You are on page 1of 376

NATURALLY DERIVED SCAFFOLDS FOR TREATMENT

OF VOLUMETRIC MUSCLE LOSS

BY

HANNAH BESSER BAKER

A Dissertation Submitted to the Graduate Faculty of

WAKE FOREST UNIVERSITY SCHOOL OF ARTS AND SCIENCES

in Partial Fulfillment of the Requirements

for the Degree of

DOCTOR OF PHILOSOPHY

Biomedical Engineering

August, 2015

Winston-Salem, NC

Approved By:

George J. Christ, Ph.D., Advisor and Chair

Graça Almeida-Porada, M.D., Ph.D.

Aaron S. Goldstein, Ph.D.

Robert W. Grange, Ph.D.

David L. Kaplan, Ph.D.

Aaron M. Mohs, Ph.D.


DEDICATION

This dissertation is dedicated to my mother, Sheena Besser Baker, whose untiring work

ethic and resilience have been my inspiration and for having instilled in me a stubborn

independence and resourcefulness for which I am truly grateful.

ii
ACKNOWLEDGEMENTS

This work would not have been possible without the guidance and support of many

people over the last five years and for that I am sincerely grateful. I would like to first

acknowledge and thank my advisor Dr. George Christ for his support and mentorship, for

providing me with many amazing opportunities to work first hand on several aspects of

translational research, and for helping me develop as a researcher by holding me to a high

standard.

I would also like to thank my committee members: Dr. Aaron Mohs, Dr. Graça

Almeida-Porada, Dr. Aaron Goldstein, Dr. Robert Grange, and Dr. David Kaplan for their

input on this work and support over the last couple years. I would especially like to

acknowledge Dr. Mohs for serving as my interim advisor and Dr. Koudy Williams for

taking over as principal investigator on ongoing animal protocols in the months between

Dr. Christ’s relocation to UVa and my defense. Further I would also like to acknowledge

and thank WFIRM for providing me with the opportunity to be a NIBIB T32 fellow and

for the support during my fellowship from Dr. Christ, Dr. Anthony Atala, and Joan

Schanck. I would also like to thank Dr. JP McQuilling for his comradery during our time

spent together as fellows.

Many people at WFIRM played crucial roles in completing this research. I would

like to thank the administrators who have helped make everything run smoothly, especially

Joanne Gray and Donna Tucker. A special thanks is owed to the WFIRM technicians and

especially Cathy Mathis and Cindy Zimmerman whose expertise and support in histology

and immunohistochemistry has helped tremendously. I would also like to thank the surgery

iii
core, the animal care staff, and especially Cindy Miller, Tammy Cockerham, and Cara

Clouse for assistance, guidance, and advice over the years on many animal protocols.

I owe my past and present lab family at WFIRM and UVa many thanks for

supporting me in work and life: Drs. Ben Corona, Catherine Ward, Masood Machingal,

and David Burmeister, Ashley Starr, Christine Koval, Dr. Mevan Siriwardane, Dr. Juliana

Passipieri, Manasi Madhavkar, Chris Bergman, Benjamin Rowe, Mona Zarif-Pour, and Dr.

John Scott, and collaborators and friends in the Marini lab: Kristina Stumpf and Lysette

Mutkus. A special thanks to Dr. John Scott for providing many weeks of assistance on the

rat LD project. I would also like to specially acknowledge Manasi Vadhavkar who

provided countless hours of assistance in cell culture and animal work and for teaching me

many lab techniques. Another very special thanks is owed to Chris Bergman, who assisted

in several facets of every project in this dissertation and especially for having endured

hundreds of hours of organ bath testing for the rat LD project, for which I am extremely

appreciative.

I am exceptionally thankful to have conducted these studies start to finish alongside

Dr. Juliana Passipieri. I have learned so much from Juliana who has a remarkable skill set

and knowledge base. Together we have been an effective and efficient team. I am going to

miss working with Juliana tremendously.

