. The

unrecovered samples may have moved as the translucent nature of the material matched the

subcutaneous environment into which it was injected. We have studied the degradation rate of a

similar base gel (collagen:chitosan) in vivo using biotin labeling and 1 week subcutaneous

Page 76: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

60

implantation (Chiu and Radisic 2011). It was determined that there was no appreciable

degradation of the gel after 7 days in vivo which is the time-frame consistent with the in vivo

subcutaneous study described here (Chiu and Radisic 2011). The recovered samples exhibited

localized areas of concentrated collagen deposits in contrast to the surrounding tissue, as

identified by the darker blue areas of Mason’s trichrome stained sections (Figure 3-6 A).

Confirmation that these areas where indeed injected hydrogel was done through chitosan

staining, and H&E staining of nodule sections showed no apparent differences in tissue

morphology between samples, seen in Figure 3-6 B & Figure 3-7 A respectively. There was clear

infiltration of smooth muscle actin (SMA) and Factor-VIII (F8) positive cells into the injected

hydrogels (Figure 3-6 C&D respectively), however significant vascular formations could not be

identified. Quantification of the area of positive F8 and SMA staining was done and significant

differences could only be identified in the area of positive SMA staining between the Control

and High groups (Figure 3-6 E). The infiltrating SMA+ cells were most likely reparative

myofibroblasts.

Page 77: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

61

Figure 3-6: Subcutaneous injection study

(A-D) Histological staining of subcutaneously injected hydrogel samples with encapsulated Lewis rat neonatal CMs, recovered 7 days post injection. Full recovered nodules imaged with (A) Mason’s trichrome stain

Page 78: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

62

with areas containing the recovered sample highlighted with black box and confirmed with (B) chitosan staining (positive=dark pink/red), indicated with black arrows. (C) SMA staining indicates a higher expression in the High gel group compared to the Low and Control gel groups while (D) Factor VIII staining shows no major differences between groups. Scale bars=200µm A, 250µm B-D. (E) SMA and Factor VIII positive staining displayed as percent positive pixels. Significant differences seen between Control and Low vs High in the percentage of SMA positive area. Statistical analysis performed by One-way ANOVA with Bonferroni post-tests. *P<0.05, N=5 Control, N=3 Low, N=4 High. Data expressed as Mean±SD.

In the subcutaneous model, after 1 week, there was no appreciable angiogenic effect of the

peptide as assessed by F8 and CD31 immunostaining coupled with identification of tubular

capillary structures, although there were more CD31+ cells in the High group compared to the

Low and Control peptide group (Figure 3-7 B). Vimentin staining, identifying all non-myocytes,

was similar in all groups (Figure 3-7 C). cTnT staining of recovered samples proved the ability

of the hydrogel to retain cells at the site of injection, and further that the injected CMs survived

and matured to a point where they were forming contractile apparati (Figure 3-7 D&E). T-cells

play an important role in the immune and inflammatory response to biomaterials through

infiltration, recruitment of other immune cell types, resorption, fibrosis and ultimate rejection of

foreign bodies motivating the investigation of their presence in the injected hydrogel (van Luyn

et al. 1998). Rare CD3+ T-lymphocytes were identified in all gel groups upon sub-cutaneous

implantation indicating a minimal immune response (Figure 3-7 F).

Page 79: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

63

Figure 3-7: Immunostaining for different cell populations in subcutaneously injected nodules after 7

days in vivo (A) H&E staining shows no apparent differences between groups. Scale bars=250µm. (B) CD31

immunostaining identifies endothelial cells and (C) Vimentin immunostaining identifies non-myocytes:

Page 80: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

64

fibroblasts and endothelial cells. Scale bars=20µm B, 10�m C. (D, E) Expression of cTnT (green) is seen only in the center of nodules correlating with the positive Chitosan staining in Figure 7B, and CMs expressing cTnT are seen in all groups (E). (F) CD3 immunostaining identifies rare recruited T-cells (lymphocytes). Scale bars=10µm. Images correspond to the sections presented in Figure 3-6.

3.2.3.2. Mouse MI model study

Demonstrating the hydrogel’s ability to support CM viability, localize after injection

subcutaneously, retain cells at the site of injection, and promote survival upon transplantation

were all key aspects in its development. However, it was necessary to also demonstrate that the

hydrogel was capable of gelling and localizing at the site of infarct in the beating heart with its

current formulation, and to do this a mouse LAD model was used, as described in 3.1.4.2. Two

weeks post MI surgery the mouse hearts were collected and the site of infarct identified as the

area directly below the ligation suture, seen in Figure 3-8 A. Immediately noted was that both the

MI Only and MI+Control gel hearts appeared very much dilated in comparison to a control Sham

operated heart. Tissue cross-sections encompassing the MI area showed the extreme remodelling

that occurs post MI , with both MI groups showing significant tissue loss and collagenous scar

tissue deposition (Figure 3-8 B&C). Chitosan staining done on paraffin embedded cross-sections

confirmed the gels ability to localize at the site of injection in the infarct area (Figure 3-8 D).

Rare CD3+ T-cells were also identified in both the MI Only and MI+Control gel groups,

indicating injection of Control gel does not seem to significantly alter lymphocyte immune

response (Figure 3-8 E). CD31 positive endothelial cells were seen in the infarct area in both MI

Only and in the injection location in the MI+Control group (Figure 3-9 A). Immature vasculature

and infiltration of SMA expressing cells is seen in Figure 3-9 B&C, identified by F8 and SMA

staining respectively. The most pronounced difference was identified in the amount of SMA

expressing cells present, most likely reparative myofibroblasts, in comparison to the Sham heart.

No difference was found in LV wall thickness and quantification of LV blood vessel

diameter and density showed no significant difference between groups (MI Only and

MI+Control, Figure 3-9 D&E).

Page 81: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

65

Figure 3-8: Chitosan:collagen gel is suitable for injection into the infarcted heart

A left anterior descending coronary artery ligation mouse MI model was used. (A-D) Heart gross morphology and histology 2 weeks post surgery. (A) White arrows indicate ligation site identified by a suture and

Page 82: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

66

(B) infarct area. Injection was done in the area just below the suture immediately following ligation and confirmed with histological staining and immunostaining (C-E). (C) Trichrome images show tissue remodeling and extent of infarct; Scale bars=500µm. Black outlined sections are area of focus for (D and E). (D) Positive chitosan staining is seen in the MI+Control gel group only (black arrow). Scale bars=250µm. (E) Rare CD3+ T-cells are observed in the MI Only and MI+Control Gel Groups. Scale bars=10µm.

It should be noted at this point that there were many limitations to this study, especially in the

number of animals, and further quantification of heart performance pre- and post-MI and with

and without intervention must be done. The purpose of this feasibility study was to evaluate the

applicability of the chitosan:collagen blend for injection into the heart and it did not include the

injection of the QHREDGS peptide modified hydrogels. Detailed studies evaluating cell

injection with the hydrogel and functional improvements at six weeks, the time when

pathological remodeling post MI is completed in both rat and mouse models (Krzemiński et al.

2008; Patten et al. 1998), must be pursued in future studies.

Injection of biomaterials to the infarcted heart may exert several beneficial effects that

attenuate the extent of pathological remodeling. These include mechanical stabilization of the

left ventricle, delivery of bioactive molecules and delivery of reparative cells. To assess the

mechanical effects of biomaterial injection and determine where exactly to make such injections,

Wall et al (2006) performed finite element modeling studies with ovine left ventricle (Wall et al.

2006). They concluded that multiple injections in the border zone surrounding an infarct reduced

end systolic fibre stress proportional to the fractional volume of material added. Thus, hydrogel

injection therapies may have important beneficial effects on cardiac mechanics and can reduce

pathological remodeling post-MI irrespective of the addition of cells or cytokines (Wall et al.

2006). On the same note, Fujimoto et al (2009) set out to design a hydrogel to reduce wall stress

and bulk up the left ventricle (LV) post MI in order to reduce the extent of pathological

remodeling (Fujimoto et al. 2009). They designed a synthetic, thermo-gelling, biodegradable

hydrogel co-polymer from N-isopropylacrylamide, acrylic acid and hydroxyethyl methacrylate-

poly(trimethylene carbonate) (Fujimoto et al. 2009). Showing no in vitro toxic degradation

products they injected the gel into multiple sites along the MI border zone in a rat model. Results

showed preservation of LV cavity area and contractility, and tissue in-growth into the injected

hydrogel, none of which was seen in a PBS injection control (Fujimoto et al. 2009).

Page 83: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

67

Figure 3-9: Vascularization and wall thickness in the mouse MI model (A) CD31,(B) Factor VIII, and (C) SMA staining show appreciable differences between MI and MI+Control

gel groups in comparison to the Sham group. Scale bars=20µm A, 250 µm B&C. (D) LV wall thickness was measured from Trichrome images to assess attenuation of pathological remodeling with injection of Control gel. (E) Factor VIII positive staining was used to identify vasculature and neovascularization. No significant differences were identified between the groups at 2 week time point in this feasibility study. N=3 per group for MI Only and MI+Control Gel, N=1 Sham. Data expressed as Mean±SD.

Page 84: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

68

In our previous work we used photo-cross-linkable chitosan, Az-chitosan, modified with the

novel cell-protective peptide QHREDGS (Rask, Mihic, et al. 2010; Rask, Dallabrida, et al.

2010). While the photo-cross-linkable biomaterial may be applicable for treatment of small

animals such as mice and rats, the UV penetration depth may limit the applicability of photo-

cross-linkable hydrogels in delivery to porcine or human hearts. We thus focused on a hydrogel

blend that cross-links according to a combination of thermal gelling and ionic interactions

between positively charged chitosan and negatively charged collagen. As such this hydrogel has

a larger scope of applicability and it is suitable in catheter delivery as well. The gelation is

performed at mild conditions, cross-linker free, thus is suitable for encapsulation and delivery of

many cell types. In comparison to the photo-cross-linkable gelation which occurs in 1-2 min, the

blend explored here required appreciably longer time (30 min) to gel at 37°C. However, the

delivery and localization to the heart was possible by pre-gelling the blend for ~10 min prior to

injection.

The availability of amine groups on the chitosan backbone enables covalent immobilization of

different biomolecules. Covalent immobilization of ligands is desired over controlled release if

the effects of the ligand are to be observed in the biomaterial rather than the surrounding tissue.

Additionally, covalent immobilization was reported to prolong receptor/ligand signaling (Ito

2008; Fan et al. 2007). In addition to QHREDGS, angiogenic growth factors could also be

covalently immobilized to enhance revascularization of the infarct area. We have successfully

used covalently immobilized VEGF in collagen scaffolds to enhance angiogenesis in a right

ventricular free wall repair (Miyagi et al. 2011). Wu and colleagues have developed a

temperature-sensitive aliphatic polyester hydrogel conjugated with vascular endothelial growth

factor (VEGF) showing that it provided localized and sustained VEGF function, attenuated

adverse cardiac remodeling and improved cardiac function after injection in a rat MI model study

(Wu et al. 2011).

Even though the current feasibility study was not conclusive a number of conclusions can be

made from it. First, the developed hydrogel is capable of localizing at the site of injection in a

beating heart, a result that is undoubtedly the most significant as it means no further components

must be added to the formulation to improve mechanical properties or reduce gelling time.

Subsequent to this observation is that the hydrogel does not fully degrade in vivo at two weeks,

therefore future work must include studies of the in vivo degradation rate of the hydrogel.

Page 85: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

69

Secondly, while the base hydrogel formulation (1:1 Control- No Peptide) does not appear to

improve heart function post MI it at least does not cause any further damage to the infarcted

mouse heart (in animals that survived the procedure) and is biocompatible. This is significant as

any hydrogel, scaffold, etc. destined to be implanted into the body must not cause (further)

damage, or alternatively diminish improvement, or else a lack of intervention would be the best

option. Finally, this model provides a basis for assessing the performance of hydrogel with

conjugated QHREDGS peptide, which should hopefully show significant improvement in

comparison to both no treatment and Control gel, and therefore puts into context the lack of

improvement seen in the current mouse study.

3.3. Summary

SA-1 Develop a chitosan-collagen hydrogel that supports CM viability and phenotype.

Conjugate QHREDGS and demonstrate dose dependent improvement in CM phenotype and

function in vitro.

These data indicate that a 2.5 mg/mL 1:1 m:m chitosan:collagen blend gels without addition

of exogenous cross-linkers in ~30 minutes at 37°C. Furthermore, this formulation provides the

appropriate stiffness and mechanical properties to support cardiomyocyte survival both in vitro

and in vivo. Addition of the peptide did not significantly affect the rheological properties or

porous structure of the hydrogel. XTT assay and success of obtaining beating constructs

demonstrated clear effects of the incorporation of the QHREDGS peptide on cell viability and

functional properties in vitro. In vivo, High QHREDGS gel attracted more reparative

myofibroblasts and improved the presence of cardiomyocytes in the subcutaneous model. The

developed hydrogel has been shown to successfully localize at the site of injection both

subcutaneously and at the site of infarct in a mouse MI model. Further studies are required to

determine hydrogel degradation rate in the infarcted heart as well as functional improvements

upon injection of the QHREDGS modified hydrogels with and without cells.

Nonetheless, the work done represents that toward completion of Specific Aim SA-1, and the

further animal models described above is the focus of Specific Aim SA-2 and therefore the focus

work described in Chapter 4.

Page 86: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

70

4. Utility of QH-G213-H hydrogel as a treatment for acute MI

The original work described in this chapter has been published in Circulation Heart Failure

(Reis et al. 2015), and is used with permission from Wolters Kluwer Health.

Having developed a hydrogel formulation that performed very well in vitro and was showing

promise as a delivery vehicle for both cells and cytokines/growth factors in our subcutaneous in

vivo work, it was both logical and necessary to move toward an in vivo animal MI model. As

described in 3.2.3.2, in order for the developed hydrogel to be determined sufficient in its current

formulation it would have to prove capable of being injected and localizing at the site of infarct

within the beating heart. As the mouse LAD procedure is widely accepted as a good MI model

and the surgeon (Dr. Abdul Momen, University Health Network) had vast experience performing

the MI surgeries in mice we proceeded with a study to determine the effect of the developed

hydrogel in vivo. Our preliminary mouse MI model study design was for a total of 25 animals.

However, the severe trauma induced by the LAD surgery, and in many instances followed by

injection of a large volume of gel (in comparison to overall heart volume), proved too much and

there was only ~30% survival at two weeks compared to reported ~50% survival for similar

studies (Kumar et al. 2005; Sam et al. 2000). To address this issue it was decided to switch from

mice to rats for MI model studies as the injection post MI should cause much less trauma to the

animals in comparison to the trauma from the initial MI, and the rat LAD MI model is also

widely accepted and studied (Krzemiński et al. 2008). Initial results from a trial study using a

Lewis rat LAD MI model, done in the exact same manner as that with mice proved to be very

promising. We achieved 100% survival at three weeks, and it seemed as though injection of both

Control (No Peptide) and peptide modified (QH-G213-H) 2.5mg/mL 1:1 chitosan-collagen

hydrogel were having a marked positive effect on LV remodeling post MI. These results

motivated us to pursue further studies into the effect the developed hydrogel (alone) had on LV

remodeling, and the mechanism by which it acts.

4.1. Materials & Methods

All animal experimental procedures were approved by the Animal Care Committee of the

Toronto General Research Institute and the University of Toronto Committee on Animal Care,

according to the Guide for the Care and Use of Laboratory Animals.

4.1.1. Experimental design overview

Page 87: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

71

A brief overview of the experimental design for this chapter, along with all relevant time

points and quantification done at each step are illustrated in Figure 4-1.

Figure 4-1: In vivo experimental design A brief overview of the design used for the large scale Lewis rat LAD MI model with major time-pints and

procedures performed.

4.1.2. Peptide modified chitosan-collagen hydrogel (QHG213H) QHREDGS peptide (Biomatik) was conjugated to chitosan (UP-G213, Novamatrix) using 1-

ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) chemistry as described previously (Reis et

al. 2012). Similarly, 2.5 mg/mL 1:1 mass:mass chitosan-collagen hydrogel was prepared as

described to make Control hydrogel (no peptide) or QHG213H hydrogel (651±8 nmol

peptide/mL-gel) and kept on ice for up to 3 hours until use (Reis et al. 2012).

4.1.3. Peptide modified polyethylene glycol (PEG-QHREDGS) QHREDGS peptide was conjugated to polyethylene glycol (PEG) by incubating 8.0 mg

acrylate-PEG-NHS (MW 3500; Jenkem Technologies, Allen, TX) in a solution of 50 mM Tris

pH 8.5 containing 0 mM or 29 mM (24 mg/mL) QHREDGS solution for a final volume of 200

µL, for 3h at 700rpm. After 3 h, the reaction solutions were dialyzed extensively against ddH2O;

lyophilized and resuspended in ddH2O, to which 80 mg polyethylene glycol diacrylate (PEGDA

Mn 700; Sigma Aldrich, Oakville, ON) and 0.8 mg 2-Hydroxy-2-methyl-propiophenone was

added; 150 µL of this solution was added to a circular silicon mold (11-mm diameter, 1-mm

thick) placed atop a glass microscope slide and a polyvinyl coverslip was placed on top of the

mold. The hydrogel was placed under UV light for 10 min to polymerize, washed for 10 min in

phosphate buffered saline (PBS) then incubated at 4°C for 1 hr. After 1 hr the cover-slip was

gently peeled off the hydrogel, the mold was removed and the hydrogel was placed in 70% EtOH

under UV for 1 hr to sterilize, then placed in sterile PBS for 1 hr at 37°C.

Neonatal rat CMs or cardiac fibroblasts, isolated as previously described (Rask, Dallabrida, et

al. 2010; Radisic et al. 2003), were washed and resuspended in equal volumes of PBS or 50

Page 88: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

72

µg/mL anti-β1-integrin antibody (α-CD29; BD Biosciences, Mississauga, ON) at 37°C for 20

min. The CMs or cardiac fibfroblasts were then spun for 5 min at 200g, resuspended in CM

media, seeded onto the PEG-hydrogels (22,000 cells per hydrogel) in a volume of 10 µL and

incubated at 37°C for 50 min. After 50 min, the hydrogels were washed once with PBS, fixed

with 4% paraformaldehyde solution for 20 min, blocked for 1 hr in 5% FBS, 0.3% Triton X-100

in PBS, washed thrice in PBS, stained with DAPI for 15 min, then washed thrice with PBS.

Washed hydrogels were imaged in PBS using an Olympus IX-81 fluorescent microscope with

CellSens Dimension Imaging software (Olympus America Inc., Center Valley, PA). The surface

of each hydrogel was imaged in its entirety and the total number of cells was determined using

the maxima plug-in filter of ImageJ software (NIH, Bathesda, MD) and summing the values

determined from each individual image for a given hydrogel. The total number of adherent cells

was normalized by dividing by the total number of cell that adhered to the PEG hydrogels that

did not contain any QHREDGS peptide for a given experiment.

4.1.4. In vivo studies Lewis rats (weighing 200–250 g) were obtained from Charles River Laboratories (Saint-

Constant, QC, Canada), and myocardial infarction (MI) was generated under general anesthesia

by occluding the left anterior descending (LAD) coronary artery as previously described (Kan et

al. 2007). The Animal Care Committee of the University Health Network approved all animal

procedures. Experiments were performed according to the Guide to the Care and Use of

Experimental Animals from the Canadian Council on Animal Care. Injection of PBS (MI Only),

Control gel (no peptide), or QHG213H (peptide modified) hydrogel was done immediately

following LAD ligation and prior to closure of the thorax. Injection (3 locations, 50 µL total)

was performed using 28-guage syringes (BD Biosciences) by inserting the needle into the peri-

infarct LV wall and directing into the developing scar (MI zone). Syringes with PBS or hydrogel

were prepared prior to surgeries and kept on ice, then warmed at ~37°C for 10 min prior to

injection (Reis et al. 2012).

4.1.4.1. Assessment of cardiac function (6 Week time point)

N=8 animals per group were used for a long term 6 week study. Cardiac function was

evaluated by echocardiography (Sequoia C256 System, Siemens Medical; 15 MHz linear array

transducer) before MI (pre-ligation baseline), 3 and 6 weeks post MI. M-mode images were

obtained in the parasternal short-axis view at the level of the papillary muscles. The

Page 89: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

73

measurements were performed by a single, blinded examiner. LV internal diastolic dimension

(LVIDd) and internal systolic dimension (LVIDs) were measured. Three consecutive cardiac

cycles were recorded and averaged. Percent fractional shortening (%FS) was calculated as

follows: %FS = (LVIDd − LVIDs) / LVIDd 100 (Kan et al. 2007). The 6 week time point was

selected as it is assumed the heart has completed remodeling by this time. PBS, Control gel (no

peptide), or QHG213H (peptide modified) hydrogel was injected into 3 locations in the LV wall

(50 µL total) surrounding the infarct using a 28-gauge insulin syringe. At 3 weeks post injection

rats with no infarct or with a very large infarct were excluded from the study; only those

exhibiting a 20-40% fractional shortening as determined by echocardiography were included.

One animal from each group did not meet the criteria and were excluded, reducing numbers to

N=7/group. Cardiac function was also assessed at the end of the study (6 weeks after treatment)

with a pressure-volume catheter as previously described (Z. Sun et al. 2008). Briefly, the rats

were anesthetized and intubated with mixed oxygen and room air by a rodent ventilator (Harvard

apparatus, Canada). A pressure-volume catheter-tipped pressure transducer (2F, Millar

Instruments, USA) was inserted into LV cavity via right carotid artery. Volumes were measured

by conductance, and end-systolic and end-diastolic values were recorded. The ejection fraction,

dP/dt Max, dP/dt Min, end systolic pressure-volume relationship (ESPV) and preload recruitable

stroke work (PRSW) were calculated.

After the pressure-volume analysis was complete, hearts were perfusion fixed with 10%

formaldehyde, excised, and sectioned into five 2-mm thick slices as previously described (Kan et

al. 2007; Z. Sun et al. 2008).

4.1.4.2. 24 hr time point

N=6 animals per group were used in a short 24 hr study to elucidate the mechanism by which

the injected hydrogel preserved cardiac function. Surgery and injections were performed in the

same manner as described above and animals were monitored overnight. The animals were

anaesthetized 24 hr after injection with isoflurane, intubated, ventilated, and maintained with 2%

isoflurane. The heart was accessed in the same manner as performed for LAD surgery and

arrested using 10% KCl injected directly into the LV. Hearts were then immediately excised and

the right ventricle and atria were removed. The LV was then sectioned into a MI region, border

region, and remote region. These sections were placed in separate cryo-tubes and snap frozen in

Page 90: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

74

liquid N2. Frozen tissues were subsequently ground using standard mortar and pestle with liquid

N2 and kept at -80°C prior to RNA and protein extraction.

4.1.4.3. In vivo hydrogel lifespan

To assess the in vivo lifespan of our hydrogel, chitosan was conjugated with the near infrared

dye Dy-Light 800 (Thermo Scientific, Waltham, MA) and injected into the heart of live animals.

Briefly, 1 vial of Dy-Light 800 label (suitable for 1 mg of protein) was dissolved in 57.7 μL PBS

and mixed immediately with 92.3 μL of chitosan at 20 mg/mL in 0.9% normal saline (final

reaction volume of 150 μL and chitosan concentration of 12.31 mg/mL). The mixture was

protected from light, placed on a microtube shaker at 650 rpm for 1 hr, and then stored at 4°C

until use. Upon use, the Dy-Light labelled chitosan was used to make hydrogels as per the

standard protocol (Reis et al. 2012), kept on ice, and then 50 μL of the labelled gel was injected

directly into 3 locations of the healthy LV wall of N=7 Lewis rats. Rats (anesthetized under

isoflurane) were then imaged using a Kodak In-Vivo FX Pro Imager (Kodak Molecular Imaging

Systems), with a 5 sec exposure X-ray image overlaid with a 40 sec exposure fluorescent image

(760 nm excitation, 830 nm emission). Full-sized images were processed to illustrate fluorescent

decrease over time by applying the same threshold and gamma adjustment to all images using

Kodak MI 4.0 software (Kodak Molecular Imaging Systems). Inset images were processed to

highlight fluorescent localization by applying the optimal brightness/contrast and threshold to

each individual image using ImageJ (NIH). N=3 rats were sacrificed 1 hr after injection, and

N=4 were monitored up to 14 days post injection. Excised hearts were stored at -80°C until use,

at which time they were homogenized and fluorescently imaged to quantify degradation.

4.1.5. Immunohistochemistry Samples for immunohistochemical and immunofluorescent staining were prepared as

described above. For paraffin embedding and sectioning, as well as hematoxylin and eosin

(H&E), Mason’s trichrome, smooth muscle actin (SMA), Factor VIII (FVIII), and CD31

staining, the fixed samples were sent to the Pathology Research Program (PRP) at the University

Health Network. Chitosan staining was performed using Cibacron Brilliant Red-3BA (CBR-

3BA, Sigma Aldrich) and Weigert’s Iron Hematoxylin as described (Rossomacha, Hoemanni,

and Shive 2004; Reis et al. 2012).