I am very grateful for my friends and extended lab family in the UVa Christ Lab,

Danny Lovell, Ellen Mintz, and Dr. Juliana Passipieri, for their assistance and support

throughout the last several months as I was traveling between Winston-Salem,

Charlottesville, and Baltimore. A very special thank you to Danny and the outstanding UVa

undergrads for conquering an unfathomable amount of histology for the rat LD project.

iv
My time in graduate school would not have been nearly as enjoyable without the

support of my family and the friendships I have made. I would like to acknowledge my

family members for their love and support throughout my grad school years: my mother

Sheena Baker, my father Steve Baker, my step-mother Cheryl Baker, and my brothers

and sisters: Mark Nater, Kristan Nater, Sarah and my brother in law Mike Burns, Amber

and my brother in law Jimmar Wilson, and Rosemary Baker.

I would also like to recognize the great friendships and endless support of fellow

graduate students which have made this time so wonderfully fun and deserve

acknowledgement: Anthony and Jess Santago, Kiks Kowalczewski, JP and Alyssa

McQuilling, Nick Vavalle and Heather Wallin, Tanner Hill, Jill and David Hobson, Jenni

and Chris Jordan, Aleks and Steph Skardal, Megan Johnston, Chris and Sarah Jo Bergman,

Roche De Guzman, Dipen Vyas, Matthew Brovold and Samantha Caldwell, Ashley

Wagoner, Daniel Deegan, Shaida Moghaddassi, Emmy McKee, Sean and Jess Murphy,

Peter Alexander and Amie Severino, Leah and Chris Stuart, Cale and Stephanie

Fahrenholtz, Brittany Eldridge, Jessica Swanner, Hesh Devarasetty, Sam Pendergraft,

Colin Fennelly, Stephen Rego and Amritha Kidiyoor, Kadie Vanderman, Saloomeh

Mokhtari, Elie Zakhem, and Susan Zhao.

I have been incredibly lucky to have great roommates and amazing friends in Emma

Moran and Doug Evans, Liz Graham and Kevin Gurysh, and Danny Lovell who have made

my time in Winston so much fun. I also owe several friends outside of Winston a big thank

you for their continuous support academically and in life over the years: Allison

Hulchanski, Matt Bailey, Jessica Marino, David Axe, Eric Weissmann, Shekhar Gadkaree,

Dan Mendelson, and Monica Lentz.

v
I would like to give a special shout out to Daniel Stovall and Shreya Raghavan for

being such great and supportive friends and helping me generously throughout the process

of applying for post graduate opportunities and beyond and to Jes Bolduc for being such a

great listener throughout this crazy time of life.

Finally, I want to specially acknowledge Abritee Dhal and Mike Kelen. Abritee has

truly kept me sane throughout our friendship in grad school and Mike has been wonderfully

understanding, patient, and helpful over the last two years. Abritee and Mike have been so

selflessly supportive especially during these last incredibly stressful and crazy couple of

years and I thank them both dearly for it.

vi
TABLE OF CONTENTS

List of Figures and Tables………….…………………………..……………………….viii

List of Abbreviations……...…………………………………...………………………..xvi

Abstract…...……………………………………………………………………………..xix

CHAPTER

I. Introduction……...……………………………………………..………………….1

II. Cell and Growth Factor Loaded Keratin Hydrogels for Treatment of Volumetric

Muscle Loss in a Mouse Model……………………………………………….....60

III. Keratin Hydrogel Enhances In Vivo Skeletal Muscle Function in a Rat Model of

Volumetric Muscle Loss………………………………………………………....99

IV. Investigation of Tissue Engineered Muscle Repair Constructs for Use in Cleft Lip

Secondary Revision Procedures in a Rat Model………………………………..134

V. Summary, Conclusions, and Future Directions…………………………….......241

APPENDIX

1. Implantation of In Vitro Tissue Engineered Muscle Repair Constructs and Bladder

Acellular Matrix Partially Restore In Vivo Skeletal Muscle Function in a Rat Model

of Volumetric Muscle Loss Injury……………….………………...……...……269

2. Ethical Considerations in Tissue Engineering Research: Case Studies in

Translation……………………………………………………………………..308

4. Growth Factor Release from Keratin Hydrogel Formulation Used in Mouse LD

and Rat TA Models (Unpublished Data from KeraNetics, LLC)…..................351

Curriculum Vitae……..…………………………………………………………..……353

vii
LIST OF FIGURES AND TABLES

CHAPTER I: Introduction

Figure 1. Skeletal Muscle Form and Histology of Immature and Mature Myofibers........6