Sections stained for cardiac troponin-T (cTnT) and TUNEL were deparaffinised and

rehydrated with subsequent baths (3 each) of 100% xylene, 100% EtOH, 95% EtOH, 75% EtOH,

Page 91: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

75

and distilled water for 3 min per change. Antigen retrieval was performed by microwaving slides

immersed in Tris-EDTA buffer for 5 min, permeabilized with 0.1% Triton-X, 0.1% Sodium

Citrate in Tris-buffered saline (TBS) solution (10 min), and blocked using Dako Serum Free

Protein Block (Dako Canada, Inc., Burlington, ON X0909) for 30 min at room temperature, with

3x3 min TBS washes between steps. Primary antibody (mouse monoclonal cTnT antibody,

Thermo Scientific MS-295-P, diluted 1:200) was applied to sections for 2 hr at room temperature

followed by secondary antibody (Alexafluor 594 goat anti-mouse IgG (H + L), Invitrogen) at

1:400 dilution for 1 hr at room temperature. TUNEL label and enzyme (Roche Applied Science,

Indianapolis, IN, 11767291910) were applied as per manufacturer’s instructions. DAPI (Sigma

Aldrich) nuclear stain was applied at a 1:1000 dilution for 10 min, and finally slides were

mounted using Dako Fluorescence Mounting Medium (Dako Canada, Inc., S3023), and imaged.

4.1.6. Quantitative PCR Whole heart or heart sections frozen at -80°C were ground using a mortar and pestle and

liquid N2. RNA was extracted with TRIZOL Reagent following the manufacturer’s protocol.

Isolated RNA was resuspended in 200 μL RNAse/DNAse free water and quantified using a

NanoDrop 1000. cDNA synthesis and RT-qPCR were performed as previously described (Nunes

et al. 2013). All oligonucleotide sequences used are listed in Table C-1 (Appendix C). Data was

standardized between samples to reflect gene expression changes of the progressing scar,

quantifying gene expression in the border zone compared to the established MI zone. Fold

changes in gene expression between groups were analyzed and genes showing a >2-fold or <0.5-

fold change in the QHG213H gel group over the MI Only (PBS) injection group were considered

significant.

4.1.7. Western blotting Whole heart or heart sections frozen at -80°C were ground using a mortar and pestle and

liquid N2. Protein was isolated using Pro-Prep Protein Extraction Solution (iNtRON

Biotechnology, Inc., Seongnam, KR) according to the manufacturer’s instructions. Samples were

run on Novex® Tris-Glycine 1.5-mm, 10-well, 10% Gels (Life Technologies) for 1.5 hr at 125 V

then transferred to nitrocellulose membrane by wet transfer at 125 V for 2.5 hr. Membranes were

probed with: α-phospho-MAPK (1/1000 in 5% BSA w/v in PBS with 0.1% Tween20), MAPK

(1/1000), α-ILK (1/1000), or GAPDH (Millipore, MAB374, 1:10,000) antibodies. Secondary

antibodies used were peroxidase-conjugated (DAKO, P0448 or P0447, 1:2000). Membranes

Page 92: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

76

were developed with ECL reagent Luminata Classico Substrate (Millipore) and exposed to film.

The films were scanned and densitometry was performed using ImageJ software.

4.1.8. Image analysis techniques Morphometric analysis was performed as described previously (Kan et al. 2007; Z. Sun et al.

2008). TUNEL/DAPI cell counts were made using a macro developed in Olympus CellSens

Dimension software and tested on control images. TUNEL/cTnT MI border zone quantification

was based on 2 independent images on either side of the MI zone in each animal, and MI zone

quantification performed by averaging at least 4 independent images within the scar. All counts

and normalizations were performed for each image and averaged to get a single value per

sample. Quantifying differences in vasculature in the MI zone and border zone of hearts excised

3 and 6 weeks post MI was done using SMA and FVIII/CD31 stained sections as described

previously (Chiu and Radisic 2010).

Automation of the image quantification process eliminates human errors and bias between

images, ensuring consistent quantification among all images and more accurate results.

4.1.9. Statistical analysis Statistical analysis was performed using SPSS Statistics 17.0 and GraphPad Prism 5.0.

Differences between experimental groups were analyzed using one or two-way ANOVA with

Bonferroni post-hoc tests unless otherwise specified. P<0.05 was considered significant for all

statistical tests. Results were plotted with GraphPad Prism 5.0, with all data being reported as

mean ± standard deviation (Mean±SD), unless otherwise noted.

4.2. Results

4.2.1. In vivo degradation (lifespan) Animals imaged 1 hr after injection of the Dy-light 800 labelled hydrogel showed localization

of labelled hydrogel within the heart; with unconjugated dye and gel washout in what appeared

to be the chest of the animals (Figure 4-2). Images taken 24 hr after injection similarly showed

localization of labelled hydrogel within the heart while the fluorescence in the chest that was

visible in the 1 hr images was no longer evident, rather waste gel/dye appeared to be present in

the stool of the animals. Subsequent images at 3, 5, 7, and 14 days post injection show continued

localization of labelled hydrogel in the heart; however the extent of fluorescence decreased with

time. By 14 days post injection only a minimal amount of fluorescent label was visible (Figure

Page 93: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

77

4-2). Staining for the presence of chitosan in heart sections derived from hearts excised 3 weeks

post injection confirmed the presence of hydrogel. However, positive chitosan staining was

evident in only ~20% of animal hearts indicating the in vivo lifespan of the hydrogel in the heart

was approximately 3 weeks (Figure 4-2).

The remaining (N=4) animals were sacrificed after imaging on day 14, the excised hearts

were homogenized, and the fluorescence intensity was measured and compared to N=3 hearts

excised 1 hr post injection. The results indicated that approximately 60% of the gel present at 1

hr is cleared by 14 days (~40% remained on day 14, Figure C-1, Appendix C).

Figure 4-2: In vivo lifespan of Dy-light 800 labelled chitosan-collagen gel Dy-light 800 labelled gel was detectable in the heart of live animals using a Kodak In-Vivo FX Pro imager

from 1 hour (hr) up to 14 days (d) post injection (visualized on day 14 with increased exposure, ↑exp.). Insets. Enlarged and enhanced view of the region indicated by the white square. Chitosan staining of heart sections extracted 3 weeks (Wk) post injection showed continued presence of chitosan in small islands in the infarct area (purple dots); however this was only observed in a minority (~20%) of samples indicating almost complete clearance of the hydrogel from the heart.

4.2.2. Functional Data All animals exhibited nearly identical echocardiographic parameters prior to surgery

irrespective of their grouping. The LAD coronary artery ligation procedure resulted in significant

ventricular dysfunction and LV dilation. These were seen as a decrease in fractional shortening

and an increase in both LV internal dimension at systole and diastole (LVIDs and LVIDd,

respectively) at 3 weeks, which was exacerbated by 6 weeks (Figure 4-3 A-C). Interestingly

there was a significant amelioration of cardiac remodeling, measured by cardiac fractional

shortening and LVIDs, in the Control gel (no peptide) group as compared to the MI Only (PBS)

Page 94: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

78

group at both 3 and 6 weeks, however the QHG213H gel group outperformed both the MI Only

(PBS) and Control gel groups (Figure 4-3 A). Most of the decrease in fractional shortening for

the MI Only and Control gel groups occurred in the first three weeks after MI, as there was no

significant reduction in fractional shortening between 3 and 6 weeks for either group, while the

decrease was lesser and more gradual with QHG213H gel injection (Figure 4-3 A&D).

Measurement of LVIDs showed that gel treatments with and without the QHREDGS peptide

significantly reduced LV remodeling during systole (LVIDs) over the monitoring period (Figure

4-3 B), but did not have a marked effect on the diastolic dimension (LVIDd, Figure 4-3 C).

There was a statistically significant level of interaction between factors Group and Time for %FS

and LVIDs (P<0.001) but not for LVIDd (Figure 4-3 A-C).

At 6 weeks post MI, hearts were further evaluated using load dependent and load independent

measurements to assess cardiac function and LV volumes. Steady-state (load dependent) ejection

fraction, dP/dt Max, and dP/dt Min were all significantly better in the QHG213H gel group as

compared to either the Control gel or MI Only (PBS) groups (Figure 4-4 A-C). Notably,

injection of the Control gel (no peptide) itself showed a significant improvement in cardiac

function as compared with the MI Only (PBS) group (Figure 4-4 A-C). Furthermore, the smallest

end systolic LV volumes were seen in the QHG213H group, followed by the Control gel group,

which were both significantly lower than the MI Only (PBS) group (Figure 4-4 D). However, no

difference was observed in the end-diastolic LV volumes between groups (Figure 4-4 E).

Page 95: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

79

Figure 4-3: Cardiac function post MI measured by echocardiography QHG213H gel injection significantly maintained cardiac function (measured by %fractional shortening,

%FS) at 21 days (3 weeks) and 42 days (6 weeks) post LAD surgery as compared to the Control gel (no peptide) and MI Only (PBS) injection groups; and the Control gel injection induced a significant maintenance in %FS relative to the PBS injection (MI Only) (A). Injection of gel with or without peptide significantly reduced LV dilation during systole at 3 and 6 weeks relative to the PBS injection (MI Only); and the QHG213H gel induced a significant improvement in LVIDs relative to the Control gel (B). No difference in diastolic LV dilation (LVIDd) was seen over the monitoring period (C). %FS and LVIDs had statistically significant interaction between factors Group and Time (P<0.0001), but no interaction effect was present for LVIDd. (D) Post-hoc pair-wise comparisons for factor “Days after Injection” for data from (A-C). Data analyzed by two-way repeated measures ANOVA and Holm-Sidak post-hoc analysis and expressed as Mean±SD, N=7/group.*P<0.01, **P<0.001 vs MI Only; #P<0.01 vs Control gel.

Page 96: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

80

Figure 4-4: Load dependent cardiac function and LV volumes at 6 weeks post MI Steady-state pressure-volume analysis was used for load dependent cardiac analysis after heart excision.

Injection of the Control gel (no peptide) significantly improved cardiac function compared to MI Only (PBS), and injection of the QHG213H gel further improved function (A-C). End systolic LV volume was significantly lower in the QHG213H gel group vs the Control gel and MI Only (PBS) groups, and in the Control gel group vs MI Only (PBS) group (D). The end LV diastolic volume was unchanged between groups (E). Data expressed as Mean±SD, N=7/group, *P<0.05, **P<0.01 vs MI Only (PBS), #P<0.05 vs Control gel.

The load independent end systolic pressure-volume relationship (ESPVR, Figure 4-5 A) and

preload recruitable stroke work (PRSW, Figure 4-5 B) were significantly improved with

injection of gels with or without peptide as compared to the MI Only (PBS) group. However,

addition of the peptide to the gel enhanced its protective effects by inducing further

improvements to these parameters Figure 4-5 A&B).

Page 97: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

81

Figure 4-5: Load independent pressure-volume analysis at 6 weeks post MI Occlusion pressure-volume analysis was used for load independent cardiac functional analysis after excising

hearts. Significant differences were seen in both end systolic pressure (A) and stroke work (B) between the QHG213H gel group and either the Control gel MI Only (PBS) groups, as well as between the Control gel group and the MI Only (PBS) group. Data expressed as Mean±SD, N=7/group, *P<0.05.

4.2.3. Gross morphology and histology PBS injected (MI Only) hearts appeared larger than Control (no peptide) gel and QHG213H

gel injected hearts; and the extent of remodeling appeared more extensive in the PBS injected

heart cross sections (Figure 4-6 A). Quantification of the scar fraction and MI zone scar

thickness from fixed sections showed that the MI Only (PBS) hearts underwent the most

extensive remodeling; that the Control gel injected group had significantly smaller fractional scar

coverage and significantly greater scar thickness than the MI Only (PBS) group; and that the

QHG213H gel group had significantly smaller fractional scar coverage and significantly greater

scar thickness than either the Control gel or MI Only groups (Figure 4-6 B&C).

Page 98: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

82

Figure 4-6: Gross heart morphology at 6 weeks post MI Representative whole heart and heart sections (from just below suture to apex of heart, 5 sections per heart)

illustrate differences in gross morphology between groups (A). Fractional MI scar coverage (B) and MI scar thickness (C) showed significant differences between groups, with QHG213H gel having significantly smaller scar coverage and larger scar wall thickness than both Control gel and MI Only (PBS) groups, as determined from gross section images (A). Data expressed as Mean±SD, N=7/group, *P<0.05, **P<0.01 vs MI Only (PBS), #P<0.05 vs Control gel.

Heart sections were histochemically stained and imaged for the different treatment groups.

Differences in scar thickness and area between treatment groups was apparent and there also

appeared to be more healthy tissue (red) within the MI zone in the QHG213H group in

comparison to the other groups based on Masson’s trichrome staining (Figure C-2 A, Appendix

C). Sections were additionally stained for SMA (a marker of smooth muscles cells present in

Page 99: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

83

mature vasculature), as well as both CD31 and FVIII (markers of endothelial cells and found in

both neo- and mature vasculature) (Figure C-2 B-D, Appendix C). The mean vessel density and

diameter of total vascularization (FVIII+, Figure 4-7 top panel) and mature vasculature (SMA+,

Figure 4-7 bottom panel) were quantified for both the MI and border zones. No difference in

vasculature metrics was seen in the MI zone, or in the FVIII+ vessels in the border zone.

However, there were significantly larger mature SMA+ vessels in the border zone for the

QHG213H gel treatment group as compared with either the MI Only (PBS) and Control gel

groups.

Figure 4-7: Vascularization at 6 weeks post MI Vascularization within the MI and border zones was quantified at 6 weeks from heart cross sections. Vessel

density and diameters were quantified from SMA stained sections to assess mature vasculature, and from FVIII stained sections for total (neo- and mature) vascularization. The QHG213H gel group had significantly larger mature SMA+ vessels in the border zone. Data expressed as Mean±SD; N=7/group.

Page 100: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

84

4.2.4. Cardiomyocyte survival mechanism TUNEL and cTnT staining was used to determine the number of apoptotic CMs in the MI and

border zones (at 6 weeks post infarct), in order to determine if the functional and morphological

improvements seen with the Control and/or QHG213H gel injections could be attributable to a

reduction in CM apoptosis. Fewer TUNEL+ (apoptotic) cells were visible within the border

region of the Control and QHG213H gel groups as compared to the MI Only group (Figure 4-8

A). Quantification of the TUNEL staining revealed that gel treatment with or without peptide

significantly reduced the fraction of apoptotic cells in the border zone (P=0.005 MI Only vs.

Control gel, P=0.012 MI Only vs QHG213H gel; Figure 4-8 B). In the MI zone, there was no

difference in the apoptotic cell fraction among the treatment groups, however the QHG213H gel

group had a higher proportion of cTnT+ cells than either the MI Only (PBS, P=0.008) and

Control gel (P=0.005) groups (Figure 4-8 C) at six weeks after treatment.

To gain further insight into the CM survival mechanism, RNA isolated from tissues from the

MI and border zones of hearts excised 24 hr after treatment were subjected to RT-qPCR and a

total of 27 genes were assessed, including genes conventionally ascribed as apoptotic,

necroptotic, pro-survival, anti- or pro-inflammatory, and cardiac-specific. Notably, between the

QHG213H gel and MI Only (PBS) groups, there was a 4.50- and 2.29-fold increase in the pro-

inflammatory interleukin (IL)-6 and IL-1β gene expression, respectively; a 7.69-fold decrease in

the pro-apoptotic caspase-9 (CASP9) gene expression; a 2.32-fold decrease in the pro-survival

PI3K gene expression; and a 2.17-fold increase in the anti-apoptotic BCL2 gene expression

(Figure C-3, Appendix C). In the case of all the aforementioned genes, the addition of

QHREDGS peptide to the gel resulted in a larger fold change in the expression levels than the

Control gel (Figure C-3, Appendix C). It must be noted that none of these fold changes were

statistically significant and we can only comment on the trend observed.

Additionally, Western blot analysis was performed to determine if integrin-linked kinase

(ILK) and MAPK were implicated in the CM survival mechanism as both proteins have been

shown to be up-regulated by QHREDGS to promote stem cell survival in recent work (Dang et

al. 2014). In analyzing border zone tissues we did not see a difference in ILK protein levels or

MAPK phosphorylation between the groups, however we did see a significant increase in MAPK

protein expression in the QHG213H peptide gel group over the Control gel and MI Only (PBS)

groups (Figure C-4, Appendix C).

Page 101: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

85

To determine if the interaction of CMs and cardiac fibroblasts with the QHREDGS peptide

was dependent on the β1-integrin subunit (identified as the receptor in endothelial cells and stem

cells) polyethylene glycol (PEG) hydrogels with and without the QHREDGS peptide were

generated (Miklas et al. 2013; Dang et al. 2014). PEG was chosen as the base hydrogel because it

is non-fouling in short term culture scenarios and does not promote cell attachment; tissue

culture plastic (TCP) was used as a positive control for cell attachment. The peptide

concentration in the PEG hydrogel was 11.7±4.5mg/mL (9.7±3.7nM) (Feric et al. 2014). The

addition of the QHREDGS peptide to the PEG hydrogel significantly increased the number of

adherent cells relative to PEG alone (Figure 4-9A&B). Pre-incubation of either cell type with an

anti-β1-integrin (CD29) antibody reduced CM adhesion to the QHREDGS-PEG hydrogel to a

level comparable to that of the PEG hydrogel without peptide (Figure 4-9A&B).

Figure 4-8: Apoptosis at 6 weeks post MI Sections from hearts extracted at 6 weeks were stained for TUNEL (red), cTnT (green), and DAPI nuclear

stain (blue). Representative images showed fewer TUNEL+ cells in the Control gel and QHG213H gel groups than in the MI Only (PBS) group in the border zone (Scale bars=100µm) (A). Quantification of the fraction TUNEL+ cells (# of TUNEL+ cells/# of DAPI+ cells determined per image) in the border zone showed a significantly smaller TUNEL+ cell fraction for both the Control and QHG213H gel groups relative to the MI

Page 102: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

86

Only (PBS) group, but no statistical difference between the gel treatment groups (B). In the MI Zone, there was no difference in the TUNEL+ cell fraction between groups, however the QHG213H group showed a significant increase in cTnT expression relative to either the MI Only (PBS) and Control gel groups (C). Data expressed as Mean±SD,* P<0.05, ** P<0.01, N=7/group.

Figure 4-9: Cardiac cell attachment to PEG and PEG-QHREDGS gels Rat NCMs (A) or cardiac fibroblasts (B) were seeded onto PEG only (no peptide) or PEG-QHREDGS disks with or without

the pre-incubation with an anti-β1-integrin (CD29) antibody. After 50 min, the attached cells were counted. The data is normalized to the PEG only gels. Two way ANOVA showed both NCMs and cardiac fibroblasts have significantly more attached cells to PEG-QHREDGS surfaces over PEG alone, but the difference is abolished when pre-incubated with anti-β1-integrin antibody. Data expressed as Mean±SE, * P<0.05, ** P<0.01, N=6/group.

4.3. Discussion

We have demonstrated, by means of in vivo tracking that our chitosan-collagen hydrogel can

localize within the beating heart and can remain in situ for at least two weeks after injection. We

have also shown by chitosan staining of heart sections that 3 weeks after injection hydrogel was

present in ~20% of samples. Moreover, we found a significant improvement in cardiac function

at 6 weeks post MI in an LAD rat model with injection of the chitosan-collagen hydrogel, and

further functional improvements with injection of the QHREDGS conjugated chitosan-collagen

hydrogel. Finally in terms of mechanism, we found that adhesion of rat NCMs to the QHREDGS

peptide is mediated by β1-type integrin receptors; and that in the border zone protein levels of the

pro-survival molecule MAPK was significantly increased by the presence of QHREDGS in the

hydrogel at 24 hr. We observed trends toward up-regulated gene expression of the pro-

inflammatory interleukins IL-6 and IL-1β and the anti-apoptotic protein BCL2, whereas gene

expression of the pro-apoptotic enzyme caspase-9 appeared to be down regulated.

Hydrogel degradation is most often characterized in vitro or by histological staining of

excised in vivo samples (Bouten et al. 2011; Das et al. 2012). While techniques for on-line in

vivo quantification of hydrogel distribution and degradation exist for subcutaneous and intra-

Page 103: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

87

muscular injections (biotin labeling, in vivo fluorescent imaging systems, MRI, PET, etc), using

these systems for tracking hydrogels in the heart prove extremely difficult for a number of

reasons. These include the dynamic nature of the heart, which continues to beat even when the

animal is sedated, the depth at which the heart is located within the animal, and the often small

volume of material injected therein (~20-200 µL) (Bouten et al. 2011; Ruggiero et al. 2012). The

near infrared dye Dy-Light 800 was used in this study specifically due to its compatibility with

the Kodak In-Vivo Imaging System. This is because this system has the potential to provide on-

line information regarding the localization and lifespan of the hydrogel and the potential to

minimize the number of animals required for the study by eliminating the need for heart tissue

extractions at various time points from which to assess the distribution and degradation of the

hydrogel. Furthermore, the use of a near infrared dye permits deep tissue imaging without

affecting the surrounding tissue as in the case of long term radiolabels used for PET imaging, or

the inaccurate degradation profiles that can result from macrophage uptake of the contrast agents

(e.g. iron oxides or gadolinium) used for MRIs (Ruggiero et al. 2012). However, near infrared

fluorescence imaging has its own limitation in that it provides the lowest spatial resolution.

By imaging the injected labelled hydrogel over a 14 day period, we were able to confirm

localization and retention of the hydrogel within the heart (Figure 4-2), however full

quantification of the degradation rate proved more challenging. Quantification from the images

was not possible because the 2D imaging system could not provide the necessary 3D information

about the volume of injected material and because of distortion in the images due to the constant

beating of the heart over the 40 sec imaging period. Furthermore, we did not fully quantify the

conjugation efficiency, however as the mechanism of DyLight800 conjugation to chitosan is the

same as that for peptide conjugation used we assumed efficiency was about equal at 50%.

Quantification was therefore attempted using excised tissue samples and required two more

assumptions: (i) the bulk of unbound dye was removed from the gel within the first hour

following injection and (ii) the gel present at 1 hr after injection represented the total amount of

gel that was successfully localized. With these assumptions, a comparison of the amount of gel

present at 14 days and the amount present at 1 hr post injection resulted in the information that

approximately 60% of the gel was degraded or removed from the heart over a two week period

(Figure C-1, Appendix C).

Page 104: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

88

One question that has been raised concerning the in vivo presence of the gel is what happens

to it after being removed from the heart, and specifically why there is presence of fluorescence

(thus chitosan) in the animal’s urine and lack thereof in the kidneys or liver. It is recognized that

the in vivo biodistribution of chitosan is one of the least studied aspects of the compound, with

molecular weight, deacetylation, and particle sizes all having an effect (Kean and Thanou 2010).

Two studies in particular describe the clearance of water soluble sugars of similar size to the

chitosan we used (Yamaoka, Tabata, and Ikada 1993; Onishi and Machida 1999). The first study,

looking into the clearance of dextrans and pollulans of varying MW from the blood finds that

even the largest sugars (100s of kDa) are processed and accumulate in animal excrements within

hours (Yamaoka, Tabata, and Ikada 1993). The second explored the biodistribution of FITC

labelled chitosan (MW ~100kDA) injected directly into the peritoneum in a mouse model. They

found that injected chitosan was completely absorbed from the peritoneal cavity in 14hr, and that

there was rapid renal clearance with 25% being excreted 1hr after injection, and 100% 14 hr post

injection (Onishi and Machida 1999). In our case, initial leaking of injected gel into the chest

cavity would be rapidly cleared as described by Onishi and Machida (1999). For hydrogel that

leaks directly into the left ventricle, it would again be carried directly to the waste processing

organs and again cleared quickly (Yamaoka, Tabata, and Ikada 1993). Similarly, hydrogel that

has successfully gelled and localized within the LV is degraded over time. As there is no

exogenous crosslinking providing structure, or creating large masses of chitosan-collagen

composites, degradation of the collagen holding the hydrogel together releases uncrosslinked

chitosan with a maximum MW of 600kDa which would not be expected to accumulate in vivo

and be quickly cleared as waste through the urine.

We further determined that the amount of QHG213H (peptide modified) hydrogel that

localized in the heart and its lifespan in the heart was sufficient to beneficially affect the cardiac

remodeling process post MI. This is evidenced by the improved cardiac functional outcomes

seen with gel injections with the QHREDGS peptide as compared to both the gel alone and the

PBS injected (MI Only) rats 6 weeks post MI (Figure 4-3, Figure 4-4, & Figure 4-5).

Specifically, in terms of cardiac output parameters, the QHG213H (peptide modified) gel

treatment resulted in a 16% improvement in ejection fraction (from 26% to 42%) and an 8%

improvement in fractional shortening (from 23% to 31%) from the MI Only (PBS) treatment

group (Figure 4-3 & Figure 4-4). Moreover, these improvements in cardiac output with

Page 105: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

89

QHG213H gel injection translate into an improvement/maintenance in ejection fraction and

fractional shortening from severely impaired (MI Only) to mildly impaired (QHG213H gel), on a

normal–mild–moderate–severe scale of impairment (R. M. Lang et al. 2006). At 6 weeks post-

MI, we observed a significant decrease in the systolic dimension and volumes but did not find

any difference in the diastolic parameters. This may be because while systolic function decreases

significantly due to CM necrosis post-MI, a longer time frame (e.g. 12 weeks) may be required

to observe changes in diastolic parameters due to matrix remodeling and scar expansion.

In terms of the morphological changes induced by QHG213H gel injection 6 weeks post MI,

we observed a decrease in infarct size and retention of infarct wall thickness with gel injections

relative to the PBS injections, with addition of the QHREDGS peptide to the gel providing the

least amount of adverse remodeling. These morphological improvements in the infarct area could

have been responsible for the increased ejection fraction and fractional shortening with gel

injections relative to the PBS injection. The gel may have stabilized the infarct wall and

increased its thickness by acting as a temporary scaffold that altered the surrounding tissue

properties and reduced wall stress. This is unlikely, however, because the storage modulus of the

Control (no peptide) and QHG213H (peptide modified) gels were quantified to be ~55Pa and

~45Pa respectively, well below the reported stiffness of native rat myocardium (1kPa at the end

of diastole to 140kPa at the end of systole) (Venugopal et al. 2012; Reis et al. 2012). Also, we do

not see a difference in end LV diastolic volume between groups, which would be expected if the

hydrogel were aiding LV mechanics and altering the geometry (Figure 4-4). It is therefore more

probable that the hydrogel affected cardiac function post MI by acting at the cellular level,

possibly by promoting cell survival directly via reducing apoptosis or indirectly via increasing

cell-cell contacts.

Indeed, quantifying the CM presence in the MI zone 6 weeks post injection showed a

significantly higher proportion of CMs in the QHG213H injected group as compared to the

Control (no peptide) gel and MI Only (PBS) groups. While there was no difference in apoptotic

cell numbers in this same region at 6 weeks (Figure 4-8), this is not unexpected as apoptosis

would be expected to level off by this time point since most cell death occurs within the first 72

hr post-MI (Sutton and Sharpe 2000). Interestingly, apoptosis was reduced in the border region

of hearts treated by either Control (no peptide) or QHG213H gel as compared to PBS injection at

6 weeks (Figure 4-8), which might suggest that the gel provided an environment that is more

Page 106: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

90

conducive to cell survival in the long term. Furthermore, the QHG213H gel injection resulted in

larger, mature vessels in the border region at 6 weeks relative to the Control (no peptide) gel and

MI Only (PBS) injection groups (Figure 4-7). This may be because Ang1 angiogenic effects are

mediated through the receptor Tie2, while the peptide QHREDGS is the Ang1 integrin-binding

site(Dallabrida et al. 2005), and may not elicit identical angiogenic responses. Other Ang1-

derived peptides (e.g. Vasculotide) have been demonstrated to bind Tie2 and are angiogenic

(Van Slyke et al. 2009). Moreover, the Ang1-CM interaction is integrin-dependent and Tie2-

independent (Dallabrida et al. 2005); while QHREDGS has been demonstrated to interact with

integrins on endothelial cells (α5β1 and αvβ3) and induced pluripotent stem cells (β1-type) to

promote cell adhesion and survival (Miklas et al. 2013; Dang et al. 2014).It is therefore possible

that the higher CM proportion in the QHG213H injected group could be due to maintained

vascularization, which enabled more cells to acquire the necessary nutrients and oxygen for

survival.

In order to further elucidate the mechanism by which the QHG213H gel injection resulted in

improved cardiac functional and morphometric measures at 6 weeks post MI relative to the PBS

injection, we investigated the effect of treatment on the post MI acute phase (24 hr) wherein

critical apoptotic and immune/inflammatory responses predominate (Figure C-3, Appendix C). A

comparison of the gene expression in the tissue derived from the border zone to that of the MI

zone revealed trend toward increased (2.17-fold) anti-apoptotic BCL2 expression and decreased

(0.13-fold) pro-apoptotic CASP9 expression in the QHG213H gel group relative to the MI Only

(PBS) group. We also observed trend toward increased gene expression of the pro-inflammatory

cytokines IL-6 and IL-1β (4.5- and 2.29-fold, respectively) and decreased (0.43-fold) expression

of PI3K, a pro-survival signaling molecule. However it must be noted that none of these fold

differences were significant, and that the relative contributions of the various signaling pathways

to CM hypertrophy and survival is complex (Nian et al. 2004; Hwang et al. 2001; Fahmi et al.

2013). Hence, while IL-6 is most commonly associated with detrimental post-MI inflammation it

has also been found to promote CM survival in the acute phase of cardiac injury (Fahmi et al.

2013), likely due to the fact that there is a benefit to inflammation in the short term with respect

to repair. Similarly, IL-1β is associated with fibrosis post MI and has classically been viewed as

promoting detrimental post infarct remodeling; however studies in mice have shown that IL-1β

activation is critical in the acute phase of MI and inhibition leads to worse outcomes (Hwang et

Page 107: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

91

al. 2001). These findings highlight the importance of timing in the activation of various

genes/proteins in post infarct inflammation and long term remodeling (Nian et al. 2004; Fahmi et

al. 2013). Notably, the Control gel (no peptide) had an effect on the expression levels of many of

these same genes (Figure C-3, Appendix C), which might explain the intermediate effects we

observed with injection of the gel without peptide on cardiac function and morphology relative to

that of the QHG213H gel and MI Only (PBS) treatment groups. In general, the addition of the

QHREDGS peptide to the gel seems to exacerbate the effects of the gel alone, however in some

instances the effect of the QHG213H gel appears to be distinctive. For example, expression of

the anti-apoptotic gene BCL2 decreased (0.67-fold) in the Control (no peptide) gel treatment

group relative to the MI Only (PBS) group, whereas the QHG213H gel group had increased (>2-

fold) BCL2 expression relative to the MI Only (PBS) group. These results therefore suggest that

the effect on BCL2 expression is dependent upon the QHREDGS peptide.

We have recently shown that the integrin subunits α5, β3, and β1 and the downstream effectors

ILK and MAPK are involved in the QHREDGS-specific pro-survival responses in stem cells and

endothelial cells (Miklas et al. 2013; Dang et al. 2014). While we were unable to detect up-

regulation of these genes at 24 hr (Figure C-3, Appendix C) nor up-regulation of the ILK protein

or of MAPK phosphorylation between the treatment groups, we did observe a significant

increase in MAPK protein expression in the QHG213H gel group compared to either the Control

(no peptide) gel or the MI Only (PBS) groups (Figure C-4, Appendix C). Here again the timing

of the assessment is important as phosphorylation and translation events can occur within

minutes to hours, whereas transcriptional events can occur within hours to days. Thus choosing

an appropriate time point to capture all of these events, while at the same time minimizing the

number of necessary animals, is a daunting task. Herein we did not observe up-

regulation/activation of the genes/proteins of interest, with the exception of MAPK protein;

however we were limited by our assessment at a single time point (24 hr). As such, the fact that

we did not observe differences between the groups is likely more indicative that we did not

capture the up-regulation/activation events in the tissues effectively, rather than that these events

did not occur. An extensive and individualized time course for the events of interest would be

required to fully delineate the mechanism by which the QHG213H gel promotes CM survival in

the post infarct milieu.

Page 108: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

92

Using QHREDGS peptide synthesized to a PEG hydrogel we showed that both CMs and

cardiac fibroblasts bind to QHREDGS through β1-type integrin receptors (Figure 4-9A&B). We

have previously demonstrated that QHREDGS interacts with endothelial cells through α5β1 and

αvβ3 integrins to promote cell survival (Miklas et al. 2013). Moreover, this data might provide

some insight into the in vivo mechanism. Two of the four isoforms of the β1-integrin subunit are

present on the CM surface and are involved in cell-matrix interactions. The β1D isoform is

present in adult myocytes and is important for firm anchoring of the myocyte to the matrix,

which provides a mechanical advantage during contraction. The β1A isoform is present in fetal

myocytes and promotes cell mobility and proliferation at the expense of efficient contractility

(M. Sun 2003). On exposure to cytokines such as TNFα (present in high amounts post MI) the

β1D isoform is replaced by the fetal β1A isoform, which can lead to reduced cardiac function and

detrimental remodeling (M. Sun 2003). β1 is also implicated in cardiac fibroblast anchoring to

the ECM (specifically collagen gels), which through further stabilization of the ECM imparts

beneficial effects to CMs (M. Sun 2003). Taken together, it is possible that the binding of cardiac

cells—both myocytes and non-myocytes—to the QHREDGS peptide through β1-type integrins

after MI leads to stabilization of cell-ECM contacts (protects transition of β1D to β1A in CMs

specifically), inhibiting apoptosis, and modulating genes associated with inflammation in the

acute phase (IL-6 and IL-1β), which results in increased CM survival. This then modifies the

chronic inflammation which would normally occur, resulting in a reduction in pathological

remodeling such that impairment to cardiac function is minimized (Figure 4-10).

Figure 4-10: Proposed QHREDGS mediated mechanism It is suspected that binding of cardiac cells to QHREDGS through β1-integrins leads to up-regulation of anti-

apoptotic BCL2 and reduction in pro-apoptotic CASP9, as well as up-regulation of the inflammatory cytokines

Page 109: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

93

IL-6 and IL-1β. This leads to constructive inflammation in the acute phase and results in early stabilization of the scar and reduced CM loss within the infarct zone, resulting in long term improved cardiac function and reduced detrimental pathological remodeling compared to hydrogel treatment alone or no treatment.

An analysis of animal MI studies performed with treatments ranging from cell sheet

implantation to co-injection of cells with hydrogel post MI, and animal monitoring anywhere

from 4 to 8 weeks post treatment, was performed to see how well the data from our study

compared to that of other groups. Major reported outcomes from these studies are presented in

Table 4-1. The data are presented as a relative change in the outcome of the treatment group to

the control group (in all cases PBS, saline, or media injection) at the end point of the study. For

example, the ejection fraction at 6 weeks post MI in our study was 41.6% for the QHG213H

treatment group, and 26.2% for the MI Only (PBS) control group, giving a 62% increase in the

QHG213H group over the MI Only group at the end of the experiment. In comparison to these

studies, our results are very encouraging in that all our metrics are equivalent to or better than

those reported by the other groups. To properly assess the impact of this work, one must consider

the general simplicity of our treatment compared to others assessed. The mouse embryonic stem

cells delivered in a chitosan-β-glycerol phosphate hydrogel (Lu et al. 2009), the human

embryonic stem cell derived cardiac cells injected in a thymosin β-4 conjugated to PEG hydrogel

(Kraehenbuehl et al. 2011), or the bone marrow stem cells delivered with α-CD/MPEG-PCL-

MPEG synthetic hydrogel (T. Wang, Jiang, et al. 2009) resulted in 22%, 25%, and 77% increases

in ejection fraction respectively. We accomplished a 62% increase in ejection fraction with a

naturally thermogelling hydrogel made from natural components to which a seven amino acid

peptide was immobilized, and without the use of cells. Furthermore, while other hydrogel

formulations (both natural and synthetic) may allow for a higher degree of control of material

properties, the use of exogenous chemical cross-linkers (or harsh cross-linking mechanisms)

raise questions about their safety, whereas the mild, physiologic conditions in which our

formulation gels mimics the natural process of ECM formation in vivo and makes it a safe,

attractive choice for use in the heart as well as to potentially deliver cells in the future

(Nicodemus and Bryant 2008). Finally, the gelling mechanism and gelling time allow for

delivery to the ventricle wall using minimally invasive, simple catheter injection. A full list

describing the studies considered in the comparative-analysis is provided in Table C-2

(Appendix C), and graphical representation of the data is presented in Figure C-5 (Appendix C).

Page 110: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

94

Table 4-1: Comparative-analysis of reported MI studies

%Relative change†: Median MeanStandard

Deviation# of

Groups QHG213H Scar Thickness 67% 69% 22% 7 53% Fractional Scar Area -48% -52% 21% 5 -34% Fractional Shortening 41% 53% 33% 6 36% Ejection Fraction 43% 54% 40% 6 62% *(Wu et al. 2011; Memon et al. 2005; Landa et al. 2008; Tsur-Gang et al. 2009; Christman, Fok, et al. 2004; Dai et al. 2005; Lu et al. 2009; T. Wang, Jiang, et al. 2009; Fujimoto et al. 2009; Kraehenbuehl et al. 2011) †Data is presented here as %relative change in treatment vs control groups at end point of given studies.

4.4. Summary

SA-2 Assess effect of the developed hydrogel in a rat MI model.

This is the first study to evaluate the effect of the angiopoietin-1-derived peptide QHREDGS

on remodelling post-MI. Injection of the developed QHREDGS peptide modified chitosan-

collagen (QHG213H) hydrogel into the LV of rats with acute MI showed significant

improvement in cardiac morphological and functional measurements at 6 weeks post MI relative

to the Control gel (no peptide) or MI Only (PBS) injections. Scar thickness was improved by

53%, fractional scar area reduced by 34%, fractional shortening improved by 36% and ejection

fraction improved by 62% compared to PBS injected hearts. The in vivo lifespan of the hydrogel

was assessed using an on-line technique and localization of the injected hydrogel to the LV wall

of treated hearts was demonstrated. The retention of the hydrogel in situ for up to two weeks was

also demonstrated and removal of the majority of the hydrogel was determined to occur by 3

weeks post injection. The injected QHREDGS-modified chitosan-collagen hydrogel was also

found to maintain a higher proportion of CMs in the MI zone, and of large mature vessels within

the border zone of infarcted hearts. We postulate that binding of cardiac cells to QHREDGS

peptide via β1-type integrin receptors on the cell surface promotes constructive acute

inflammation, inhibiting apoptosis, and attenuates adverse remodeling resulting in improved

cardiac functional outcomes post MI. While effects of Angiopoietin-1 are considered to be

largely Tie2 receptor mediated, the novelty of our work lies in the expanded insight into integrin-

mediated mechanisms by which Angiopoientin-1 derived peptide, QHREDGS, acts on cardiac

Page 111: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

95

cells in vivo. The developed delivery system, consisting of collagen:chitosan hydrogel with

covalently immobilized QHREDGS, could also form a basis for a new therapeutic that is fully

chemically defined and based on biocompatible, biodegradable and inexpensive molecules.

Page 112: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

96

5. Recommendations for future work

This thesis describes the work that has been done towards development of a hydrogel for

cardiac tissue engineering (Specific Aim SA-1), culminating in the publication of Reis et al.

(2012). It continues to describe much of the work done in testing the developed hydrogel in vivo

in a rat model of acute myocardial infarction (Specific Aim SA-2); the original work described in

Reis et al. (2015). Future work must further address SA-2. iii (determining mechanism of action).

5.1. Specific Aim SA-2. i. Show ability to localize in vivo and quantify duration of

its in vivo presence.

Further to the discussion of the in vivo biodistribution of the hydrogel and our data concerning

its presence within the heart one more questions remains: does the peptide modified chitosan

affect any other organs in the body? As seen in the previous discussion, once cleared from the

heart (through degradation of collagen) chitosan and peptide modified chitosan is likely then

processed and rapidly cleared through animals excrements. However, an in depth study of this

has not been done. We have not yet assessed whether addition of the peptide to chitosan makes it

more likely to localize in the kidneys or liver, or even if rapidly cleared does processing it affect

either organ. Recent work in our lab points to the survival promoting activity of QHREDGS

peptide with a number of different cell types (e.g. CMs, FBs, iPSCs, ECs) thus it is likely to

interact with other cells in the body. It is likely that in vitro culture of either liver or kidney cells

in the presence of soluble QHREDGS-chitosan will show if interaction produces any significant

effects, however it would be more interesting to see these effects in vivo in context of post-MI

healing and inflammatory milieu that is present after such insult. Collecting and analyzing these

tissues in tandem with heart tissues during an MI model would provide further insights into the

in vivo biodistribution and life span of our hydrogel.

5.2. Specific Aim SA-2. iii. Determine mechanism of action.

There remain a number of avenues that need to be explored in further determining the

mechanism through which we see improvement after MI with peptide modified hydrogel

treatment. First, the α-integrin sub-unit through which cardiomyocytes attach to QHREDGS

peptide is still undetermined. Similar sub-unit blocking experiments as described in determining

the β-integrin sub-unit should be performed to determine the complement α-sub-unit (many

purified antibodies readily available from BD Biosciences).

Page 113: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

97

Furthermore, the pathway activated through integrin binding to QHREDGS peptide is not

well or fully characterized. Our results point to higher IL-6 and IL-1β with peptide treatment

(neither were statistically significant), although MAPK activation is implicated from our Western

blot analysis. Recent work assessing inflammatory cytokines in the Border Zone tissues excised

24 hours after treatment were similarly inconclusive and suggests two possibilities: (i) the

hydrogel treatments do not elicit a significantly different inflammatory response than PBS

injection after infarct, or (ii) the time at which animals were sacrificed (24 hrs) was not ideal for

detecting those differences. Full understanding of this activation pathway must be determined.

Cardiomyocytes were the cell type largely studied throughout the current work to understand

their interaction with QHREDGS peptide. However, it is known from our in vitro work that

cardiac endothelial cells and various other non-cardiac cells interact positively with QHREDGS

(Miklas et al. 2013; Dang et al. 2014; Feric et al. 2014). In addition, we recently showed cardiac

fibroblasts bind to QHREDGS peptide through β1-integrins and we have little understanding of

the peptides interaction with inflammatory cells such as monocytes and macrophages.

We have not worked with monocytes before; however work has been done by other groups

into the Ang1/Tie-2 system in human monocyte chemotaxis, attachment, and M1/M2

differentiation (Ahmad et al. 2010; Seok et al. 2013). The reported work found that Ang-1

induces chemotaxis of monocytes in a manner that is independent of both Tie-2 and integrin

binding. In addition, they showed that Ang-1 binding to human umbilical endothelial cells was

partially Tie-2 and integrin dependent, whereas Ang-1 binding to monocytes was independent of

both integrins and Tie-2 (Ahmad et al. 2010). In terms of monocyte differentiation they found

that Ang1 is a potent stimulator of p38 and Erk1/2 MAPKs, induces pro-inflammatory monocyte

activation, and that this is Tie-2 independent. The Ang1 mediated stimulation promoted TNF-α

expression as well as a slight increase in IL-6. It was also shown that Ang1 switched macrophage

differentiation toward a pro-inflammatory M1 phenotype, even in the presence of an anti-

inflammatory (pro M2 phenotype) mediator. As we saw slight increases in IL-6 and MAPK in

our system it may suggest that monocytes do indeed attach to QHREDGS peptide, through

integrins or otherwise, and this may promote pro-inflammatory responses. However, none of our

results showed any statistically different expression levels among groups to suggest M1 vs. M2

macrophage activation.

Page 114: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

98

The in vitro work also does not take into account the great amount of cross-talk between

various cell types and the complex immune response post-MI that may negate or promote any

effect the peptide may have on any one of the various cell types present. Basic mechanistic

understanding of the anti-apoptotic effect of QHREDGS peptide on various cell types (e.g.

monocytes, CMs, FBs, ECs) could be done in vitro using one of many ischemia models and up-

regulation of various cytokines assessed, however the problem would remain of how these

cytokines affect the post-MI immune/inflammatory response (Wei et al. 2013; Yue et al. 2000;

Ma et al. 2006). This can only be assessed with large scale in vivo models, which themselves

pose many issues. In the simplest case, where our tissue collection time points were not ideal to

assess effect, a large study would be required to sacrifice many animals at many points during

the acute phase after MI (first few days) to better understand the up/down-regulation and

expression of the many proteins involved and find statistical significance. If our original

hypothesized mechanism is true then understanding the exact roles IL6 or IL1β play in terms of

cell recruitment and what those infiltrating cells do post-MI becomes more complex. Both the

control case of MI Only (PBS injection) and the treatment (peptide modified gel injection) elicit

an immune/inflammatory response, as does the MI surgery itself. The classic models of using

knock-out animals or antibodies against specific proteins will not work when both the control

and treatment case elicit similar responses. For macrophage infiltration to MI zone and

assessment of M1/M2 phenotype may require autologus intravenous injections of labelled cells

and tracking outcomes.

Page 115: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

99

6. Conclusions & Contributions to New Knowledge

As discussed previously, the issue of cardiac disease and the need for clinically translatable

treatment options is one of the most pressing health concerns of developed society. Post MI

pathological cardiac remodeling activities include infarct expansion, ventricular dilation, and

scar tissue formation. While necessary for LV stabilization and immediate survival, these

activities are very disconcerting in the short and long term. Overall, the dilation of the ventricle

has been shown to directly decrease patients’ survival and may ultimately lead to congestive

heart failure. The extent of remodeling is affected by the size of the infarct, how much the infarct

heals, and ventricular wall stresses.

There are routine clinical treatments which attempt to prevent the pathological remodeling

that occurs post-infarction. These methods include both pharmacological methods that aim to

inhibit remodeling pathways, as well as targeting the cause of MI (blocked or partially blocked

coronary arteries). Reperfusion of blocked arteries (through methods such as thrombolysis and

angioplastys) to restore blood flow to the infarcted myocardium is standard procedure which has

been shown to have beneficial effects including limiting detrimental remodeling and partially

healing the myocardium (Sutton and Sharpe 2000; Touchstone et al. 1989; Marino, Zanolla, and

Zardini 1989; Baks et al. 2006). While promising, reperfusion is not without issues as the

mechanism of repair is not fully understood and restoring blood flow to injured cells can lead to

further damage and have severe detrimental effects (Verma et al. 2002). Pharmacological

methods used to treat MI are only partially effective and there are many questions about the

safety of long term use (Sutton and Sharpe 2000; Beltrame 2008; McMurray et al. 2006;

Braunwald et al. 2004), even though they have shown effectiveness in the short term.

Currently, no methods to completely prevent these pathological events exist, however

researchers all over the world are exploring approaches and treatment options that can stabilize

the scar faster, reduce the expansion of the infarct, and decrease ventricular wall stresses that

lead to further detrimental remodeling events. Left ventricular assist devices have improved

greatly over the years and are increasingly used as a bridge to transplantation, and some have

been approved as a destination therapy, providing individuals ineligible for a heart transplant

with a long-term therapy option (Wilson et al. 2009; Kirklin et al. 2013). However this is not the

most desirable option and it would be preferred to prevent heart failure altogether, thus

Page 116: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

100

treatments that limit the remodeling that occurs post-infarction will improve the survival and

quality of life of MI patients (Pfeffer and Braunwald 1990).

To address these issues we sought to utilize the novel Ang1 derived peptide QHREDGS along

with two common biomaterials, chitosan and collagen, to create a novel treatment option for MI.

Chitosan and collagen have been used before in the treatment of cardiac dysfunction and tissue

engineering as whole (Lu et al. 2009; Rask, Dallabrida, et al. 2010; Garbern et al. 2011;

Suuronen et al. 2006; Y. Zhang et al. 2008; Wu et al. 2011). However, typical formulations of

chitosan-collagen hydrogels use some sort of crosslinking mechanism to speed gelation, improve

mechanical integrity, and perhaps make them more amenable for use in vivo. Our first success in

this project was demonstrating that combinations of the two components could form

mechanically stable hydrogels, without the use of exogenous chemical crosslinkers, suitable for

CM cell culture, and that specifically a 2.5 mg/mL 1:1 m:m ratio was optimal for culturing

cardiac cells. Furthermore, the functional groups on both chitosan and collagen, as well as their

ionic nature, allows for not only conjugation of various molecules (e.g. QHREDGS) but also

release of small molecules in a drug delivery system. Work concurrent to this showed that a 2:1

collagen to chitosan ratio was optimal for the controlled release of Tβ4 and suitable for the

endothelial and smooth muscle cell growth into vascular structures from epicardial explants

when cultured in the Tβ4 containing gel (Chiu and Radisic 2011; Chiu, Montgomery, et al.

2012).

We were subsequently able to demonstrate that the hydrogel was able to localize at the site of

injection in the LV of live animals and track its in vivo biodistribution on line to show its lifespan

in the heart to be 2-3 weeks. While many would dismiss the relatively low mechanical properties

of our gel (in terms of elastic storage modulus) as being irrelevant for use in the heart we were

successful in demonstrating otherwise. To our knowledge this is the first time a non-exogenously

cross-linked chitosan-collagen blend has been used in the heart. As mentioned previously

varying the ratio of chitosan and collagen also allows for a level of tailoring of the mechanical

and ionic properties of the gels for use in other applications.

The most important findings of this work, however, had to do with the potential of

QHREDGS peptide in treating acute MI as well as furthering the understanding of the underlying

mechanisms through which it acts. Our in vitro work clearly demonstrated stable conjugation of

QHREDGS to chitosan provided long-term signaling as well as measurable improvements in CM

Page 117: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

101

performance when encapsulated in peptide modified chitosan-collagen hydrogels. We showed

that CM attachment to QHREDGS is indeed integrin dependant and implicated the β1-integrin

subunit as being integral to attachment and signaling. Our in vivo results clearly demonstrate the

potential of this novel hydrogel in treating acute MI to maintain higher levels of cardiac function

and reduce pathological remodeling. A major contribution of this work is in the understanding of

how both chitosan-collagen hydrogel and peptide-modified hydrogel succeed in producing these

post-MI benefits, in terms of maintaining higher levels of CMs within the developed scar and

reducing scar expansion. From our results it would seem injection of either gel does not induce a

significantly different immune response from PBS injection post-MI, but does result in better

cardiac outcomes. Furthermore, we were able to indentify some trends in inflammatory cytokine

expression that warrants further investigation and raises interesting observations about cell

recruitment and survival mechanisms in the acute phase after infarct.

In looking forward, this work has provided the basis for a novel MI treatment and opened up

new research avenues into how the heart responds to gel treatment after infarct, peptide

immobilization and the long-term impacts of cell exposure to such compounds, and whether this

formulation can have similar successes in larger animal models – leading to the question can it

be successful clinically?

6.1. Publications & Contributions

a. Journal articles

Xiao Y., Reis L.A., Zhao Y., & Radisic M. (2015). Modifications of biomaterials with

immobilized growth factors or peptides for tissue engineering applications. Methods.

Submitted.

Zhang B., Montgomery M., Chamberlain M.D., Wells L.A., Pahnke A., Massé S., Kim J.,

Reis L.A., Abdulah M., Nunes S.S., Nanthakumar K., Sefton M.V., & Radisic M.

(2014). AngioChip: a biodegradable scaffold with built-in vasculature for organ-on-a-

chip engineering and direct surgical anastomosis. Nature Materials. Under Review.

Reis L.A., Chiu L.L.Y., Wu J., Feric N., Momen A., Li R.K., & Radisic M. (2015).

Hydrogels with integrin-binding angiopoietin-1-derived peptide, QHREDGS, for

treatment of acute myocardial infarction. Circulation: Heart Failure. DOI:

10.1161/CIRCHEARTFAILURE.114.001881.

Page 118: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

102

Reis L.A., Chiu L.L.Y., Feric N., Fu L., & Radisic M. (2014). Biomaterials in myocardial

tissue engineering. Journal of Tissue Engineering & Regenerative Medicine. DOI:

10.1002/term.1944.

Miklas J.W., Nunes S.S., Sofla A., Reis L.A., Pahnke A., Xiao Yun., Laschinger C., &

Radisic M. (2014). Bioreactor for modulation of cardiac microtissue phenotype by

combined static stretch and electrical stimulation. Biofabrication. 6: 024113.

Miklas J.W., Dallabrida S.M., Reis L.A., Ismail N., Rupnik M., & Radisic M. (2013).

QHREDGS enhances tube formation, metabolism, and survival of endothelial cells in

collagen-chitosan hydrogels. PLOS One. 8(8): e72956.

Chiu L.L.Y., Reis L.A., Momen A., & Radisic M. (2012). Controlled release of thymosin

β4 from injected collagen-chitosan hydrogels promotes angiogenesis and prevents

tissue loss after myocardial infarction. Regenerative Medicine. 7(4): 523-33.

Chiu L.L.Y., Iyer R.K., Reis L.A., Nunes S.S., & Radisic M. (2012). Cardiac Tissue

Engineering: Current State and Perspectives. Frontiers in Bioscience. 17: 1533-1550.

Reis L.A., Chiu L.L.Y., Liang Y., Huynh K., & Radisic M. (2011). A peptide-modified

chitosan-collagen hydrogel for cardiac cell culture and delivery. Acta Biomaterilia.

8(3): 1022-36.

Iyer R.K., Chiu L.L.Y., Reis L.A., & Radisic M. (2011). Engineered cardiac tissues.

Current Opinion in Biotechnology. 22(5): 706-714.

Rask F., Mihic A., Reis L. A., Dallabrida S., Ismail N., Sider K., Simmons C., Weisel R.,

Li R.K., & Radisic M. (2010). Hydrogels modified with QHREDGS peptide support

heart cell survival in vitro and after sub-cutaneous implantation. Soft Matter. 6: 5089-

99.

b. Book chapters

Reis L.A., Chiu L.L.Y., Feric N., Fu L., & Radisic M. (January 2014). Injectable

biomaterials for cardiac repair. In Li R.K. & Weisel R.D. (EDs), Cardiac

Regeneration & Repair (Vol. II, Ch. 3). Cambridge: Woodhead Publishing.

Audet J., Clarke G., Lee W., Ma J., Onishi K., Reis L.A., & Zandstra P. (2013).

Combining in silico and in vitro techniques to engineer pluripotent stem cell fate. In

Micou M.K. & Kilkenny D. (EDs.), A Laboratory Course in Tissue Engineering (1st

ed., p. 173-196). Boca Raton: CRC Press.

Page 119: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

103

Odedra D., Chiu L.L.Y., Reis L.A., Rask F., Chiang K., Radisic M. (2011). Cardiac Tissue

Engineering. In J. A. Burdick & R. L. Mauck (EDs.), Biomaterials for Tissue

Engineering Applications: A review of past & future trends (1st ed., p. 562). Vienna:

Springer Vienna.

c. Oral presentations

Reis L.A., Chiu L.L.Y., Wu J., Feric N., Momen A., Li R.K., & Radisic M. (May 2014). A

peptide modified hydrogel therapy for acute myocardial infarction. 2014 Annual

University of Toronto Institute of Biomaterials & Biomedical Engineering Scientific

Day, Toronto, ON, CAN.

Reis, L.A., Chiu, L.L.Y., Wu, J., Momen A., Li, R.K., & Radisic, M. (Oct. 2012). A

peptide modified hydrogel for cardiac tissue preservation after acute myocardial

infarction. 2012 Biomedical Engineering Society Annual Meeting, Atlanta, GA, USA.

Reis, L.A., Chiu, L.L.Y., Wu, J., Momen A., Li, R.K., & Radisic, M. (April 2012). A

peptide modified hydrogel for cardiac regeneration. 5th Annual Regenerative

Medicine Symposium, Toronto, ON, CAN.

d. Poster presentations

Reis L.A., Chiu L.L.Y., Wu J., Momen A., Li R.K., & Radisic M. (May 2013). A peptide

modified hydrogel for treatment after acute myocardial infarction. 2013 Canadian

Biomaterials Society Annual Meeting, Ottawa, ON, CAN.

Reis L.A., Chiu L.L.Y., Wu J., Momen A., Li R.K., & Radisic M. (September 2012). A

peptide modified hydrogel for cardiac tissue preservation after acute myocardial

infarction. An Afternoon of Engineering Innovation, Toronto, ON, CAN.

Reis L.A., Chiu L.L.Y., Wu J., Momen A., Li R.K., & Radisic M. (June 2012). A peptide

modified hydrogel for cardiac regeneration. CIHR 2012 Canadian Student Health

Research Forum, Winnipeg, MB, CAN.

Reis L.A., Chiu L.L.Y., Wu J., Momen A., Li R.K., & Radisic M. (May 2012). A peptide

modified hydrogel to reduce long term effect of acute myocardial infarction. 2012

Annual University of Toronto Institute of Biomaterials & Biomedical Engineering

Scientific Day, Toronto, ON, CAN.

Reis L.A., Chiu L.L.Y., Liang Y., Huynh K., Momen A., & Radisic M. (May 2011). A

peptide modified collagen-chitosan hydrogel for the site-specific delivery of

Page 120: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

104

cardiomyocytes to the heart. 2011 Annual University of Toronto Institute of

Biomaterials & Biomedical Engineering Scientific Day, Toronto, ON, CAN.

Reis L.A., Huynh K., Momen A., & Radisic M. (June 2010). Development of a peptide

modified collagen-chitosan hydrogel for the site-specific delivery of cardiomyocytes to

the heart. 2010 Canadian Biomaterials Society Annual Meeting, Kingston, ON, CAN.

Page 121: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

105

7. References

Ahmad, Shakil, Melissa J Cudmore, Keqing Wang, Peter Hewett, Rahul Potluri, Takeshi Fujisawa, and Asif Ahmed. 2010. “Angiopoietin-1 Induces Migration of Monocytes in a Tie-2 and Integrin-Independent Manner.” Hypertension 56 (3) (September): 477–83. doi:10.1161/HYPERTENSIONAHA.110.155556. http://www.ncbi.nlm.nih.gov/pubmed/20696992.

Baks, Timo, Robert-Jan van Geuns, Elena Biagini, Piotr Wielopolski, Nico R Mollet, Filippo Cademartiri, Willem J van der Giessen, et al. 2006. “Effects of Primary Angioplasty for Acute Myocardial Infarction on Early and Late Infarct Size and Left Ventricular Wall Characteristics.” Journal of the American College of Cardiology 47 (1) (January 3): 40–4. doi:10.1016/j.jacc.2005.09.008. http://www.ncbi.nlm.nih.gov/pubmed/16386662.

Balasubramanian, Sundaravadivel, and Dhandapani Kuppuswamy. 2003. “RGD-Containing Peptides Activate S6K1 through beta3 Integrin in Adult Cardiac Muscle Cells.” The Journal of Biological Chemistry 278 (43) (October): 42214–24. doi:10.1074/jbc.M303428200. http://www.ncbi.nlm.nih.gov/pubmed/12909616.

Beltrame, John F. 2008. “Nitrate Therapy in Acute Myocardial Infarction: Potion or Poison?” Cardiovascular Drugs and Therapy / Sponsored by the International Society of Cardiovascular Pharmacotherapy 22 (3) (June): 165–8. doi:10.1007/s10557-008-6103-1. http://www.ncbi.nlm.nih.gov/pubmed/18347964.

Beltrami, Antonio P, Laura Barlucchi, Daniele Torella, Mathue Baker, Federica Limana, Stefano Chimenti, Hideko Kasahara, et al. 2003. “Adult Cardiac Stem Cells Are Multipotent and Support Myocardial Regeneration.” Cell 114 (6) (September): 763–76. http://www.ncbi.nlm.nih.gov/pubmed/14505575.

Berger, J, M Reist, J.M Mayer, O Felt, and R Gurny. 2004. “Structure and Interactions in Chitosan Hydrogels Formed by Complexation or Aggregation for Biomedical Applications.” European Journal of Pharmaceutics and Biopharmaceutics 57 (1) (January): 35–52. doi:10.1016/S0939-6411(03)00160-7. http://linkinghub.elsevier.com/retrieve/pii/S0939641103001607.

Bhana, Bashir, Rohin K Iyer, Wen Li Kelly Chen, Ruogang Zhao, Krista L Sider, Morakot Likhitpanichkul, Craig A Simmons, and Milica Radisic. 2010. “Influence of Substrate Stiffness on the Phenotype of Heart Cells.” Biotechnology and Bioengineering 105 (6) (April 15): 1148–60. doi:10.1002/bit.22647. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=20014437.

Bittira, Bindu, Jin-Qiang Kuang, Abdulaziz Al-Khaldi, Dominique Shum-Tim, and Ray C J Chiu. 2002. “In Vitro Preprogramming of Marrow Stromal Cells for Myocardial Regeneration.” The Annals of Thoracic Surgery 74 (4) (October): 1154–9; discussion 1159–60. http://www.ncbi.nlm.nih.gov/pubmed/12400761.

Page 122: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

106

Blackburn, Nick J.R., Tanja Sofrenovic, Drew Kuraitis, Ali Ahmadi, Brian McNeill, Chao Deng, Katey J. Rayner, Zhiyuan Zhong, Marc Ruel, and Erik J. Suuronen. 2014. “Timing Underpins the Benefits Associated with Injectable Collagen Biomaterial Therapy for the Treatment of Myocardial Infarction.” Biomaterials (November): 1–11. doi:10.1016/j.biomaterials.2014.11.004. http://linkinghub.elsevier.com/retrieve/pii/S0142961214011600.

Boateng, Samuel Y, Syed S Lateef, William Mosley, Thomas J Hartman, Luke Hanley, and Brenda Russell. 2005. “RGD and YIGSR Synthetic Peptides Facilitate Cellular Adhesion Identical to that of Laminin and Fibronectin but Alter the Physiology of Neonatal Cardiac Myocytes.” American Journal of Physiology. Cell Physiology 288 (1) (January): C30–8. doi:10.1152/ajpcell.00199.2004. http://www.ncbi.nlm.nih.gov/pubmed/15371257.

Bock-Marquette, Ildiko, Ankur Saxena, Michael D White, J Michael Dimaio, and Deepak Srivastava. 2004. “Thymosin beta4 Activates Integrin-Linked Kinase and Promotes Cardiac Cell Migration, Survival and Cardiac Repair.” Nature 432 (7016) (November): 466–72. doi:10.1038/nature03000. http://www.ncbi.nlm.nih.gov/pubmed/15565145.

Bouten, C V C, P Y W Dankers, a Driessen-Mol, S Pedron, a M a Brizard, and F P T Baaijens. 2011. “Substrates for Cardiovascular Tissue Engineering.” Advanced Drug Delivery Reviews 63 (4-5) (April 30): 221–41. doi:10.1016/j.addr.2011.01.007. http://www.ncbi.nlm.nih.gov/pubmed/21277921.

Braunwald, Eugene, Michael J Domanski, Sarah E Fowler, Nancy L Geller, Bernard J Gersh, Judith Hsia, Marc a Pfeffer, Madeline M Rice, Yves D Rosenberg, and Jean L Rouleau. 2004. “Angiotensin-Converting-Enzyme Inhibition in Stable Coronary Artery Disease.” The New England Journal of Medicine 351 (20) (November 11): 2058–68. doi:10.1056/NEJMoa042739. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2556374&tool=pmcentrez&rendertype=abstract.

Cai, Chen-Leng, Xingqun Liang, Yunqing Shi, Po-Hsien Chu, Samuel L Pfaff, Ju Chen, and Sylvia Evans. 2003. “Isl1 Identifies a Cardiac Progenitor Population That Proliferates prior to Differentiation and Contributes a Majority of Cells to the Heart.” Developmental Cell 5 (6) (December): 877–89. http://www.ncbi.nlm.nih.gov/pubmed/14667410.

Camenisch, Gieri, Maria Teresa Pisabarro, Daniel Sherman, Joe Kowalski, Mark Nagel, Phil Hass, Ming-Hong Xie, et al. 2002. “ANGPTL3 Stimulates Endothelial Cell Adhesion and Migration via Integrin Alpha Vbeta 3 and Induces Blood Vessel Formation in Vivo.” The Journal of Biological Chemistry 277 (19) (May 10): 17281–90. doi:10.1074/jbc.M109768200. http://www.ncbi.nlm.nih.gov/pubmed/11877390.

Carlson, T R, Y Feng, P C Maisonpierre, M Mrksich, and a O Morla. 2001. “Direct Cell Adhesion to the Angiopoietins Mediated by Integrins.” The Journal of Biological Chemistry 276 (28) (July 13): 26516–25. doi:10.1074/jbc.M100282200. http://www.ncbi.nlm.nih.gov/pubmed/11346644.

Page 123: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

107

Chen, Hongli, Han Chen, Lingrong Liu, Ping Yuan, and Qiqing Zhang. 2008. “The Study of Improved Controlled Release of Vincristine Sulfate from Collagen-Chitosan Complex Film.” Artificial Cells, Blood Substitutes, and Immobilization Biotechnology 36 (4) (January): 372–85. doi:10.1080/10731190802239057. http://www.ncbi.nlm.nih.gov/pubmed/18649172.

Chen, Qi Zhi, Sian E. Harding, Nadire N. Ali, Alexander R. Lyon, and Aldo R. Boccaccini. 2008. “Biomaterials in Cardiac Tissue Engineering: Ten Years of Research Survey.” Materials Science and Engineering: R: Reports 59 (1-6) (February): 1–37. doi:10.1016/j.mser.2007.08.001. http://linkinghub.elsevier.com/retrieve/pii/S0927796X07001118.

Chenite, a, C Chaput, D Wang, C Combes, M D Buschmann, C D Hoemann, J C Leroux, B L Atkinson, F Binette, and a Selmani. 2000. “Novel Injectable Neutral Solutions of Chitosan Form Biodegradable Gels in Situ.” Biomaterials 21 (21) (November): 2155–61. http://www.ncbi.nlm.nih.gov/pubmed/10985488.

Chiu, Loraine L Y, Rohin K Iyer, Lewis A Reis, Sara S Nunes, and M Radisic. 2012. “Cardiac Tissue Engineering: Current State and Perspectives.” Frontiers in Bioscience 17 (1): 1533–1550. doi:10.2741/4002. http://www.bioscience.org/2012/v17/af/4002/list.htm.

Chiu, Loraine L Y, Miles Montgomery, Yan Liang, Haijiao Liu, and Milica Radisic. 2012. “Perfusable Branching Microvessel Bed for Vascularization of Engineered Tissues.” Proceedings of the National Academy of Sciences of the United States of America 109 (50) (December 11): E3414–23. doi:10.1073/pnas.1210580109. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3528595&tool=pmcentrez&rendertype=abstract.

Chiu, Loraine L Y, and Milica Radisic. 2010. “Scaffolds with Covalently Immobilized VEGF and Angiopoietin-1 for Vascularization of Engineered Tissues.” Biomaterials 31 (2) (January): 226–41. doi:10.1016/j.biomaterials.2009.09.039. http://www.ncbi.nlm.nih.gov/pubmed/19800684.

———. 2011. “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 155 (3) (November 7): 376–85. doi:10.1016/j.jconrel.2011.05.026. http://www.ncbi.nlm.nih.gov/pubmed/21663777.

Christman, Karen L, Qizhi Fang, Michael S Yee, Kandice R Johnson, Richard E Sievers, and Randall J Lee. 2005. “Enhanced Neovasculature Formation in Ischemic Myocardium Following Delivery of Pleiotrophin Plasmid in a Biopolymer.” Biomaterials 26 (10) (April): 1139–44. doi:10.1016/j.biomaterials.2004.04.025. http://www.ncbi.nlm.nih.gov/pubmed/15451633.

Christman, Karen L, Hubert H Fok, Richard E Sievers, Qizhi Fang, and Randall J Lee. 2004. “Fibrin Glue Alone and Skeletal Myoblasts in a Fibrin Scaffold Preserve Cardiac Function

Page 124: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

108

after Myocardial Infarction.” Tissue Engineering 10 (3-4): 403–9. doi:10.1089/107632704323061762. http://www.ncbi.nlm.nih.gov/pubmed/15165457.

Christman, Karen L, Andrew J Vardanian, Qizhi Fang, Richard E Sievers, Hubert H Fok, and Randall J Lee. 2004. “Injectable Fibrin Scaffold Improves Cell Transplant Survival, Reduces Infarct Expansion, and Induces Neovasculature Formation in Ischemic Myocardium.” Journal of the American College of Cardiology 44 (3) (August): 654–60. doi:10.1016/j.jacc.2004.04.040. http://www.ncbi.nlm.nih.gov/pubmed/15358036.

Crockford, David. 2007. “Development of Thymosin beta4 for Treatment of Patients with Ischemic Heart Disease.” Annals of the New York Academy of Sciences 1112 (September): 385–95. doi:10.1196/annals.1415.051. http://www.ncbi.nlm.nih.gov/pubmed/17947592.

Dahl, Shannon L M, Alan P Kypson, Jeffrey H Lawson, Juliana L Blum, Justin T Strader, Yuling Li, Roberto J Manson, et al. 2011. “Readily Available Tissue-Engineered Vascular Grafts.” Science Translational Medicine 3 (68) (February 2): 68ra9. doi:10.1126/scitranslmed.3001426. http://www.ncbi.nlm.nih.gov/pubmed/21289273.

Dai, Wangde, Loren E Wold, Joan S Dow, and Robert a Kloner. 2005. “Thickening of the Infarcted Wall by Collagen Injection Improves Left Ventricular Function in Rats: A Novel Approach to Preserve Cardiac Function after Myocardial Infarction.” Journal of the American College of Cardiology 46 (4): 714–9. doi:10.1016/j.jacc.2005.04.056. http://www.ncbi.nlm.nih.gov/pubmed/16098441.

Dallabrida, Susan M, Nesreen Ismail, Julianne R Oberle, Blanca E Himes, and Maria A Rupnick. 2005. “Angiopoietin-1 Promotes Cardiac and Skeletal Myocyte Survival through Integrins.” Circulation Research 96 (4) (March): e8–24. doi:10.1161/01.RES.0000158285.57191.60. http://www.ncbi.nlm.nih.gov/pubmed/15692086.

Dang, Lan T H, Nicole T Feric, Carol Laschinger, Wing Y Chang, Boyang Zhang, Geoffrey A Wood, William L Stanford, and Milica Radisic. 2014. “Inhibition of Apoptosis in Human Induced Pluripotent Stem Cells during Expansion in a Defined Culture Using Angiopoietin-1 Derived Peptide QHREDGS.” Biomaterials 35 (27) (September 12): 7786–99. doi:10.1016/j.biomaterials.2014.05.018. http://www.ncbi.nlm.nih.gov/pubmed/24930852.

Das, Debarun, Ziyang Zhang, Thomas Winkler, Meenakshi Mour, Christina I Günter, Michael M Morlock, Hans-günther Machens, and Arndt F Schilling. 2012. “Bioresorption and Degradation of Biomaterials.” Advances in Biochemical Engineering/biotechnology (126) (October 6): 317–333. doi:10.1007/10_2011_119. http://www.ncbi.nlm.nih.gov/pubmed/21975956.

Davis, Michael E, Patrick C H Hsieh, Tomosaburo Takahashi, Qing Song, Shuguang Zhang, Roger D Kamm, Alan J Grodzinsky, Piero Anversa, and Richard T Lee. 2006. “Local Myocardial Insulin-like Growth Factor 1 (IGF-1) Delivery with Biotinylated Peptide Nanofibers Improves Cell Therapy for Myocardial Infarction.” Proceedings of the National

Page 125: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

109

Academy of Sciences of the United States of America 103 (21) (May): 8155–60. doi:10.1073/pnas.0602877103. http://www.ncbi.nlm.nih.gov/pubmed/16698918.

Davis, Michael E, J P Michael Motion, Daria a Narmoneva, Tomosaburo Takahashi, Daihiko Hakuno, Roger D Kamm, Shuguang Zhang, and Richard T Lee. 2005. “Injectable Self-Assembling Peptide Nanofibers Create Intramyocardial Microenvironments for Endothelial Cells.” Circulation 111 (4): 442–50. doi:10.1161/01.CIR.0000153847.47301.80. http://www.ncbi.nlm.nih.gov/pubmed/15687132.

Davis, S, T H Aldrich, P F Jones, a Acheson, D L Compton, V Jain, T E Ryan, et al. 1996. “Isolation of Angiopoietin-1, a Ligand for the TIE2 Receptor, by Secretion-Trap Expression Cloning.” Cell 87 (7) (December 27): 1161–9. http://www.ncbi.nlm.nih.gov/pubmed/8980223.

Deforest, Cole a, Evan a Sims, and Kristi S Anseth. 2010. “Peptide-Functionalized Click Hydrogels with Independently Tunable Mechanics and Chemical Functionality for 3D Cell Culture.” Chemistry of Materials : A Publication of the American Chemical Society 22 (16) (August 24): 4783–4790. doi:10.1021/cm101391y. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2937999&tool=pmcentrez&rendertype=abstract.

Deng, Chao, Pingchuan Zhang, Branka Vulesevic, Drew Kuraitis, Fengfu Li, Ann Fook Yang, May Griffith, Marc Ruel, and Erik J Suuronen. 2010. “A Collagen-Chitosan Hydrogel for Endothelial Differentiation and Angiogenesis( * ).” Tissue Engineering. Part A 16 (10) (June): 3099–3109. doi:10.1089/ten.tea.2009.0504. http://www.ncbi.nlm.nih.gov/pubmed/20586613.

Dengler, Jana. 2009. “An in Vitro Model System for Cardiac Cell Therapy.” University of Toronto.

DeQuach, J A, V Mezzano, A Miglani, S Lange, G M Keller, F Sheikh, and K L Christman. 2010. “Simple and High Yielding Method for Preparing Tissue Specific Extracellular Matrix Coatings for Cell Culture.” PLoS One 5: e13039 ST – Simple and high yielding method for p. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=20885963.

Deten, Alexander, Hans Christian Volz, Sören Clamors, Sabine Leiblein, Wilfried Briest, Grit Marx, and Heinz-Gerd Zimmer. 2005. “Hematopoietic Stem Cells Do Not Repair the Infarcted Mouse Heart.” Cardiovascular Research 65 (1) (January): 52–63. doi:10.1016/j.cardiores.2004.11.009. http://www.ncbi.nlm.nih.gov/pubmed/15621033.

Dobner, Stephan, Deon Bezuidenhout, Padmini Govender, Peter Zilla, and Neil Davies. 2009. “A Synthetic Non-Degradable Polyethylene Glycol Hydrogel Retards Adverse Post-Infarct Left Ventricular Remodeling.” Journal of Cardiac Failure 15 (7) (September): 629–36. doi:10.1016/j.cardfail.2009.03.003. http://www.ncbi.nlm.nih.gov/pubmed/19700140.

Page 126: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

110

Duan, Yi, Zen Liu, John O’Neill, Leo Q. Wan, Donald O. Freytes, and Gordana Vunjak-Novakovic. 2011. “Hybrid Gel Composed of Native Heart Matrix and Collagen Induces Cardiac Differentiation of Human Embryonic Stem Cells without Supplemental Growth Factors.” Journal of Cardiovascular Translational Research 4 (5) (July 9): 605–615. doi:10.1007/s12265-011-9304-0. http://www.ncbi.nlm.nih.gov/pubmed/21744185.

Elisseeff, J, K Anseth, D Sims, W McIntosh, M Randolph, and R Langer. 1999. “Transdermal Photopolymerization for Minimally Invasive Implantation.” Proceedings of the National Academy of Sciences of the United States of America 96 (6) (March 16): 3104–7. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=15902&tool=pmcentrez&rendertype=abstract.

Eschenhagen, Thomas, Christine Fink, Ute Remmers, Hasso Scholz, Jens Wattchow, Joachim Weil, Wolfram Hubertus Zimmermann, et al. 1997. “Three-Dimensional Reconstitution of Embryonic Cardiomyocytes in a Collagen Matrix: A New Heart Muscle Model System.” The FASEB Journal : Official Publication of the Federation of American Societies for Experimental Biology 11 (8) (July): 683–94. http://www.ncbi.nlm.nih.gov/pubmed/9240969.

Fahmi, Ahmed, Nicola Smart, Anu Punn, Rita Jabr, Michael Marber, and Richard Heads. 2013. “p42/p44-MAPK and PI3K Are Sufficient for IL-6 Family cytokines/gp130 to Signal to Hypertrophy and Survival in Cardiomyocytes in the Absence of JAK/STAT Activation.” Cellular Signalling 25 (4) (April): 898–909. doi:10.1016/j.cellsig.2012.12.008. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3627957&tool=pmcentrez&rendertype=abstract.

Fan, Vivian H, Kenichi Tamama, Ada Au, Romie Littrell, Llewellyn B Richardson, John W Wright, Alan Wells, and Linda G Griffith. 2007. “Tethered Epidermal Growth Factor Provides a Survival Advantage to Mesenchymal Stem Cells.” Stem Cells (Dayton, Ohio) 25 (5) (May): 1241–51. doi:10.1634/stemcells.2006-0320. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17234993.

Feric, Nicole, Calvin Cheng, M. Cynthia Goh, Vyacheslav Dudnyk, Val Di Tizio, and Milica Radisic. 2014. “Angiopoeitin-1 Derived Peptide QHREDGS Promotes Osteoblast Differentiation, Bone Matrix Deposition and Mineralization on Biomedical Materials.” Biomaterials Science. doi:10.1039/c4bm00073k.

Fujimoto, Kazuro L, Zuwei Ma, Devin M Nelson, Ryotaro Hashizume, Jianjun Guan, Kimimasa Tobita, and William R Wagner. 2009. “Synthesis, Characterization and Therapeutic Efficacy of a Biodegradable, Thermoresponsive Hydrogel Designed for Application in Chronic Infarcted Myocardium.” Biomaterials 30 (26): 4357–68. doi:10.1016/j.biomaterials.2009.04.055. http://www.ncbi.nlm.nih.gov/pubmed/19487021.

Fujita, Masanori, Masayuki Ishihara, Yuji Morimoto, Masafumi Simizu, Yoshio Saito, Hirofumi Yura, Takemi Matsui, et al. 2005. “Efficacy of Photocrosslinkable Chitosan Hydrogel

Page 127: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

111

Containing Fibroblast Growth Factor-2 in a Rabbit Model of Chronic Myocardial Infarction.” The Journal of Surgical Research 126 (1): 27–33. doi:10.1016/j.jss.2004.12.025. http://www.ncbi.nlm.nih.gov/pubmed/15916971.

Fujita, Masanori, Masayuki Ishihara, Masafumi Simizu, Kiyohaya Obara, Toshiaki Ishizuka, Yoshio Saito, Hirofumi Yura, et al. 2004. “Vascularization in Vivo Caused by the Controlled Release of Fibroblast Growth Factor-2 from an Injectable Chitosan/non-Anticoagulant Heparin Hydrogel.” Biomaterials 25 (4) (February): 699–706. doi:10.1016/S0142-9612(03)00557-X. http://linkinghub.elsevier.com/retrieve/pii/S014296120300557X.

Garbern, Jessica C, Elina Minami, Patrick S Stayton, and Charles E Murry. 2011. “Delivery of Basic Fibroblast Growth Factor with a pH-Responsive, Injectable Hydrogel to Improve Angiogenesis in Infarcted Myocardium.” Biomaterials 32 (9) (March): 2407–16. doi:10.1016/j.biomaterials.2010.11.075. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3026890&tool=pmcentrez&rendertype=abstract.

Go, Alan S, Dariush Mozaffarian, Véronique L Roger, Emelia J Benjamin, Jarett D Berry, William B Borden, Dawn M Bravata, et al. 2013. “Heart Disease and Stroke Statistics--2013 Update: A Report from the American Heart Association.” Circulation 127 (1) (January 1): e6–e245. doi:10.1161/CIR.0b013e31828124ad. http://www.ncbi.nlm.nih.gov/pubmed/23239837.

Graber, Mark L., Douglas C. DiLillo, Bradford L. Friedman, and Enrique Pastoriza-Munoz. 1986. “Characteristics of Fluoroprobes for Measuring Intracellular pH.” Analytical Biochemistry 156 (1) (July): 202–212. doi:10.1016/0003-2697(86)90174-0. http://linkinghub.elsevier.com/retrieve/pii/0003269786901740.

Habib, Manhal, Keren Shapira-Schweitzer, Oren Caspi, Amira Gepstein, Gil Arbel, Doron Aronson, Dror Seliktar, and Lior Gepstein. 2011. “A Combined Cell Therapy and in-Situ Tissue-Engineering Approach for Myocardial Repair.” Biomaterials 32 (30) (October): 7514–23. doi:10.1016/j.biomaterials.2011.06.049. http://www.ncbi.nlm.nih.gov/pubmed/21783246.

Hao, Xiaojin, Eduardo A Silva, Agneta Månsson-Broberg, Karl-Henrik Grinnemo, Anwar J Siddiqui, Göran Dellgren, Eva Wärdell, Lars Ake Brodin, David J Mooney, and Christer Sylvén. 2007. “Angiogenic Effects of Sequential Release of VEGF-A165 and PDGF-BB with Alginate Hydrogels after Myocardial Infarction.” Cardiovascular Research 75 (1): 178–85. doi:10.1016/j.cardiores.2007.03.028. http://www.ncbi.nlm.nih.gov/pubmed/17481597.

Harada, Mutsuo, Yingjie Qin, Hiroyuki Takano, Tohru Minamino, Yunzeng Zou, Haruhiro Toko, Masashi Ohtsuka, et al. 2005. “G-CSF Prevents Cardiac Remodeling after Myocardial Infarction by Activating the Jak-Stat Pathway in Cardiomyocytes.” Nature

Page 128: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

112

Medicine 11 (3) (March): 305–11. doi:10.1038/nm1199. http://www.ncbi.nlm.nih.gov/pubmed/15723072.

Hayes, a J, W Q Huang, J Mallah, D Yang, M E Lippman, and L Y Li. 1999. “Angiopoietin-1 and Its Receptor Tie-2 Participate in the Regulation of Capillary-like Tubule Formation and Survival of Endothelial Cells.” Microvascular Research 58 (3) (November): 224–37. doi:10.1006/mvre.1999.2179. http://www.ncbi.nlm.nih.gov/pubmed/10527766.

Hayworth, Douglas. 2014. “Carbodiimide Crosslinker Chemistry.” http://www.piercenet.com/method/carbodiimide-crosslinker-chemistry.

Hwang, Myung-Woo, Akira Matsumori, Yutaka Furukawa, Koh Ono, Masaharu Okada, Atsushi Iwasaki, Masatake Hara, Tadashi Miyamoto, Masanao Touma, and Shigetake Sasayama. 2001. “Neutralization of Interleukin-1β in the Acute Phase of Myocardial Infarction Promotes the Progression of Left Ventricular Remodeling.” Journal of the American College of Cardiology 38 (5) (November): 1546–1553. doi:10.1016/S0735-1097(01)01591-1. http://linkinghub.elsevier.com/retrieve/pii/S0735109701015911.

Ifkovits, Jamie L, Elena Tous, Masahito Minakawa, Masato Morita, J Daniel Robb, Kevin J Koomalsingh, Joseph H Gorman, Robert C Gorman, and Jason a Burdick. 2010. “Injectable Hydrogel Properties Influence Infarct Expansion and Extent of Postinfarction Left Ventricular Remodeling in an Ovine Model.” Proceedings of the National Academy of Sciences of the United States of America 107 (25) (June 22): 11507–12. doi:10.1073/pnas.1004097107. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2895138&tool=pmcentrez&rendertype=abstract.

Ikeda, S., and E.A. Foegeding. 2003. “Measurement of Gel Rheology: Dynamic Tests.” In Current Protocols in Food Analytical Chemistry, edited by Ronald E. Wrolstad, Terry E. Acree, Eric A. Decker, Michael H. Penner, David S. Reid, Steven J. Schwartz, Charles F. Shoemaker, Denise Smith, and Peter Sporns, 1–9. Hoboken, NJ, USA: John Wiley & Sons, Inc. doi:10.1002/0471142913. http://doi.wiley.com/10.1002/0471142913.

Ito, Y. 2008. “Covalently Immobilized Biosignal Molecule Materials for Tissue Engineering.” Soft Matter 4: 46–56 ST – Covalently immobilized biosignal molec. doi:10.1039/b708359. <Go to ISI>://000251580600004.

Iwakura, Atsushi, Masatoshi Fujita, Kazuaki Kataoka, Keiichi Tambara, Yutaka Sakakibara, Masashi Komeda, and Yasuhiko Tabata. 2003. “Intramyocardial Sustained Delivery of Basic Fibroblast Growth Factor Improves Angiogenesis and Ventricular Function in a Rat Infarct Model.” Heart and Vessels 18 (2) (May): 93–9. doi:10.1007/s10380-002-0686-5. http://www.ncbi.nlm.nih.gov/pubmed/12756606.

Iwanaga, Koji, Hiroyuki Takano, Masashi Ohtsuka, Hiroshi Hasegawa, Yunzeng Zou, Yingjie Qin, Kenichi Odaka, Kenzo Hiroshima, Hiroyuki Tadokoro, and Issei Komuro. 2004. “Effects of G-CSF on Cardiac Remodeling after Acute Myocardial Infarction in Swine.”

Page 129: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

113

Biochemical and Biophysical Research Communications 325 (4) (December): 1353–1359. doi:10.1016/j.bbrc.2004.10.149. http://www.ncbi.nlm.nih.gov/pubmed/15555576.

Iyer, Rohin K, Loraine L Y Chiu, Lewis a Reis, and Milica Radisic. 2011. “Engineered Cardiac Tissues.” Current Opinion in Biotechnology 22 (5) (October): 706–714. doi:10.1016/j.copbio.2011.04.004. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3169735&tool=pmcentrez&rendertype=abstract.

Iyer, Rohin K., Loraine L Y Chiu, and Milica Radisic. 2009. “Microfabricated Poly(ethylene Glycol) Templates Enable Rapid Screening of Triculture Conditions for Cardiac Tissue Engineering.” Journal of Biomedical Materials Research - Part A 89 (3) (June): 616–631. doi:10.1002/jbm.a.32014. http://www.ncbi.nlm.nih.gov/pubmed/18442120.

Iyer, Rohin K., Jane Chui, and Milica Radisic. 2009. “Spatiotemporal Tracking of Cells in Tissue-Engineered Cardiac Organoids.” Journal of Tissue Engineering and Regenerative Medicine 3 (February): 196–207. doi:10.1002/term.153.

Johnson, Todd D, Stephen Y Lin, and Karen L Christman. 2011. “Tailoring Material Properties of a Nanofibrous Extracellular Matrix Derived Hydrogel.” Nanotechnology 22 (49) (December 9): 494015. doi:10.1088/0957-4484/22/49/494015. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3280097&tool=pmcentrez&rendertype=abstract.

Kan, Chung-Dann, Shu-Hong Li, Richard D Weisel, Shun Zhang, and Ren-Ke Li. 2007. “Recipient Age Determines the Cardiac Functional Improvement Achieved by Skeletal Myoblast Transplantation.” Journal of the American College of Cardiology 50 (11) (September 11): 1086–92. doi:10.1016/j.jacc.2007.06.009. http://www.ncbi.nlm.nih.gov/pubmed/17825720.

Kang, Hyun-Jae, Hyo-Soo Kim, Bon-Kwon Koo, Yong-Jin Kim, DongSoo Lee, Dae-Won Sohn, Byung-Hee Oh, and Young-Bae Park. 2007. “Intracoronary Infusion of the Mobilized Peripheral Blood Stem Cell by G-CSF Is Better than Mobilization Alone by G-CSF for Improvement of Cardiac Function and Remodeling: 2-Year Follow-up Results of the Myocardial Regeneration and Angiogenesis in Myocardi.” American Heart Journal 153 (2) (February): 237.e1–8. doi:10.1016/j.ahj.2006.11.004. http://www.ncbi.nlm.nih.gov/pubmed/17239682.

Kang, Hyun-Jae, Hyo-Soo Kim, Shu-Ying Zhang, Kyung-Woo Park, Hyun-Jai Cho, Bon-Kwon Koo, Yong-Jin Kim, et al. 2004. “Effects of Intracoronary Infusion of Peripheral Blood Stem-Cells Mobilised with Granulocyte-Colony Stimulating Factor on Left Ventricular Systolic Function and Restenosis after Coronary Stenting in Myocardial Infarction: The MAGIC Cell Randomised Clinical.” Lancet 363 (9411) (March): 751–6. doi:10.1016/S0140-6736(04)15689-4. http://www.ncbi.nlm.nih.gov/pubmed/15016484.

Page 130: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

114

Kang, Jeehoon, Ji-Yeon Yun, Jin Hur, Jin-A Kang, Jae-Il Choi, Seung Bum Ko, Jaewon Lee, et al. 2014. “Erythropoietin Priming Improves the Vasculogenic Potential of G-CSF Mobilized Human Peripheral Blood Mononuclear Cells.” Cardiovascular Research (July 31). doi:10.1093/cvr/cvu180. http://www.ncbi.nlm.nih.gov/pubmed/25082847.

Kattman, Steven J, Tara L Huber, and Gordon M Keller. 2006. “Multipotent Flk-1+ Cardiovascular Progenitor Cells Give Rise to the Cardiomyocyte, Endothelial, and Vascular Smooth Muscle Lineages.” Developmental Cell 11 (5) (November): 723–32. doi:10.1016/j.devcel.2006.10.002. http://www.ncbi.nlm.nih.gov/pubmed/17084363.

Kean, T, and M Thanou. 2010. “Biodegradation, Biodistribution and Toxicity of Chitosan.” Advanced Drug Delivery Reviews 62 (1) (January 31): 3–11. doi:10.1016/j.addr.2009.09.004. http://www.ncbi.nlm.nih.gov/pubmed/19800377.

Kim, I., H. G. Kim, J.-N. So, J. H. Kim, H. J. Kwak, and G. Y. Koh. 2000. “Angiopoietin-1 Regulates Endothelial Cell Survival Through the Phosphatidylinositol 3’-Kinase/Akt Signal Transduction Pathway.” Circulation Research 86 (1) (January 7): 24–29. doi:10.1161/01.RES.86.1.24. http://circres.ahajournals.org/cgi/doi/10.1161/01.RES.86.1.24.

Kim, In Yong, Seog-Jin Seo, Hyun-Seuk Moon, Mi-Kyong Yoo, In-Young Park, Bom-Chol Kim, and Chong-Su Cho. 2008. “Chitosan and Its Derivatives for Tissue Engineering Applications.” Biotechnology Advances 26 (1): 1–21. doi:10.1016/j.biotechadv.2007.07.009. http://www.ncbi.nlm.nih.gov/pubmed/17884325.

Kirklin, James K, David C Naftel, Robert L Kormos, Lynne W Stevenson, Francis D Pagani, Marissa a Miller, J Timothy Baldwin, and James B Young. 2013. “Fifth INTERMACS Annual Report: Risk Factor Analysis from More than 6,000 Mechanical Circulatory Support Patients.” The Journal of Heart and Lung Transplantation : The Official Publication of the International Society for Heart Transplantation 32 (2) (February): 141–56. doi:10.1016/j.healun.2012.12.004. http://www.ncbi.nlm.nih.gov/pubmed/23352390.

Kouskoff, Valerie, Georges Lacaud, Staci Schwantz, Hans Jöerg Fehling, and Gordon Keller. 2005. “Sequential Development of Hematopoietic and Cardiac Mesoderm during Embryonic Stem Cell Differentiation.” Proceedings of the National Academy of Sciences of the United States of America 102 (37) (September): 13170–5. doi:10.1073/pnas.0501672102. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1201570&tool=pmcentrez&rendertype=abstract.

Kraehenbuehl, Thomas P, Lino S Ferreira, Alison M Hayward, Matthias Nahrendorf, André J van der Vlies, Eliza Vasile, Ralph Weissleder, Robert Langer, and Jeffrey a Hubbell. 2011. “Human Embryonic Stem Cell-Derived Microvascular Grafts for Cardiac Tissue Preservation after Myocardial Infarction.” Biomaterials 32 (4) (February): 1102–9. doi:10.1016/j.biomaterials.2010.10.005. http://www.ncbi.nlm.nih.gov/pubmed/21035182.

Page 131: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

115

Kraehenbuehl, Thomas P, Lino S Ferreira, Prisca Zammaretti, Jeffrey a Hubbell, and Robert Langer. 2009. “Cell-Responsive Hydrogel for Encapsulation of Vascular Cells.” Biomaterials 30 (26) (September): 4318–24. doi:10.1016/j.biomaterials.2009.04.057. http://www.ncbi.nlm.nih.gov/pubmed/19500842.

Krzemiński, Tadeusz F, Jerzy K Nozyński, Joanna Grzyb, and Maurycy Porc. 2008. “Wide-Spread Myocardial Remodeling after Acute Myocardial Infarction in Rat. Features for Heart Failure Progression.” Vascular Pharmacology 48 (2-3): 100–8. doi:10.1016/j.vph.2008.01.002. http://www.ncbi.nlm.nih.gov/pubmed/18276196.

Kumar, Dinender, Timothy a Hacker, Jennifer Buck, Larry F Whitesell, Eugene H Kaji, Pamela S Douglas, and Timothy J Kamp. 2005. “Distinct Mouse Coronary Anatomy and Myocardial Infarction Consequent to Ligation.” Coronary Artery Disease 16 (1) (February): 41–4. http://www.ncbi.nlm.nih.gov/pubmed/15654199.

Kwon, Jin Sook, In Kyu Park, Ae Shin Cho, Sun Mi Shin, Moon Hwa Hong, Seo Yeon Jeong, Yong Sook Kim, et al. 2009. “Enhanced Angiogenesis Mediated by Vascular Endothelial Growth Factor Plasmid-Loaded Thermo-Responsive Amphiphilic Polymer in a Rat Myocardial Infarction Model.” Journal of Controlled Release : Official Journal of the Controlled Release Society 138 (2) (September 1): 168–76. doi:10.1016/j.jconrel.2009.05.023. http://www.ncbi.nlm.nih.gov/pubmed/19465071.

L’Heureux, Nicolas, Nathalie Dusserre, Gerhardt Konig, Braden Victor, Paul Keire, Thomas N Wight, Nicolas a F Chronos, et al. 2006. “Human Tissue-Engineered Blood Vessels for Adult Arterial Revascularization.” Nature Medicine 12 (3) (March): 361–5. doi:10.1038/nm1364. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1513140&tool=pmcentrez&rendertype=abstract.

Laflamme, Michael a, Kent Y Chen, Anna V Naumova, Veronica Muskheli, James a Fugate, Sarah K Dupras, Hans Reinecke, et al. 2007. “Cardiomyocytes Derived from Human Embryonic Stem Cells in pro-Survival Factors Enhance Function of Infarcted Rat Hearts.” Nature Biotechnology 25 (9) (September): 1015–24. doi:10.1038/nbt1327. http://www.ncbi.nlm.nih.gov/pubmed/17721512.

Laflamme, Michael A, and Charles E Murry. 2005. “Regenerating the Heart.” Nature Biotechnology 23 (7): 845–56. doi:10.1038/nbt1117. http://www.ncbi.nlm.nih.gov/pubmed/16003373.

Landa, Natali, Liron Miller, Micha S Feinberg, Radka Holbova, Michal Shachar, Inbar Freeman, Smadar Cohen, and Jonathan Leor. 2008. “Effect of Injectable Alginate Implant on Cardiac Remodeling and Function after Recent and Old Infarcts in Rat.” Circulation 117 (11): 1388–96. doi:10.1161/CIRCULATIONAHA.107.727420. http://www.ncbi.nlm.nih.gov/pubmed/18316487.

Page 132: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

116

Lang, Nora, Maria J Pereira, Yuhan Lee, Ingeborg Friehs, Nikolay V Vasilyev, Eric N Feins, Klemens Ablasser, et al. 2014. “A Blood-Resistant Surgical Glue for Minimally Invasive Repair of Vessels and Heart Defects.” Science Translational Medicine 6 (218) (January 8): 218ra6. doi:10.1126/scitranslmed.3006557. http://www.ncbi.nlm.nih.gov/pubmed/24401941.

Lang, Roberto M, Michelle Bierig, Richard B Devereux, Frank a Flachskampf, Elyse Foster, Patricia a Pellikka, Michael H Picard, et al. 2006. “Recommendations for Chamber Quantification.” European Journal of Echocardiography : The Journal of the Working Group on Echocardiography of the European Society of Cardiology 7 (2) (March): 79–108. doi:10.1016/j.euje.2005.12.014. http://www.ncbi.nlm.nih.gov/pubmed/16458610.

Lee, Randall J, Andrew Hinson, Sam Helgerson, Robert Bauernschmitt, and Hani N Sabbah. 2012. “Polymer-Based Restoration of Left Ventricular Mechanics.” Cell Transplantation (March 28): 1–15. doi:10.3727/096368911X637461. http://www.ncbi.nlm.nih.gov/pubmed/22469060.

Leor, Jonathan, Natali Landa, and Smadar Cohen. 2006. “Renovation of the Injured Heart with Myocardial Tissue Engineering.” Expert Review of Cardiovascular Therapy 4 (2) (March): 239–52. doi:10.1586/14779072.4.2.239. http://www.ncbi.nlm.nih.gov/pubmed/16509819.

Leor, Jonathan, Shmuel Tuvia, Victor Guetta, Ferenc Manczur, David Castel, Udi Willenz, Ors Petneházy, et al. 2009. “Intracoronary Injection of in Situ Forming Alginate Hydrogel Reverses Left Ventricular Remodeling after Myocardial Infarction in Swine.” Journal of the American College of Cardiology 54 (11): 1014–23. doi:10.1016/j.jacc.2009.06.010. http://www.ncbi.nlm.nih.gov/pubmed/19729119.

Li, Q., E.T. T Dunn, E.W. W Grandmaison, and M.F.A. F A Goosen. 1992. “Applications and Properties of Chitosan.” Journal of Bioactive and Compatible Polymers 7 (4): 370–397. doi:10.1177/088391159200700406. http://jbc.sagepub.com/cgi/doi/10.1177/088391159200700406.

Li, RK Ren Ke, Zhi Qiang ZQ Jia, RD Richard D Weisel, DAG Donald AG G Mickle, Ji Zhang, Molly K Mohabeer, Vivek Rao, and Joan Ivanov. 1996. “Cardiomyocyte Transplantation Improves Heart Function.” The Annals of Thoracic Surgery 62 (3) (August): 654–661. doi:10.1016/S0003-4975(96)00389-X. http://linkinghub.elsevier.com/retrieve/pii/S000349759600389X.

Li, Zhenqing, and Jianjun Guan. 2011. “Hydrogels for Cardiac Tissue Engineering.” Polymers 3 (2) (April 9): 740–761. doi:10.3390/polym3020740. http://www.mdpi.com/2073-4360/3/2/740/.

Lipinski, Michael J, Giuseppe G L Biondi-Zoccai, Antonio Abbate, Reena Khianey, Imad Sheiban, Jozef Bartunek, Marc Vanderheyden, et al. 2007. “Impact of Intracoronary Cell Therapy on Left Ventricular Function in the Setting of Acute Myocardial Infarction: A Collaborative Systematic Review and Meta-Analysis of Controlled Clinical Trials.” Journal

Page 133: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

117

of the American College of Cardiology 50 (18) (October 30): 1761–7. doi:10.1016/j.jacc.2007.07.041. http://www.ncbi.nlm.nih.gov/pubmed/17964040.

Liu, Ying, Lijun Sun, Yi Huan, Haitao Zhao, and Jinglan Deng. 2006. “Effects of Basic Fibroblast Growth Factor Microspheres on Angiogenesis in Ischemic Myocardium and Cardiac Function: Analysis with Dobutamine Cardiovascular Magnetic Resonance Tagging.” European Journal of Cardio-Thoracic Surgery : Official Journal of the European Association for Cardio-Thoracic Surgery 30 (1) (July): 103–7. doi:10.1016/j.ejcts.2006.03.043. http://www.ncbi.nlm.nih.gov/pubmed/16730451.

Liu, Zhiqiang, Haibin Wang, Yan Wang, Qiuxia Lin, Anning Yao, Feng Cao, Dexue Li, et al. 2012. “The Influence of Chitosan Hydrogel on Stem Cell Engraftment, Survival and Homing in the Ischemic Myocardial Microenvironment.” Biomaterials 33 (11) (April): 3093–106. doi:10.1016/j.biomaterials.2011.12.044. http://www.ncbi.nlm.nih.gov/pubmed/22265788.

Lu, Wen Ning, Shuang-Hong Lü, Hai-Bin Wang, De-Xue Li, Cui-Mi Duan, Zhi-Qiang Liu, Wen-Jun He, et al. 2009. “Functional Improvement of Infarcted Heart by Co-Injection of Embryonic Stem Cells with Temperature-Responsive Chitosan Hydrogel.” Tissue Engineering. Part A 15 (6): 1437–47. doi:10.1089/ten.tea.2008.0143. http://www.ncbi.nlm.nih.gov/pubmed/19061432.

Ma, Heng, Hai-Feng Zhang, Lu Yu, Quan-Jiang Zhang, Jia Li, Jian-Hua Huo, Xue Li, Wen-Yi Guo, Hai-Chang Wang, and Feng Gao. 2006. “Vasculoprotective Effect of Insulin in the Ischemic/reperfused Canine Heart: Role of Akt-Stimulated NO Production.” Cardiovascular Research 69 (1) (January): 57–65. doi:10.1016/j.cardiores.2005.08.019. http://www.ncbi.nlm.nih.gov/pubmed/16212952.

Maisonpierre, P. C. 1997. “Angiopoietin-2, a Natural Antagonist for Tie2 That Disrupts in Vivo Angiogenesis.” Science 277 (5322) (July 4): 55–60. doi:10.1126/science.277.5322.55. http://www.sciencemag.org/cgi/doi/10.1126/science.277.5322.55.

Majeti, N.V. V, and Ravi Kumar. 2000. “A Review of Chitin and Chitosan Applications.” Reactive & Functional Polymers 46: 1–27.

Makino, S, K Fukuda, S Miyoshi, F Konishi, H Kodama, J Pan, M Sano, et al. 1999. “Cardiomyocytes Can Be Generated from Marrow Stromal Cells in Vitro.” The Journal of Clinical Investigation 103 (5) (March): 697–705. doi:10.1172/JCI5298. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=408125&tool=pmcentrez&rendertype=abstract.

Marino, Paolo, Luisa Zanolla, and Piero Zardini. 1989. “Effect of Streptokinase on Left Ventricular Modeling and Function after Myocardial Infarction: The GISSI (Gruppo Italiano per Lo Studio Della Streptochinasi nell’Infarto Miocardico) Trial.” Journal of the American College of Cardiology 14 (5) (November): 1149–1158. doi:10.1016/0735-1097(89)90409-9. http://linkinghub.elsevier.com/retrieve/pii/0735109789904099.

Page 134: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

118

Martens, Timothy P, Amandine F G Godier, Jonathan J Parks, Leo Q Wan, Michael S Koeckert, George M Eng, Barry I Hudson, Warren Sherman, and Gordana Vunjak-Novakovic. 2009. “Percutaneous Cell Delivery into the Heart Using Hydrogels Polymerizing in Situ.” Cell Transplantation 18 (3) (January): 297–304. doi:10.3727/096368909788534915. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2771541&tool=pmcentrez&rendertype=abstract.

Martin, Cindy M., Annette P. Meeson, Scott M. Robertson, Thomas J. Hawke, James A. Richardson, Susan Bates, Sean C. Goetsch, Teresa D. Gallardo, and Daniel J. Garry. 2004. “Persistent Expression of the ATP-Binding Cassette Transporter, Abcg2, Identifies Cardiac SP Cells in the Developing and Adult Heart☆.” Developmental Biology 265 (1) (January): 262–275. doi:10.1016/j.ydbio.2003.09.028. http://linkinghub.elsevier.com/retrieve/pii/S0012160603005815.

Mathers, Colin D, and Dejan Loncar. 2006. “Projections of Global Mortality and Burden of Disease from 2002 to 2030.” PLoS Medicine 3 (11) (November): e442. doi:10.1371/journal.pmed.0030442. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=1664601&tool=pmcentrez&rendertype=abstract.

Mayfield, Audrey E, Everad L Tilokee, Nicholas Latham, Brian McNeill, Bu-Khanh Lam, Marc Ruel, Erik J Suuronen, David W Courtman, Duncan J Stewart, and Darryl R Davis. 2014. “The Effect of Encapsulation of Cardiac Stem Cells within Matrix-Enriched Hydrogel Capsules on Cell Survival, Post-Ischemic Cell Retention and Cardiac Function.” Biomaterials 35 (1) (January): 133–42. doi:10.1016/j.biomaterials.2013.09.085. http://www.ncbi.nlm.nih.gov/pubmed/24099706.

McMurray, John, Scott Solomon, Karen Pieper, Shelby Reed, Jean Rouleau, Eric Velazquez, Harvey White, et al. 2006. “The Effect of Valsartan, Captopril, or Both on Atherosclerotic Events after Acute Myocardial Infarction: An Analysis of the Valsartan in Acute Myocardial Infarction Trial (VALIANT).” Journal of the American College of Cardiology 47 (4) (February 21): 726–33. doi:10.1016/j.jacc.2005.09.055. http://www.ncbi.nlm.nih.gov/pubmed/16487836.

Memon, Imran a, Yoshiki Sawa, Norihide Fukushima, Goro Matsumiya, Shigeru Miyagawa, Satoshi Taketani, Satoru K Sakakida, et al. 2005. “Repair of Impaired Myocardium by Means of Implantation of Engineered Autologous Myoblast Sheets.” The Journal of Thoracic and Cardiovascular Surgery 130 (5) (November): 1333–41. doi:10.1016/j.jtcvs.2005.07.023. http://www.ncbi.nlm.nih.gov/pubmed/16256786.

Menasché, Philippe. 2008. “Current Status and Future Prospects for Cell Transplantation to Prevent Congestive Heart Failure.” Seminars in Thoracic and Cardiovascular Surgery 20 (2) (January): 131–7. doi:10.1053/j.semtcvs.2008.03.001. http://www.ncbi.nlm.nih.gov/pubmed/18707646.

Page 135: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

119

Menasché, Philippe, Albert A Hagège, Jean-Thomas Vilquin, Michel Desnos, Eric Abergel, Bruno Pouzet, Alain Bel, et al. 2003. “Autologous Skeletal Myoblast Transplantation for Severe Postinfarction Left Ventricular Dysfunction.” Journal of the American College of Cardiology 41 (7) (April): 1078–1083. doi:10.1016/S0735-1097(03)00092-5. http://linkinghub.elsevier.com/retrieve/pii/S0735109703000925.

Miklas, Jason W, Susan M Dallabrida, Lewis a Reis, Nesreen Ismail, Maria Rupnick, and Milica Radisic. 2013. “QHREDGS Enhances Tube Formation, Metabolism and Survival of Endothelial Cells in Collagen-Chitosan Hydrogels.” PloS One 8 (8) (January): e72956. doi:10.1371/journal.pone.0072956. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3754933&tool=pmcentrez&rendertype=abstract.

Minatoguchi, Shinya, Genzou Takemura, Xue-Hai Chen, Ningyuan Wang, Yoshihiro Uno, Masahiko Koda, Masazumi Arai, et al. 2004. “Acceleration of the Healing Process and Myocardial Regeneration May Be Important as a Mechanism of Improvement of Cardiac Function and Remodeling by Postinfarction Granulocyte Colony-Stimulating Factor Treatment.” Circulation 109 (21) (June): 2572–80. doi:10.1161/01.CIR.0000129770.93985.3E. http://www.ncbi.nlm.nih.gov/pubmed/15123535.

Miyagawa, Shigeru, Yoshiki Sawa, Satoru Sakakida, Satoshi Taketani, Haruhiko Kondoh, Imran Ahmed Memon, Yukiko Imanishi, Tatsuya Shimizu, Teruo Okano, and Hikaru Matsuda. 2005. “Tissue Cardiomyoplasty Using Bioengineered Contractile Cardiomyocyte Sheets to Repair Damaged Myocardium: Their Integration with Recipient Myocardium.” Transplantation 80 (11) (December): 1586–1595. doi:10.1097/01.tp.0000181163.69108.dd. http://content.wkhealth.com/linkback/openurl?sid=WKPTLP:landingpage&an=00007890-200512150-00013.

Miyagi, Y, L L Chiu, M Cimini, R D Weisel, M Radisic, and R K Li. 2011. “Biodegradable Collagen Patch with Covalently Immobilized VEGF for Myocardial Repair.” Biomaterials 32: 1280–90 ST – Biodegradable collagen patch with co. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=21035179.

Muller-Ehmsen, J. 2002. “Rebuilding a Damaged Heart: Long-Term Survival of Transplanted Neonatal Rat Cardiomyocytes After Myocardial Infarction and Effect on Cardiac Function.” Circulation 105 (14) (March): 1720–1726. doi:10.1161/01.CIR.0000013782.76324.92. http://circ.ahajournals.org/cgi/doi/10.1161/01.CIR.0000013782.76324.92.

Müller-Ehmsen, Jochen, Peter Whittaker, Robert a Kloner, Joan S Dow, Tsuyoshi Sakoda, Tiffany I Long, Peter W Laird, and Larry Kedes. 2002. “Survival and Development of Neonatal Rat Cardiomyocytes Transplanted into Adult Myocardium.” Journal of Molecular and Cellular Cardiology 34 (2) (February): 107–16. doi:10.1006/jmcc.2001.1491. http://www.ncbi.nlm.nih.gov/pubmed/11851351.

Page 136: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

120

Murry, Charles E, Mark H Soonpaa, Hans Reinecke, Hisako O Hidehiro Nakajima, Michael Rubart, Kishore B S Pasumarthi, Jitka Ismail Virag, et al. 2004. “Haematopoietic Stem Cells Do Not Transdifferentiate into Cardiac Myocytes in Myocardial Infarcts.” Nature 428 (6983) (April): 664–8. doi:10.1038/nature02446. http://www.ncbi.nlm.nih.gov/pubmed/15034593.

Muzzarelli, Riccardo A. A. 1993. “Biochemical Significance of Exogenous Chitins and Chitosans in Animals and Patients.” Carbohydrate Polymers 20 (1): 7–16. doi:10.1016/0144-8617(93)90027-2. http://linkinghub.elsevier.com/retrieve/pii/0144861793900272.

Naito, Hiroshi, Ivan Melnychenko, Michael Didié, Karin Schneiderbanger, Pia Schubert, Stephan Rosenkranz, Thomas Eschenhagen, and Wolfram Hubertus Zimmermann. 2006. “Optimizing Engineered Heart Tissue for Therapeutic Applications as Surrogate Heart Muscle.” Circulation 114: 72–79. doi:10.1161/CIRCULATIONAHA.105.001560.

Nelson, Devin M, Zuwei Ma, Kazuro L Fujimoto, Ryotaro Hashizume, and William R Wagner. 2011. “Intra-Myocardial Biomaterial Injection Therapy in the Treatment of Heart Failure: Materials, Outcomes and Challenges.” Acta Biomaterialia 7 (1) (January): 1–15. doi:10.1016/j.actbio.2010.06.039. http://www.ncbi.nlm.nih.gov/pubmed/20619368.

Nelson, Devin M, Zuwei Ma, Cory E Leeson, and William R Wagner. 2012. “Extended and Sequential Delivery of Protein from Injectable Thermoresponsive Hydrogels.” Journal of Biomedical Materials Research. Part A 100 (3) (March): 776–85. doi:10.1002/jbm.a.34015. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3266437&tool=pmcentrez&rendertype=abstract.

Nian, Min, Paul Lee, Neelam Khaper, and Peter Liu. 2004. “Inflammatory Cytokines and Postmyocardial Infarction Remodeling.” Circulation Research 94 (12) (June 25): 1543–53. doi:10.1161/01.RES.0000130526.20854.fa. http://www.ncbi.nlm.nih.gov/pubmed/15217919.

Nicodemus, Garret D, and Stephanie J Bryant. 2008. “Cell Encapsulation in Biodegradable Hydrogels for Tissue Engineering Applications.” Tissue Engineering. Part B, Reviews 14 (2) (June): 149–65. doi:10.1089/ten.teb.2007.0332. http://www.ncbi.nlm.nih.gov/pubmed/18498217.

Nunes, Sara S, Jason W Miklas, Jie Liu, Roozbeh Aschar-Sobbi, Yun Xiao, Boyang Zhang, Jiahua Jiang, et al. 2013. “Biowire: A Platform for Maturation of Human Pluripotent Stem Cell-Derived Cardiomyocytes.” Nature Methods 10 (8) (August): 781–7. doi:10.1038/nmeth.2524. http://www.ncbi.nlm.nih.gov/pubmed/23793239.

Nygren, Jens M, Stefan Jovinge, Martin Breitbach, Petter Säwén, Wilhelm Röll, Jürgen Hescheler, Jalal Taneera, Bernd K Fleischmann, and Sten Eirik W Jacobsen. 2004. “Bone Marrow-Derived Hematopoietic Cells Generate Cardiomyocytes at a Low Frequency

Page 137: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

121

through Cell Fusion, but Not Transdifferentiation.” Nature Medicine 10 (5) (May): 494–501. doi:10.1038/nm1040. http://www.ncbi.nlm.nih.gov/pubmed/15107841.

Odedra, Devang, Loraine L Y Chiu, Lewis A Reis, Fiona Rask, Katherine Chiang, and Milica Radisic. 2011. “Cardiac Tissue Engineering.” In Biomaterials for Tissue Engineering Applications: A Review of Past & Future Trends, edited by Jason A. Burdick and Robert L. Mauck, 1st Ed., 562. Vienna: Springer Vienna. doi:10.1007/978-3-7091-0385-2. http://www.springerlink.com/index/10.1007/978-3-7091-0385-2.

Oh, Hidemasa, Steven B Bradfute, Teresa D Gallardo, Teruya Nakamura, Vinciane Gaussin, Yuji Mishina, Jennifer Pocius, et al. 2003. “Cardiac Progenitor Cells from Adult Myocardium: Homing, Differentiation, and Fusion after Infarction.” Proceedings of the National Academy of Sciences of the United States of America 100 (21) (October): 12313–8. doi:10.1073/pnas.2132126100. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=218755&tool=pmcentrez&rendertype=abstract.

Okada, Masaho, Thomas R Payne, Hideki Oshima, Nobuo Momoi, Kimimasa Tobita, and Johnny Huard. 2010. “Differential Efficacy of Gels Derived from Small Intestinal Submucosa as an Injectable Biomaterial for Myocardial Infarct Repair.” Biomaterials 31 (30) (October): 7678–83. doi:10.1016/j.biomaterials.2010.06.056. http://www.ncbi.nlm.nih.gov/pubmed/20674011.

Onishi, H, and Y Machida. 1999. “Biodegradation and Distribution of Water-Soluble Chitosan in Mice.” Biomaterials 20 (2) (January): 175–82. http://www.ncbi.nlm.nih.gov/pubmed/10022787.

Orlic, Donald, Jan Kajstura, Stefano Chimenti, Igor Jakoniuk, S M Anderson, B Li, J Pickel, et al. 2001. “Bone Marrow Cells Regenerate Infarcted Myocardium.” Nature 410 (6829) (April): 701–5. doi:10.1038/35070587. http://www.ncbi.nlm.nih.gov/pubmed/11287958.

Orlic, Donald, Jan Kajstura, Stefano Chimenti, Federica Limana, Igor Jakoniuk, Federico Quaini, Bernardo Nadal-Ginard, David M. Bodine, Annarosa Leri, and Piero Anversa. 2001. “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 98 (18) (August): 10344–9. doi:10.1073/pnas.181177898. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=56963&tool=pmcentrez&rendertype=abstract.

Ott, Harald C, Thomas S Matthiesen, Saik Kia Goh, Lauren D Black, Stefan M Kren, Theoden I Netoff, and Doris A Taylor. 2008. “Perfusion-Decellularized Matrix: Using Nature’s Platform to Engineer a Bioartificial Heart.” Nature Medicine 14 (2) (February): 213–21. doi:10.1038/nm1684. http://www.ncbi.nlm.nih.gov/pubmed/18193059.

Ozawa, Tsukasa, Donald A G Mickle, Richard D Weisel, Nobuya Koyama, Sumiko Ozawa, and Ren-Ke Li. 2002. “Optimal Biomaterial for Creation of Autologous Cardiac Grafts.”

Page 138: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

122

Circulation 106 (12 Suppl 1) (September 24): I176–82. doi:10.1161/01.cir.0000032901.55215.cc. http://www.ncbi.nlm.nih.gov/pubmed/12354729.

Pantilat, Steven Z, and Anthony E Steimle. 2004. “Palliative Care for Patients with Heart Failure.” JAMA : The Journal of the American Medical Association 291 (20) (May 26): 2476–82. doi:10.1001/jama.291.20.2476. http://jama.ama-assn.org/content/291/20/2476.short.

Papapetropoulos, A. 2000. “Angiopoietin-1 Inhibits Endothelial Cell Apoptosis via the Akt/Survivin Pathway.” Journal of Biological Chemistry 275 (13) (March 24): 9102–9105. doi:10.1074/jbc.275.13.9102. http://www.jbc.org/cgi/doi/10.1074/jbc.275.13.9102.

Patten, RD, MJ Aronovitz, L. Deras-Mejia, N.G. Pandian, G.G. Hanak, J.J. Smith, M.E. Mendelsohn, and M.A. Konstam. 1998. “Ventricular Remodeling in a Mouse Model of Myocardial Infarction.” American Journal of Physiology-Heart and Circulatory Physiology 274 (5): H1812. http://ajpheart.physiology.org/content/274/5/H1812.short.

Pfeffer, M. a., and E. Braunwald. 1990. “Ventricular Remodeling after Myocardial Infarction. Experimental Observations and Clinical Implications.” Circulation 81 (4) (April 1): 1161–1172. doi:10.1161/01.CIR.81.4.1161. http://circ.ahajournals.org/cgi/doi/10.1161/01.CIR.81.4.1161.

Prasitsilp, M, R Jenwithisuk, K Kongsuwan, N Damrongchai, and P Watts. 2000. “Cellular Responses to Chitosan in Vitro: The Importance of Deacetylation.” Journal of Materials Science. Materials in Medicine 11 (12) (December): 773–8. http://www.ncbi.nlm.nih.gov/pubmed/15348059.

Pulapura, S., and J. Kohn. 1992. “Trends in the Development of Bioresorbable Polymers for Medical Applications.” Journal of Biomaterials Applications 6 (3) (January): 216–250. doi:10.1177/088532829200600303. http://jba.sagepub.com/cgi/doi/10.1177/088532829200600303.

Puppi, D., F. Chiellini, a.M. Piras, and E. Chiellini. 2010. “Polymeric Materials for Bone and Cartilage Repair.” Progress in Polymer Science 35 (4) (April): 403–440. doi:10.1016/j.progpolymsci.2010.01.006. http://linkinghub.elsevier.com/retrieve/pii/S0079670010000134.

Radisic, Milica, Michelle Euloth, Liming Yang, Robert Langer, Lisa E Freed, and Gordana Vunjak-Novakovic. 2003. “High-Density Seeding of Myocyte Cells for Cardiac Tissue Engineering.” Biotechnology and Bioengineering 82 (4) (May 20): 403–14. doi:10.1002/bit.10594. http://www.ncbi.nlm.nih.gov/pubmed/12632397.

Radisic, Milica, Jos Malda, Eric Epping, Wenliang Geng, Robert Langer, and Gordana Vunjak-Novakovic. 2006. “Oxygen Gradients Correlate with Cell Density and Cell Viability in Engineered Cardiac Tissue.” Biotechnology and Bioengineering 93 (2) (February 5): 332–43. doi:10.1002/bit.20722. http://www.ncbi.nlm.nih.gov/pubmed/16270298.

Page 139: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

123

Radisic, Milica, Hyoungshin Park, Helen Shing, Thomas Consi, Frederick J Schoen, Robert Langer, Lisa E Freed, and Gordana Vunjak-Novakovic. 2004. “Functional Assembly of Engineered Myocardium by Electrical Stimulation of Cardiac Myocytes Cultured on Scaffolds.” Proceedings of the National Academy of Sciences of the United States of America 101 (52) (December 28): 18129–34. doi:10.1073/pnas.0407817101. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=539727&tool=pmcentrez&rendertype=abstract.

Rane, Aboli a, Joyce S Chuang, Amul Shah, Diane P Hu, Nancy D Dalton, Yusu Gu, Kirk L Peterson, Jeffrey H Omens, and Karen L Christman. 2011. “Increased Infarct Wall Thickness by a Bio-Inert Material Is Insufficient to Prevent Negative Left Ventricular Remodeling after Myocardial Infarction.” PloS One 6 (6) (January): e21571. doi:10.1371/journal.pone.0021571. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3121880&tool=pmcentrez&rendertype=abstract.

Rask, Fiona. 2009. “The Development of an Injectible Hydrogel for the Site-Specific Delivery of Cardiomyocytes to the Heart.” University of Toronto.

Rask, Fiona, Susan M Dallabrida, Nesreen S Ismail, Zohreh Amoozgar, Yoon Yeo, Maria A Rupnick, and Milica Radisic. 2010. “Photocrosslinkable Chitosan Modified with Angiopoietin-1 Peptide, QHREDGS, Promotes Survival of Neonatal Rat Heart Cells.” Journal of Biomedical Materials Research. Part A 95 (1) (October): 105–17. doi:10.1002/jbm.a.32808. http://www.ncbi.nlm.nih.gov/pubmed/20540095.

Rask, Fiona, Anton Mihic, Lewis Reis, Susan M Dallabrida, Nesreen S Ismail, Krista Sider, Craig. A Simmons, et al. 2010. “Hydrogels Modified with QHREDGS Peptide Support Cardiomyocyte Survival in Vitro and after Sub-Cutaneous Implantation.” Soft Matter 6 (20): 5089–5099. doi:10.1039/c0sm00362j. http://xlink.rsc.org/?DOI=c0sm00362j.

Reffelmann, Thorsten, and Robert A Kloner. 2003. “Cellular Cardiomyoplasty—cardiomyocytes, Skeletal Myoblasts, or Stem Cells for Regenerating Myocardium and Treatment of Heart Failure?” Cardiovascular Research 58 (2) (May): 358–368. doi:10.1016/S0008-6363(02)00739-3. http://cardiovascres.oxfordjournals.org/cgi/doi/10.1016/S0008-6363(02)00739-3.

Reinecke, Hans, Glen H MacDonald, Stephen D Hauschka, and Charles E Murry. 2000. “Electromechanical Coupling between Skeletal and Cardiac Muscle. Implications for Infarct Repair.” The Journal of Cell Biology 149 (3) (May): 731–40. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2174851&tool=pmcentrez&rendertype=abstract.

Reinecke, Hans, Veronica Poppa, and Charles E Murry. 2002. “Skeletal Muscle Stem Cells Do Not Transdifferentiate into Cardiomyocytes after Cardiac Grafting.” Journal of Molecular and Cellular Cardiology 34 (2) (February): 241–9. doi:10.1006/jmcc.2001.1507. http://www.ncbi.nlm.nih.gov/pubmed/11851363.

Page 140: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

124

Reinecke, Hans, Ming Zhang, Trudy Bartosek, and Charles E Murry. 1999. “Survival, Integration, and Differentiation of Cardiomyocyte Grafts: A Study in Normal and Injured Rat Hearts.” Circulation 100 (2): 193–202. http://circ.ahajournals.org/cgi/content/abstract/circulationaha;100/2/193.

Reis, Lewis A, Loraine L Y Chiu, Nicole Feric, Lara Fu, and Milica Radisic. 2014a. Injectable Biomaterials for Cardiac Regeneration and Repair. Edited by Ren-Ke Li and Richard D Weisel. Cardiac Regeneration & Repair Volume II: Biomaterials & Tissue Engineering. 1st ed. Cambridge, UK: Elsevier. doi:10.1533/9780857096715.1.49.

———. 2014b. “Biomaterials in Myocardial Tissue Engineering.” Journal of Tissue Engineering and Regenerative Medicine (July 28). doi:10.1002/term.1944. http://www.ncbi.nlm.nih.gov/pubmed/25066525.

Reis, Lewis A, Loraine L Y Chiu, Yan Liang, Kent Hyunh, Abdul Momen, and Milica Radisic. 2012. “A Peptide-Modified Chitosan-Collagen Hydrogel for Cardiac Cell Culture and Delivery.” Acta Biomaterialia 8 (3) (March): 1022–1036. doi:10.1016/j.actbio.2011.11.030. http://www.ncbi.nlm.nih.gov/pubmed/22155066.

Reis, Lewis A, Loraine L Y Chiu, Jun Wu, Nicole Feric, Momen Abdul, Ren-Ke Li, and Milica Radisic. 2015. “Hydrogels with Integrin-Binding Angiopoietin-1-Derived Peptide, QHREDGS, for Treatment of Acute Myocardial Infarction.” Circulation Heart Failure. doi:10.1161/CIRCHEARTFAILURE.114.001881.

Roger, Véronique L, Alan S Go, Donald M Lloyd-Jones, Emelia J Benjamin, Jarett D Berry, William B Borden, Dawn M Bravata, et al. 2012. “Heart Disease and Stroke Statistics--2012 Update: A Report from the American Heart Association.” Circulation 125 (1) (January 3): e2–e220. doi:10.1161/CIR.0b013e31823ac046. http://www.ncbi.nlm.nih.gov/pubmed/22179539.

Rossomacha, Evgeny, Caroline D. Hoemanni, and Matthew S. Shive. 2004. “Simple Methods for Staining Chitosan in Biotechnological Applications.” Journal of Histotechnology 27 (1) (March): 31–36. doi:10.1179/his.2004.27.1.31. http://www.maneyonline.com/doi/abs/10.1179/his.2004.27.1.31.

Rufaihah, Abdul Jalil, Srirangam Ramanujam Vaibavi, Marian Plotkin, Jiayi Shen, Venkateswaran Nithya, Jing Wang, Dror Seliktar, and Theodoros Kofidis. 2013. “Enhanced Infarct Stabilization and Neovascularization Mediated by VEGF-Loaded PEGylated Fibrinogen Hydrogel in a Rodent Myocardial Infarction Model.” Biomaterials 34 (33) (November): 8195–202. doi:10.1016/j.biomaterials.2013.07.031. http://www.ncbi.nlm.nih.gov/pubmed/23891519.

Ruggiero, Alessandro, Daniel L J Thorek, Jamal Guenoun, Gabriel P Krestin, and Monique R Bernsen. 2012. “Cell Tracking in Cardiac Repair: What to Image and How to Image.” European Radiology 22 (1) (January): 189–204. doi:10.1007/s00330-011-2190-7.

Page 141: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

125

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3229694&tool=pmcentrez&rendertype=abstract.

Ruvinov, Emil, Jonathan Leor, and Smadar Cohen. 2011. “The Promotion of Myocardial Repair by the Sequential Delivery of IGF-1 and HGF from an Injectable Alginate Biomaterial in a Model of Acute Myocardial Infarction.” Biomaterials 32 (2) (January): 565–78. doi:10.1016/j.biomaterials.2010.08.097. http://www.ncbi.nlm.nih.gov/pubmed/20889201.

Sakakibara, Yutaka, Kazunobu Nishimura, Keiichi Tambara, Masaya Yamamoto, Fanglin Lu, Yasuhiko Tabata, and Masashi Komeda. 2002. “Prevascularization with Gelatin Microspheres Containing Basic Fibroblast Growth Factor Enhances the Benefits of Cardiomyocyte Transplantation.” The Journal of Thoracic and Cardiovascular Surgery 124 (1) (July): 50–56. doi:10.1067/mtc.2002.121293. http://linkinghub.elsevier.com/retrieve/pii/S0022522302000399.

Sam, Flora, Douglas B Sawyer, Donny L Chang, Franz R Eberli, Soeun Ngoy, Mohit Jain, Jay Amin, Carl S Apstein, and Wilson S Colucci. 2000. “Progressive Left Ventricular Remodeling and Apoptosis Late after Myocardial Infarction in Mouse Heart.” American Journal of Physiology. Heart and Circulatory Physiology 279 (1) (July): H422–8. http://www.ncbi.nlm.nih.gov/pubmed/10899082.

Segers, Vincent F M, and Richard T Lee. 2011. “Biomaterials to Enhance Stem Cell Function in the Heart.” Circulation Research 109 (8) (September 30): 910–22. doi:10.1161/CIRCRESAHA.111.249052. http://www.ncbi.nlm.nih.gov/pubmed/21960724.

Segers, Vincent F M, Tomotake Tokunou, Luke J Higgins, Catherine MacGillivray, Joseph Gannon, and Richard T Lee. 2007. “Local Delivery of Protease-Resistant Stromal Cell Derived Factor-1 for Stem Cell Recruitment after Myocardial Infarction.” Circulation 116 (15) (October 9): 1683–92. doi:10.1161/CIRCULATIONAHA.107.718718. http://www.ncbi.nlm.nih.gov/pubmed/17875967.

Seif-Naraghi, Sonya B, Michael a Salvatore, Pam J Schup-Magoffin, Diane P Hu, and Karen L Christman. 2010. “Design and Characterization of an Injectable Pericardial Matrix Gel: A Potentially Autologous Scaffold for Cardiac Tissue Engineering.” Tissue Engineering. Part A 16 (6) (June): 2017–27. doi:10.1089/ten.TEA.2009.0768. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2949214&tool=pmcentrez&rendertype=abstract.

Seok, Seung Hyeok, Jong-Ik Heo, Ji-Hye Hwang, Yi-Rang Na, Jang-Hyuk Yun, Eun Hui Lee, Jong-Wan Park, and Chung-Hyun Cho. 2013. “Angiopoietin-1 Elicits pro-Inflammatory Responses in Monocytes and Differentiating Macrophages.” Molecules and Cells 35 (6) (June): 550–6. doi:10.1007/s10059-013-0088-8. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3887877&tool=pmcentrez&rendertype=abstract.

Page 142: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

126

Shake, Jay G, Peter J Gruber, William a Baumgartner, Guylaine Senechal, Jennifer Meyers, J Mark Redmond, Mark F Pittenger, and Bradley J Martin. 2002. “Mesenchymal Stem Cell Implantation in a Swine Myocardial Infarct Model: Engraftment and Functional Effects.” The Annals of Thoracic Surgery 73 (6) (June): 1919–26; discussion 1926. http://www.ncbi.nlm.nih.gov/pubmed/12078791.

Shi, Chunmeng, Ying Zhu, Xinze Ran, Meng Wang, Yongping Su, and Tianmin Cheng. 2006. “Therapeutic Potential of Chitosan and Its Derivatives in Regenerative Medicine.” The Journal of Surgical Research 133 (2) (June 15): 185–92. doi:10.1016/j.jss.2005.12.013. http://www.ncbi.nlm.nih.gov/pubmed/16458923.

Singelyn, Jennifer M, Jessica a DeQuach, Sonya B Seif-Naraghi, Robert B Littlefield, Pamela J Schup-Magoffin, and Karen L Christman. 2009. “Naturally Derived Myocardial Matrix as an Injectable Scaffold for Cardiac Tissue Engineering.” Biomaterials 30 (29) (October): 5409–16. doi:10.1016/j.biomaterials.2009.06.045. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2728782&tool=pmcentrez&rendertype=abstract.

Singelyn, Jennifer M, Priya Sundaramurthy, Todd D Johnson, Pamela J Schup-Magoffin, Diane P Hu, Denver M Faulk, Jean Wang, et al. 2012. “Catheter-Deliverable Hydrogel Derived from Decellularized Ventricular Extracellular Matrix Increases Endogenous Cardiomyocytes and Preserves Cardiac Function Post-Myocardial Infarction.” Journal of the American College of Cardiology 59 (8) (February 21): 751–63. doi:10.1016/j.jacc.2011.10.888. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3285410&tool=pmcentrez&rendertype=abstract.

Smart, Nicola, Catherine a Risebro, Athalie a D Melville, Kelvin Moses, Robert J Schwartz, Kenneth R Chien, and Paul R Riley. 2007. “Thymosin beta4 Induces Adult Epicardial Progenitor Mobilization and Neovascularization.” Nature 445 (7124) (January): 177–82. doi:10.1038/nature05383. http://www.ncbi.nlm.nih.gov/pubmed/17108969.

Soonpaa, Mark H, and Loren J Field. 1998. “Survey of Studies Examining Mammalian Cardiomyocyte DNA Synthesis.” Circulation Research 83: 15–26. http://circres.ahajournals.org/cgi/content/abstract/83/1/15.

Sun, M. 2003. “Temporal Response and Localization of Integrins beta1 and beta3 in the Heart After Myocardial Infarction: Regulation by Cytokines.” Circulation 107 (7) (February 17): 1046–1052. doi:10.1161/01.CIR.0000051363.86009.3C. http://circ.ahajournals.org/cgi/doi/10.1161/01.CIR.0000051363.86009.3C.

Sun, Zhuo, Jun Wu, Hiroko Fujii, Jiang Wu, Shu-Hong Li, Svetlana Porozov, Adina Belleli, Valentin Fulga, Yael Porat, and Ren-Ke Li. 2008. “Human Angiogenic Cell Precursors Restore Function in the Infarcted Rat Heart: A Comparison of Cell Delivery Routes.” European Journal of Heart Failure 10 (6) (June): 525–33. doi:10.1016/j.ejheart.2008.04.004. http://www.ncbi.nlm.nih.gov/pubmed/18490195.

Page 143: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

127

Sürder, Daniel, Robert Manka, Viviana Lo Cicero, Tiziano Moccetti, Kaspar Rufibach, Sabrina Soncin, Lucia Turchetto, et al. 2013. “Intracoronary Injection of Bone Marrow-Derived Mononuclear Cells Early or Late after Acute Myocardial Infarction: Effects on Global Left Ventricular Function.” Circulation 127: 1968–1979. doi:10.1161/CIRCULATIONAHA.112.001035.

Sutton, M G, and N Sharpe. 2000. “Left Ventricular Remodeling after Myocardial Infarction: Pathophysiology and Therapy.” Circulation 101 (25) (June): 2981–8. http://www.ncbi.nlm.nih.gov/pubmed/10869273.

Suuronen, Erik J, John P Veinot, Serena Wong, Varun Kapila, Joel Price, May Griffith, Thierry G Mesana, and Marc Ruel. 2006. “Tissue-Engineered Injectable Collagen-Based Matrices for Improved Cell Delivery and Vascularization of Ischemic Tissue Using CD133+ Progenitors Expanded from the Peripheral Blood.” Circulation 114 (1 Suppl): I138–44. doi:10.1161/CIRCULATIONAHA.105.001081. http://www.ncbi.nlm.nih.gov/pubmed/16820563.

Suuronen, Erik J, Pingchuan Zhang, Thierry G Mesana, Drew Kuraitis, John P Veinot, Xudong Cao, Angela Melhuish, Daniel McKee, Fengfu Li, and Marc Ruel. 2009. “An Acellular Matrix-Bound Ligand Enhances the Mobilization, Recruitment and Therapeutic Effects of Circulating Progenitor Cells in a Hindlimb Ischemia Model.” The FASEB Journal : Official Publication of the Federation of American Societies for Experimental Biology 23 (5): 1447–58. doi:10.1096/fj.08-111054. http://www.ncbi.nlm.nih.gov/pubmed/19136616.

Takahashi, Kazutoshi, Koji Tanabe, Mari Ohnuki, Megumi Narita, Tomoko Ichisaka, Kiichiro Tomoda, and Shinya Yamanaka. 2007. “Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors.” Cell 131 (5) (November): 861–72. doi:10.1016/j.cell.2007.11.019. http://www.ncbi.nlm.nih.gov/pubmed/18035408.

Takahashi, Kazutoshi, and Shinya Yamanaka. 2006. “Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors.” Cell 126 (4) (August): 663–676. doi:10.1016/j.cell.2006.07.024. http://www.ncbi.nlm.nih.gov/pubmed/16904174.

Tan, Wei, Raj Krishnaraj, and Tejal A Desai. 2001. “Evaluation of Nanostructured Composite CollagenChitosan Matrices for Tissue Engineering.” Tissue Engineering 7 (2): 203–210.

Touchstone, D a, G a Beller, T W Nygaard, C Tedesco, and S Kaul. 1989. “Effects of Successful Intravenous Reperfusion Therapy on Regional Myocardial Function and Geometry in Humans: A Tomographic Assessment Using Two-Dimensional Echocardiography.” Journal of the American College of Cardiology 13 (7) (June): 1506–13. http://www.ncbi.nlm.nih.gov/pubmed/2723266.

Traverse, Jay H. 2011. “Effect of Intracoronary Delivery of Autologous Bone Marrow Mononuclear Cells 2 to 3 Weeks Following Acute Myocardial Infarction on Left

Page 144: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

128

Ventricular Function<subtitle>The LateTIME Randomized Trial</subtitle>.” JAMA: The Journal of the American Medical Association 306: 2110. doi:10.1001/jama.2011.1670.

———. 2012. “Effect of the Use and Timing of Bone Marrow Mononuclear Cell Delivery on Left Ventricular Function After Acute Myocardial Infarction.” JAMA: The Journal of the American Medical Association 308 (22): 1. doi:10.1001/jama.2012.28726. http://jama.jamanetwork.com/article.aspx?doi=10.1001/jama.2012.28726.

Treiser, Matthew, Sascha Abramson, Robert Langer, and Joachim Kohn. 2010. “Degradable and Resorbable Biomaterials.” In Biomaterials Science: An Introduction to Materials in Medicine, edited by Buddy D. Ratner, Allan S. Hoffman, Frederick J. Schoen, and Jack E. Lemons, Third Ed., 179–195. Waltham, MA, USA: Elsevier. doi:10.1016/B978-0-08-087780-8.00021-8. http://dx.doi.org/10.1016/B978-0-08-087780-8.00021-8.

Tsur-Gang, Orna, Emil Ruvinov, Natalie Landa, Radka Holbova, Micha S Feinberg, Jonathan Leor, and Smadar Cohen. 2009. “The Effects of Peptide-Based Modification of Alginate on Left Ventricular Remodeling and Function after Myocardial Infarction.” Biomaterials 30 (2) (January): 189–95. doi:10.1016/j.biomaterials.2008.09.018. http://www.ncbi.nlm.nih.gov/pubmed/18849071.

Van Luyn, M J, I M Khouw, P B van Wachem, E H Blaauw, and J a Werkmeister. 1998. “Modulation of the Tissue Reaction to Biomaterials. II. The Function of T Cells in the Inflammatory Reaction to Crosslinked Collagen Implanted in T-Cell-Deficient Rats.” Journal of Biomedical Materials Research. http://www.ncbi.nlm.nih.gov/pubmed/9468048.

Van Slyke, Paul, Jennifer Alami, Daniel Martin, Michael Kuliszewski, Howard Leong-Poi, Michael V Sefton, and Dan Dumont. 2009. “Acceleration of Diabetic Wound Healing by an Angiopoietin Peptide Mimetic.” Tissue Engineering. Part A 15 (6) (June): 1269–80. doi:10.1089/ten.tea.2007.0400. http://www.ncbi.nlm.nih.gov/pubmed/18939935.

Venugopal, Jayarama Reddy, Molamma P Prabhakaran, Shayanti Mukherjee, Rajeswari Ravichandran, Kai Dan, and Seeram Ramakrishna. 2012. “Biomaterial Strategies for Alleviation of Myocardial Infarction.” Journal of the Royal Society, Interface / the Royal Society 9 (66) (January 7): 1–19. doi:10.1098/rsif.2011.0301. http://www.ncbi.nlm.nih.gov/pubmed/21900319.

Verma, Subodh, Paul W M Fedak, Richard D Weisel, Jagdish Butany, Vivek Rao, Andrew Maitland, Ren-Ke Li, Bikramjit Dhillon, and Terrence M Yau. 2002. “Fundamentals of Reperfusion Injury for the Clinical Cardiologist.” Circulation 105 (20) (May 21): 2332–6. doi:10.1161/01.CIR.0000016602.96363.36. http://circ.ahajournals.org/cgi/doi/10.1161/01.CIR.0000016602.96363.36.

Vunjak-Novakovic, Gordana, Nina Tandon, Amandine Godier, Robert Maidhof, Anna Marsano, Timothy P Martens, and Milica Radisic. 2010. “Challenges in Cardiac Tissue Engineering.” Tissue Engineering. Part B, Reviews 16 (2) (April): 169–87. doi:10.1089/ten.TEB.2009.0352.

Page 145: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

129

http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2946883&tool=pmcentrez&rendertype=abstract.

Wall, Samuel T, Joseph C Walker, Kevin E Healy, Mark B Ratcliffe, and Julius M Guccione. 2006. “Theoretical Impact of the Injection of Material into the Myocardium: A Finite Element Model Simulation.” Circulation 114 (24) (December 12): 2627–35. doi:10.1161/CIRCULATIONAHA.106.657270. http://www.ncbi.nlm.nih.gov/pubmed/17130342.

Wang, Haibin, Zhiqiang Liu, Dexue Li, Xuan Guo, F Kurtis Kasper, Cuimi Duan, Jin Zhou, Antonios G Mikos, and Changyong Wang. 2012. “Injectable Biodegradable Hydrogels for Embryonic Stem Cell Transplantation: Improved Cardiac Remodelling and Function of Myocardial Infarction.” Journal of Cellular and Molecular Medicine 16 (6) (June): 1310–20. doi:10.1111/j.1582-4934.2011.01409.x. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3227757&tool=pmcentrez&rendertype=abstract.

Wang, Haibin, Xuelian Zhang, Yanmin Li, Yitong Ma, Ye Zhang, Zhiqiang Liu, Jin Zhou, et al. 2010. “Improved Myocardial Performance in Infarcted Rat Heart by Co-Injection of Basic Fibroblast Growth Factor with Temperature-Responsive Chitosan Hydrogel.” The Journal of Heart and Lung Transplantation : The Official Publication of the International Society for Heart Transplantation 29 (8) (August): 881–7. doi:10.1016/j.healun.2010.03.016. http://www.ncbi.nlm.nih.gov/pubmed/20466563.

Wang, Limin, and Jan P Stegemann. 2011. “Glyoxal Crosslinking of Cell-Seeded Chitosan/collagen Hydrogels for Bone Regeneration.” Acta Biomaterialia 7 (6) (June): 2410–7. doi:10.1016/j.actbio.2011.02.029. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3085611&tool=pmcentrez&rendertype=abstract.

Wang, Tao, Xue-Jun Jiang, Qi-Zhu Tang, Xiao-Yan Li, Tao Lin, De-Qun Wu, Xian-Zheng Zhang, and Emmy Okello. 2009. “Bone Marrow Stem Cells Implantation with Alpha-cyclodextrin/MPEG-PCL-MPEG Hydrogel Improves Cardiac Function after Myocardial Infarction.” Acta Biomaterialia 5 (8): 2939–44. doi:10.1016/j.actbio.2009.04.040. http://www.ncbi.nlm.nih.gov/pubmed/19426843.

Wang, Tao, De-Qun Wu, Xue-Jun Jiang, Xian-Zheng Zhang, Xiao-Yan Li, Jin-Feng Zhang, Zhao-Bin Zheng, Renxi Zhuo, Hong Jiang, and Congxin Huang. 2009. “Novel Thermosensitive Hydrogel Injection Inhibits Post-Infarct Ventricle Remodelling.” European Journal of Heart Failure 11 (1) (January): 14–9. doi:10.1093/eurjhf/hfn009. http://www.ncbi.nlm.nih.gov/pubmed/19147452.

Warren, Luigi, Philip D Manos, Tim Ahfeldt, Yuin-han Loh, Hu Li, Frank Lau, Wataru Ebina, Pankaj K. Mandal, Zachary D. Smith, and Alexander Meissner. 2010. “Highly Efficient Reprogramming to Pluripotency and Directed Differentiation of Human Cells with Synthetic Modified mRNA.” Cell Stem Cell In Press (September): 1–13.

Page 146: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

130

doi:10.1016/j.stem.2010.08.012. http://linkinghub.elsevier.com/retrieve/pii/S1934590910004340.

Wei, Guo, Yue Guan, Ying Yin, Jialin Duan, Dan Zhou, Yanrong Zhu, Wei Quan, Miaomiao Xi, and Aidong Wen. 2013. “Anti-Inflammatory Effect of Protocatechuic Aldehyde on Myocardial Ischemia/Reperfusion Injury In Vivo and In Vitro.” Inflammation 36 (3) (June): 592–602. doi:10.1007/s10753-012-9581-z. http://www.ncbi.nlm.nih.gov/pubmed/23269534.

Whittingstall, Peter. 2003. “Dynamic or Oscillatory Testing of Complex Fluids.” In Current Protocols in Food Analytical Chemistry, edited by Ronald E. Wrolstad, Terry E. Acree, Eric A. Decker, Michael H. Penner, David S. Reid, Steven J. Schwartz, Charles F. Shoemaker, Denise Smith, and Peter Sporns, 1–11. Hoboken, NJ, USA: John Wiley & Sons, Inc. doi:10.1002/0471142913.fah0301s07.

Williams, D. F. 1987. Definitions in Biomaterials : Proceedings of a Consensus Conference of the European Society for Biomaterials, Chester, England, March 3-5, 1986. Vol. 4. Amsterdam, New York: Elsevier Science Ltd.

Wilson, Sean R, Gilbert H Mudge, Garrick C Stewart, and Michael M Givertz. 2009. “Evaluation for a Ventricular Assist Device: Selecting the Appropriate Candidate.” Circulation 119 (16) (April 28): 2225–32. doi:10.1161/CIRCULATIONAHA.109.850610. http://www.ncbi.nlm.nih.gov/pubmed/19398678.

World Health Organization. 2011a. “THE TOP TEN CAUSES OF DEATH: The 10 Leading Causes of Death by Broad Income Group, 2008.” http://www.who.int/mediacentre/factsheets/fs310_2008.pdf.

———. 2011b. “Global Status Report on Noncommunicable Diseases 2010.” World Health. Geneva, Switzerland. http://www.who.int/nmh/publications/ncd_report_full_en.pdf.

Wu, Jun, Faquan Zeng, Xi-Ping Huang, Jennifer C-Y Chung, Filip Konecny, Richard D Weisel, and Ren-Ke Li. 2011. “Infarct Stabilization and Cardiac Repair with a VEGF-Conjugated, Injectable Hydrogel.” Biomaterials 32 (2) (January): 579–86. doi:10.1016/j.biomaterials.2010.08.098. http://www.ncbi.nlm.nih.gov/pubmed/20932570.

Yamamoto, T, N Suto, T Okubo, a Mikuniya, H Hanada, S Yagihashi, M Fujita, and K Okumura. 2001. “Intramyocardial Delivery of Basic Fibroblast Growth Factor-Impregnated Gelatin Hydrogel Microspheres Enhances Collateral Circulation to Infarcted Canine Myocardium.” Japanese Circulation Journal 65 (5) (May): 439–44. http://www.ncbi.nlm.nih.gov/pubmed/11348050.

Yamaoka, Tetsuji, Yasuhiko Tabata, and Yoshito Ikada. 1993. “Body Distribution Profile of Polysaccharides after Intravenous Administration.” Drug Delivery 1 (1) (January): 75–82. doi:10.3109/10717549309031345. http://www.informapharmascience.com/doi/abs/10.3109/10717549309031345.

Page 147: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

131

Yang, Lei, Mark H Soonpaa, Eric D Adler, Torsten K Roepke, Steven J Kattman, Marion Kennedy, Els Henckaerts, et al. 2008. “Human Cardiovascular Progenitor Cells Develop from a KDR+ Embryonic-Stem-Cell-Derived Population.” Nature 453 (7194): 524–8. doi:10.1038/nature06894. http://www.ncbi.nlm.nih.gov/pubmed/18432194.

Yeo, Yoon, Wenliang Geng, Taichi Ito, Daniel S Kohane, Jason A Burdick, and Milica Radisic. 2007. “Photocrosslinkable Hydrogel for Myocyte Cell Culture and Injection.” Journal of Biomedical Materials Research. Part B, Applied Biomaterials 81 (2): 312–22. doi:10.1002/jbm.b.30667. http://www.ncbi.nlm.nih.gov/pubmed/16969828.

Yu, Jiashing, Karen L Christman, Eric Chin, Richard E Sievers, Maythem Saeed, and Randall J Lee. 2009. “Restoration of Left Ventricular Geometry and Improvement of Left Ventricular Function in a Rodent Model of Chronic Ischemic Cardiomyopathy.” The Journal of Thoracic and Cardiovascular Surgery 137 (1) (January): 180–7. doi:10.1016/j.jtcvs.2008.08.036. http://www.ncbi.nlm.nih.gov/pubmed/19154923.

Yu, Jiashing, Yiping Gu, Kim T Du, Shirley Mihardja, Richard E Sievers, and Randall J Lee. 2009. “The Effect of Injected RGD Modified Alginate on Angiogenesis and Left Ventricular Function in a Chronic Rat Infarct Model.” Biomaterials 30 (5): 751–6. doi:10.1016/j.biomaterials.2008.09.059. http://www.ncbi.nlm.nih.gov/pubmed/19010528.

Yue, T.-L., C. Wang, J.-L. Gu, X.-L. Ma, S. Kumar, J. C. Lee, G. Z. Feuerstein, H. Thomas, B. Maleeff, and E. H. Ohlstein. 2000. “Inhibition of Extracellular Signal-Regulated Kinase Enhances Ischemia/Reoxygenation-Induced Apoptosis in Cultured Cardiac Myocytes and Exaggerates Reperfusion Injury in Isolated Perfused Heart.” Circulation Research 86 (6) (March 31): 692–699. doi:10.1161/01.RES.86.6.692. http://circres.ahajournals.org/cgi/doi/10.1161/01.RES.86.6.692.

Zammaretti, Prisca, and Marisa Jaconi. 2004. “Cardiac Tissue Engineering: Regeneration of the Wounded Heart.” Current Opinion in Biotechnology 15 (5) (October): 430–4. doi:10.1016/j.copbio.2004.08.007. http://www.ncbi.nlm.nih.gov/pubmed/15464373.

Zhang, M, D Methot, V Poppa, Y Fujio, K Walsh, and C E Murry. 2001. “Cardiomyocyte Grafting for Cardiac Repair: Graft Cell Death and Anti-Death Strategies.” Journal of Molecular and Cellular Cardiology 33 (5) (May): 907–21. doi:10.1006/jmcc.2001.1367. http://www.ncbi.nlm.nih.gov/pubmed/11343414.

Zhang, Shuguang, Todd C Holmes, Curtis Lockshin, and Alexander Rich. 1993. “Spontaneous Assembly of a Self-Complementary Oligopeptide to Form a Stable Macroscopic Membrane.” Proceedings of the National Academy of Sciences of the United States of America 90 (April): 3334–3338.

Zhang, Xuelian, Haibin Wang, Xiang Ma, Azhati Adila, Baozhu Wang, Fen Liu, Bangdang Chen, Changyong Wang, and Yitong Ma. 2010. “Preservation of the Cardiac Function in Infarcted Rat Hearts by the Transplantation of Adipose-Derived Stem Cells with Injectable Fibrin Scaffolds.” Experimental Biology and Medicine (Maywood, N.J.) 235 (12)

Page 148: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

132

(December): 1505–15. doi:10.1258/ebm.2010.010175. http://www.ncbi.nlm.nih.gov/pubmed/21127347.

Zhang, Yan, Jean N DaSilva, Stephanie Thorn, Robert a DeKemp, Rob S Beanlands, Marc Lamoureux, Marc Ruel, and Erik J Suuronen. 2008. “Collagen-Based Matrices Improve the Delivery of Transplanted Circulating Progenitor Cells: Development and Demonstration by Ex Vivo Radionuclide Cell Labeling and in Vivo Tracking with Positron-Emission Tomography.” Circulation. Cardiovascular Imaging 1 (3) (November): 197–204. doi:10.1161/CIRCIMAGING.108.781120. http://www.ncbi.nlm.nih.gov/pubmed/19808543.

Zhao, Zhi-Qing, John D Puskas, Di Xu, Ning-Ping Wang, Mario Mosunjac, Robert a Guyton, Jakob Vinten-Johansen, and Robert Matheny. 2010. “Improvement in Cardiac Function with Small Intestine Extracellular Matrix Is Associated with Recruitment of C-Kit Cells, Myofibroblasts, and Macrophages after Myocardial Infarction.” Journal of the American College of Cardiology 55 (12) (March 23): 1250–61. doi:10.1016/j.jacc.2009.10.049. http://www.ncbi.nlm.nih.gov/pubmed/20298933.

Zimmermann, W.-H. 2001. “Tissue Engineering of a Differentiated Cardiac Muscle Construct.” Circulation Research 90 (2) (December 13): 223–230. doi:10.1161/hh0202.103644. http://circres.ahajournals.org/cgi/doi/10.1161/hh0202.103644.

Zimmermann, Wolfram Hubertus, Ivan Melnychenko, Gerald Wasmeier, Michael Didié, Hiroshi Naito, Uwe Nixdorff, Andreas Hess, et al. 2006. “Engineered Heart Tissue Grafts Improve Systolic and Diastolic Function in Infarcted Rat Hearts.” Nature Medicine 12 (4) (April): 452–8. doi:10.1038/nm1394. http://www.ncbi.nlm.nih.gov/pubmed/16582915.

Zimmermann, Wolfram-Hubertus, Ivan Melnychenko, and Thomas Eschenhagen. 2004. “Engineered Heart Tissue for Regeneration of Diseased Hearts.” Biomaterials 25 (9) (April): 1639–1647. doi:10.1016/S0142-9612(03)00521-0. http://linkinghub.elsevier.com/retrieve/pii/S0142961203005210.

Page 149: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

133

Appendix A. Reaction schematics and chemical structures

Figure A-1: Polyelectrolytic complexation of chitosan and collagen

(A) Collagen has both carboxyl and amine groups, although it has many more negatively charged carboxyl groups (pI~4.8). In acidic medium most carboxyl groups are quenched and sparse charge along collagen fibers exist. Increasing the pH increases ionic strength and attraction between collagen fibers, and causes spontaneous organization through both covalent bonding and ionic interaction between collagen fibers (only ionic interactions shown). As the reaction is endothermic, an increase in temperature greatly assists and speeds up the gelation process. (B) Addition of chitosan to a collagen mixture greatly increases the number of free, charged amines but does not interfere with the spontaneous organization of collagen fibers (which promotes the formation of a hydrogel). Chitosan associates with collagen fibers through ionic interaction, forming thicker chitosan-collagen complexed fibers. (C) Further addition of chitosan creates even thicker overall chitosan-collagen fibers with chitosan essentially quenching all available negative charge on collagen fibers, and further sequesters collagen fibers. (A-C) Collagen only hydrogels form highly interconnected networks with thinner individual fibers, meaning the hydrogels are easily deformed but do not break apart easily. The addition of chitosan creates larger overall fibers, however reduces the number of cross-links that can be formed between network fibers. The resultant hydrogel resists deformation more than pure collagen and retains network strength to resist tearing. Too much chitosan essentially quenches all available carboxyl groups on collagen, forming very clustered fibers (thick), but very low fibre network interconnectivity. The resultant hydrogel is therefore very brittle. (Inspired by Berger et al. (2004)).

Page 150: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

134

Figure A-2: Modified chitosans used

Figure A-3: EDC/Sulfo-NHS reaction chemistry for conjugation of biomolecules

Page 151: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

135

Figure A-4: QHREDGS conjugation to PEG

Page 152: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

136

Appendix B. In vitro construct CM distribution

Figure B-1: In vitro cell leakage and migration Brightfield images of cell/gel constructs seeded in 12 well tissue culture plates at 1hr (30 minutes post

gelation) and after 120hrs in culture. Low magnification image (top row) and high magnification (bottom) show minimal cell leakage out of the gel during seeding and gelation as well as after 5 days in culture. Scale bars=200µm top, 50µm bottom left, 100µm bottom right.

Page 153: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

137

Figure B-2: Quantification of in vitro construct viability (A) Live/dead (PI, red/CFDA, green) images of Control and High peptide constructs grown in vitro after

120 hrs. Images were taken at the edge of constructs and in the very center (middle). Scale bars=100µm. (B) Live/dead quantification was performed using ImageJ and shows no significant differences in viability between groups at the two regions, however the edge region has significantly higher viability then in the center of constructs (N=4/group). Data expressed as Mean±SD, ** P<0.01, *** P<0.0001.

Page 154: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

138

Figure B-3: High QHREDGS hydrogel constructs contain elongated cardiomyocytes with visible cross-striations

High magnification cardiac troponin T immunostaining of in vitro formalin fixed paraffin embedded tissue constructs. cTnT (green), DAPI (blue). Arrow indicates cross-striations. Scale bars=25µm.

Page 155: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

139

Appendix C. Utility of QHG213H hydrogel for MI treatment published

supplementary data

Table C-1: Genes and primers used in RT-qPCR

Gene Abbr. Forward Reverse

Glyceraldehyde 3-phosphate dehydrogenase

GAPDH GCTGAGAATGGGAAGCTG AGTGATGGCATGGACTGT

B-cell lymphoma 2 BCL2 CTGGTGGACAACATCGCTCTG GGTCTGCTGACCTCACTTGTG

B-cell lymphoma extra large

BCL-XL CATATAACCCCAGGGACAGC GTCATGCCCGTCAGGAAC

Phosphatidylinositol-4,5-bisphosphate 3-kinase

PI3K AGCCACAGGTGAAAATACGG TTTTCTTTCCGCAACAGCTT

BH3 interacting-domain death agonist

BID AGCAGGTGATGAACTGGACC AGACGTCACGGAGCAGAGAT

BCL2-associated X protein BAX TCCAGGATCGAGCAGA AAGTAGAAGAGGGCAACC

Caspase 3 CASP-3 AACCTCAGAGAGACATTCATGG GAGTTTCGGCTTTCCAGTCA

Caspase 9 CASP-9 CTCCTGCGGCGATGC CCACTGGGGTGAGGTTTC

Receptor interacting protein 1

RIP1 GTGGTGAAGCTACTGGGCAT AGGAAGCCACACCAAGATCG

Receptor interacting protein 3

RIP3 CACACCAGCAGGGACATCAT TTTGGTGCGTTCCAGGTGTA

Tumor necrosis factor α TNFα TGCCTCAGCCTCTTCTCATT GCTTGGTTTGCTACGAC

Interleukin 1β IL-1β CTGTGACTCGTGGGATGATG GGGATTTTGTCGTTGCTTGT

Interleukin 6 IL6 TCCTACCCCAACTTCCAATGCTC TTGGATGGTCTTGGTCCTTAGCC

Intercellular adhesion molecule 1

ICAM1 ACAAGTCCGTGCCTTTAGCTC GATCACGAAGCCCGCAATG

Vascular cell adhesion molecule 1

VCAM1 TAAGTTACACAGCAGTCAAATGGA CACATACATAAATGCCGGAATCTT

Matrix metalloproteinase 2 MMP2 AAGTCTGAAGAGTGTGAAGT GTGAAGGAGAAGGCTGATT

Transforming growth factor β1

TGFβ1 TGCTTCAGCTCCACAGAGAA TGGTTGTAGAGGGCAAGGAC

Monocyte chemoattractant proetein 1

MCP1 TTCACAGTTGCTGCCTGTAG TCTGATCTCACTTGGTTCTGG

Macrophage migration inhibitory factor 1

MIF1 CCA GGA CCG CAA CTA CAG CAA GGG CTC AAG CGA AGG TGG AAC

Interleukin 10 IL10 TTGAACCACCCGGCATCT CCAAGGAGTTGCTCCCGTTA

Tissue inhibitor of matrix metalloproteinase 1

TIMP1 CGCGGGCCGTTTAAGG GTTTGCAAGGGATGGCTGAA

Vascular endothelial growth factor

VEGF CCCACGACAGAAGGAGAGCA GCACACAGGACGGCTTGAA

Cardiac troponin T cTnT AGGCTCTTCATGCCCAAC TGTTCTCGAAGTGAGCCTC

α myosin heavy chain αMHC TGATGACTCCGAGGAGCT TGACACAGACCCTTGAGCA

Β myosin heavy chain β MHC CCTCGCAATATCAAGGGAAA TACAGGTGCATCAGCTCCA

Page 156: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

140

Integrin linked kinase ILK CCCCACGTGTGTAAGCTCAT AGCCCCATAACTGGGGTAGT

Integrin subunit α5 α5 CCAGAGCAAGAGCCGGATAG GCGTTCTTGTCACCCAGGTA

Integrin sub-unit β1 β1 CAATGAGGGTCGTGTTGGGA CCGTTGGACCTATCGCAGTT

Table C-2: Description of studies considered for comparative-analysis

Group/Paper Control Treatment Treatment

Time (After MI)

Monitoring Time (After Treatment)

(Christman et al. 2004) 0.5% BSA in PBS

Skeletal myoblasts in Fibrin glue 1 weeks 5 weeks

(Memon et al. 2005) DMEM injection

Myoblast cell sheets 2 weeks 8 weeks

(Dai et al. 2005) Saline injection Bovine dermal collagen 1 week 6 weeks

(Landa et al. 2008) Saline injection Ca-cross linked alginate 1 week 8 weeks

(Tsur-Gang et al. 2009) Saline injection Ca-cross linked alginate 1 week 8 weeks

(Lu et al. 2009) PBS mESCs in temperature responsive chitosan-β-GP hydrogel

1 week 4 weeks

(Wang et al. 2009) DMEM/F12 medium

Bone marrow stem cells in α-CD/MPEG-PCL-MPEG hydrogel

1 week 4 weeks

(Fujimoto et al. 2009) PBS Thermoresponsive poly(NIPAAm-co-AAC-co-HEMAPTMC)

2 weeks 8 weeks

(Wu et al. 2011) PBS VEGF conjugated PVL-b-PEG-PVL hydrogel

1 week 4 weeks

(Kraehenbuehl et al. 2011)

PBS hESC derived ELCs & SMLCs in Tβ4-PEG hydrogel

1 hour 6 weeks

Our work PBS Chitosan-collagen-QHREDGS hydrogel

Imm. 6 weeks

Page 157: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

141

Figure C-1: Quantification of hydrogel lifespan Whole heart fluorescence was quantified from homogenized hearts excised 1 hr and 14 days after injection with Dylight-800

labelled chitosan-collagen gel. Assuming that after 1 hr the injected material has gelled fully and that unconjugated dye has been removed along with un-gelled material by this time, the ~60% reduction in fluorescence on day 14 is indicative that ~40% of the labelled hydrogel localized in the heart remained after two weeks.

Page 158: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

142

Figure C-2: Histological staining 6 weeks post MI (A) Mason’s trichrome, (B) smooth muscle actin (SMA), (C) CD31, and (D) Factor VIII (FVIII) stained

sections of hearts excised 6 weeks post MI. Black outlined sections in (A) are enlarged in (B-D). A large collagenous scar is seen in all groups (A), but there appears to be more SMA, CD31 and FVIII staining in the Control gel and QHG213H gel groups as compared to MI Only (PBS) group, suggesting increased vascularization (B-D). Scale bars = 1 mm (A), 50 µm (B-D), N=7/group.

Page 159: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

143

Figure C-3: RT-qPCR of MI and border zone tissues from hearts excised 24 hrs post MI Hearts subjected to MI and immediate treatment were excised 24 hrs later, sectioned into MI, border, and remote regions,

snap frozen then ground and used for RT-qPCR. Overall, 27 genes conventionally associated with apoptosis, necroptosis (Necrop), survival, anti- and pro-inflammatory responses, as well as cardiac specific genes were assessed. Data was standardized in samples to reflect changes in gene expression in the progressing border zone to the established MI zone. No significance was determined between groups. Data expressed as Mean±SD, N=5/group.

Page 160: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

144

Figure C-4: Western blot analysis of border zone tissue from hearts excised 24 hrs post MI

Western blot analysis of border zone heart tissues showed no differences in the levels of ILK or phospho-MAPK (PMAPK) protein expression between groups. The MAPK protein expression was significantly increased in the QHG213H gel group compared to the Control gel and MI Only (PBS) groups. Data expressed as Mean±SD, * P<0.05, N=5/group.

Page 161: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

145

Figure C-5: Reported cardiac morphological and functional data from studies used in meta-analysis The differences between Control treatment (Saline or PBS injection) and therapy (hydrogel injection, cell treatment, patches)

in various measures of cardiac remodeling and function after acute MI as reported by difference studies are illustrated (Top panel, and bottom left). The relative change in the outcome of these measures between all groups is illustrated (bottom right).

Page 162: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

146

Appendix D. Unpublished rat MI model supplementary material

After sacrificing, excising, and fixing hearts it was fairly obvious that the MI Only and

Control gel groups were very dilated in comparison to the Sham operated heart (consistent with

the results seen in the mouse MI model described earlier), but that the QHG213H gel injected

heart appeared much more similar in size (Figure D-1 A). Two cross sections of each heart were

taken in the area just below the suture and illustrate the extent of MI in each group (white

scarring, Figure D-1 B) and is made further evident in the Mason’s trichrome staining done on

all sections (Figure D-1 C). The blue collagenous scar deposition seen in the trichrome stained

sections seemed much less apparent in the QHG213H gel group compared to MI Only and

Control gel, and overall tissue cross section appeared very similar to that of the healthy heart

(Sham).

Figure D-1: Heart morphology & histology 3Wk post MI A LAD rat MI model was used to assess hydrogel utility in mitigating remodeling. (A) Ligation site is

clearly marked by the suture and surrounding MI zone (white), and remodeling extent is further apparent in tissue cross sections (B). Scale bars=1mm (A-B). (C) Trichrome images show tissue remodeling and extent of infarct. Scale bars=1mm. Black outlined sections are area of focus for Figure D-2. (D). N=2 Sham, N=6 rest.

Staining for H&E, SMA, F8, and CD31 was also performed on all 3Wk sections (Figure D-2

A-D respectively). SMA expression in the MI zone of all test groups appeared greater than

Page 163: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

147

endogenous expression seen in the Sham hearts, although expression in the QHG213H group

seemed localized to the gel injection site and appeared to be much less expression than the MI

Only and Control gel groups (Figure D-2 A). F8 and CD31 expression, both endothelial cell

markers used to identify neovascularization, were similar within groups and were consistent with

the trend observed in the SMA stained sections (Figure D-2 C&D). Interestingly, however, in the

QHG213H gel group both F8 and CD31 staining appeared limited to the gel border and

infiltrating cells were not seen within the gel as seen with the SMA staining.

Figure D-2: Further histological staining 3Wks post MI (A) H&E, (B) SMA, (C) F8, & (D) CD31staining illustrate infiltrating myofibroblasts and neovascularization

in the MI zone. Marked differences can be seen between all test groups and the healthy, Sham tissue. MI Only and Control gel groups have very similar expression profiles for all stains, but QHG213H gel group appears to have much more normal expression of all markers, although clear infiltration of SMA + cells into the injection site can be seen. Scale bars=50µm, N=2 Sham, N=6 rest.

Many different metrics were used in an attempt to quantify the extent of remodeling at 3Wks

post MI. First, left ventricle (LV) wall thickness in the MI zone was measured at numerous

Page 164: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

148

points (~5 locations per sample) to get an average value per section, then sample, and mean

value per group (Figure D-3 A&B). An obvious trend was seen with the MI Only group having

the lowest MI zone LV wall thickness, followed by the Control and then QHG213H gel groups;

however no significant difference between measurements was apparent Figure D-3 B). Adobe

Photoshop CS3 was used quantify MI scar area and total tissue area from trichrome stained

images (shaded areas, Figure D-3 C&D). Values were used to get a measure of fractional scar

coverage (MI scar area/total tissue area) between groups, and compare tissue loss (or

maintenance) by comparing mean total tissue area between groups (Figure D-3 D&F). The trend

of the MI Only group having the greatest fractional scar coverage and lowest mean total tissue

area, and the QHG213H gel group having the best was consistent with the MI zone LV wall

thickness data, however a significant difference was only seen between MI Only and QHG213H

gel groups in terms of mean total tissue area (p=0.007). Both the interior and exterior scar extent

were also measured as a final metric of remodeling at 3Wks, as different groups report many

different metrics and therefore they were performed for completeness (Figure D-4). Both

measures maintained the overall trend seen in Figure D-3, however neither are statistically

significant.

While only one measure (total tissue area) showed statistical significance in the ability of

QHG213H gel to limit remodeling at 3Wks, this time point was only an intermediate and it was

not entirely surprising we did not see more significance between groups. As previously stated, in

both mouse and rat MI models pathological remodeling is not complete until six weeks post MI,

therefore while the trends observed at 3Wks were promising the same metrics at 6Wks were the

truly important measures (Patten et al. 1998; Krzemiński et al. 2008).

Page 165: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

149

Figure D-3: 3Wk MI quantification measurements (A) MI zone wall thickness measurements were made at various spots (~5) along the scar length and

averaged to get a measurement per sample and (B) mean LV wall thickness between groups. (C) MI scar area was measured and normalized to overall heart tissue area per sample (E) to get a measure of (D) fractional scar coverage between groups, and to compare (F) the maintenance or loss of heart tissue. **P<0.01, one way ANOVA, Bonferroni post tests. Data expressed as Mean±SD, N=6/group.

Page 166: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

150

Figure D-4: 3Wk MI scar extent assessment Interior (A, red line) compared to LV capacity (perimeter, blackline). Shows clear trend towards lower extent

in QHG213H group (B) which correlates with rest of data. Exterior, same thing (C&D). Data expressed as Mean±SD, N=6/group.

Stained sections from the hearts excised 6 weeks post treatment were imaged using a slide

scanner at Princess Margaret Hospital, allowing for much higher resolution and greater

measuring accuracy when quantifying the extent of remodeling from 6Wk trichrome images

compared to those from 3Wk. The same metrics were used to quantify remodeling extent at

6Wks as done at 3Wks and the same trend remained with QHG213H gel group having the

highest scar thickness, lowest fractional scar tissue coverage, and highest LV tissue area,

followed by Control gel and then MI Only (Figure D-5 A-C, respectively) however the results

were not significant as seen with the quantification done on the gross tissue sections (Figure 4-6).

Two further metrics were assessed; the relative scar thickness compared to healthy LV and the

Page 167: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

151

infarct expansion index (Figure D-5 D&E respectively). The two graphs show the expected trend

of QHG213H gel performing best, followed by Control gel and finally MI Only (PBS), but there

remained a lack of significance between groups.

Figure D-5: 6Wk MI quantification measurements (from histology pictures)

Page 168: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

152

Measurements done in the same manner as that for 3Wk samples, however with higher resolution for greater accuracy and more MI zone wall thickness measurements (>10 per sample) to get overall (A) MI zone scar thickness, (B) fractional MI zone scar tissue coverage (with respect to left ventricle (LV) area only), and (C) total LV tissue area. LV area was considered for the 6Wk samples as some RVs were missing from sections, and it was much easier to eliminate RV than it was for 3Wk samples. Data expressed as Mean±SD, N=4/group.

Quantifying differences in vasculature in the MI zone at 3Wks post MI was done using SMA,

F8, and CD31 stained sections. Mean vessel density and diameters between groups were

compared for mature SMA expressing vasculature at 3Wks (Figure D-6 A), or total vasculature

(Figure D-6 B). While images may have hinted at a trend being apparent, the data suggested

there were no differences in vascularization between groups, with large standard deviations

indicating there was a large difference within the groups themselves.

Figure D-6: Vascularization within the MI scar zone at 3Wks post infarct Vascularization within the MI zone was quantified at 3Wks. Vessel density and diameter were quantified

from SMA stained sections to assess mature vasculature within the MI zone (A) and from F8 (B) stained sections for neovascularization. No discernible trend can be seen and there is no significant difference between groups. Data expressed as Mean±SD. N=5 MI Only, N=6 Control, N=3 QHG213H.

The TUNEL label and enzyme are used to label DNA strand breaks for detecting and

quantitating late stage apoptotic cell death at a single cell level. It was used to determine CM cell

death at the MI border zone and see if the functional and morphological improvements seen

could be due to reduced apoptosis and maintenance of healthy CMs. TUNEL/cTNT staining

showed a limited number of apoptotic cells in all groups at 3Wks (Figure D-7 A), and perhaps a

Page 169: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

153

reduced number in the Control group compared to MI Only and QHG213H groups, but no

definitive conclusions could be made (Figure D-7 B&C). Counting was done using a macro

developed using Olympus CellSens Dimension software tested on TUNEL controls (Figure D-8).

Figure D-7: Apoptosis in the MI boundary zone at 3Wk Late stage apoptosis was assessed by staining sections for TUNEL (red), cTnT (green), and DAPI nuclear

stain (blue). Representative images (A, 40X magnification) from each group show sparse TUNEL positive cells, and there is no difference in number of TUNEL positive cells per image (B) or in fraction of TUNEL positive cells (C). Data expressed as Mean±SD, N=3/group. Scale bars=20µm.

The raw counts made for both DAPI+ and TUNEL+ cells from the 6Wk heart sections are

seen in Figure D-9. The counts were made from multiple images taken per section at two varying

magnifications, 40X (Figure D-9 A) and 10X (Figure D-9 B) as a method to test variability in

counts at the two magnifications, which it appeared there was not. Sample images used to

optimize the macro for counting are displayed in Figure D-10 A, with associated control macro

optimized counts for both DAPI+ and TUNEL+ cells shown in Figure D-10 B. Again, the macro

was optimized for images taken at 40X or 10X magnification to ensure no differences in relative

cell counts between sections with varying magnification.

Page 170: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

154

Figure D-8: 3Wk TUNEL/cTnT staining controls TUNEL (red), cTnT (green), and DAPI nuclear stain (blue). (A) Representative 40X magnification images of

caspase treated +ve Control, and TUNEL label added only –ve Control. Obvious differences are seen and images were used to calibrate the macro used for counting TUNEL+ve and DAPI+ve cells (B). Data expressed as Mean. Scale bars=20µm.

Figure D-9: 6Wk TUNEL/cTnT staining raw counts

Page 171: A Peptide Modified Hydrogel Therapy for Acute Myocardial ... · ii A Peptide Modified Hydrogel Therapy for Acute Myocardial Infarction Lewis A. Reis Doctor of Philosophy The Institute

155

Raw TUNEL positive and Dapi positive cell numbers per image, used to make Figure 4-8, at (A) 40X and (B) 10X magnifications. Data expressed as Mean±SD, N=4/group.

Figure D-10: 6Wk TUNEL/cTnT staining controls TUNEL (red), cTnT (green), and DAPI nuclear stain (blue). Clear TUNEL positive cells seen at both 40X

and 10X caspase treated positive control (A), and none seen in the negative control images. Images were used to optimize macro for counting TUNEL and DAPI +ve cell numbers (B). Data expressed as Mean. Scale bars=20µm 40X, 100µm 10X.