Figure 2. Skeletal Muscle Injury and Repair in VML…………………………………....8

Table I. Summary of Key Growth Factors and Trophic Factors involved in Skeletal

Muscle Repair……………………………………………………………………………12

Figure 3. Platform of Technologies for TE Skeletal Muscle…………………………....26

Figure 4. Keratin Structure.………………………………………………………….…..28

Figure 5. Growth Factor Release from 20% Keratin Gels………………………………30

Figure 6. TEMR Technology……………………………………………………………33

Figure 7. Data from Study Using TEMR in Mouse LD (from Machingal, 2011 used with

permission)……………………………………………………………………………….35

Figure 8. Muscle Regeneration in a Mouse LD VML model (Corona, 2012 used with

permission)……………………………………………………………………………….36

Figure 9. Data from TEMR Study in Rat TA (from Corona, 2014 used with

permission)……………………………………………………………………………….39

CHAPTER II: Cell and Growth Factor Loaded Keratin Hydrogels for Treatment of

Volumetric Muscle Loss in a Mouse Model

Table I. List of Groups, Components, and Number of Animals (n)……….....................68

Figure 1. Surgical Flow of VML injury and Keratin Implant…………...........................69

viii
Table II. Summary of Latissimus Dorsi Muscle Physical and Functional Measurement

Data……………………………………………………………………………………....72

Figure 2. Muscle Function Analysis at 2 Months Post-Surgery…………………...……74

Figure 3. Selected Force-Frequency Curves with Fitted Dose-Response Curves……....76

Figure 4. Representative Tissue Gross Morphology…………………………..…….…..77

Figure 5. Mouse LD Cell and Tissue Morphology and Functional Protein Expression in

Uninjured, NR, and BAM Treated Tissues…………………………………………...….79

Figure 6. Mouse LD Cell and Tissue Morphology and Functional Protein Expression in

KN, KN+I, KN+b, and KN+I+b Treated Tissues…………………..……………..……..81

Figure 7. Mouse LD Cell and Tissue Morphology and Functional Protein Expression in

KN+M, KN+M+I, KN+M+b, and KN+M+I+b Treated Tissues…………………..……83

CHAPTER III: Keratin Hydrogel Enhances In Vivo Skeletal Muscle Function in a Rat

Model of Volumetric Muscle Loss

Table I. Design of Experimental Groups Submitted to VML Injury…..........................103

Figure 1. Creation of VML in Rat TA and Injection of Keratin Hydrogel………….....106

Figure 2. Contribution of Keratin Hydrogel Formulation to Muscle Regeneration at 4 and

8 Weeks…………………………………………………………………………………110

Table II. In Vivo Functional Analysis of TA Muscle at 8 Weeks Post-Injury…...….…113

Figure 3. Keratin Hydrogel Promotes Continuous Functional Recovery over Time…..115

Table III. In Vivo Functional Analysis of TA Muscle at 12 Weeks Post-Injury……....117

Figure 4. Gross Morphology and Histological Analysis of TA after Keratin Based

Hydrogel Treatment…………………………………………………………………….119

ix
CHAPTER IV: Investigation of Tissue Engineered Muscle Repair Constructs for Use in

Cleft Lip Secondary Revision Procedures in a Rat Model

Table I. Treatment Groups……………………………………………………………..140

Figure 1. Surgical Flow of Rat LD VML Injury and repair with TEMR…………..….140

Table II. Summary of Latissimus Dorsi Muscle Physical and Functional Measurement

Data at 2 Months Post-Injury…………………………….……………………………..144

Table III. Summary of Latissimus Dorsi Muscle Physical and Functional Measurement

Data at 4 Months Post-Injury………………………………………………….……..…146

Table IV. Summary of Latissimus Dorsi Muscle Physical and Functional Measurement

Data at 6 Months Post-Injury……………………………………………….………..…148

Figure 2. Force-Frequency Curve with Dose-Response Curves for Males at 2mos…...150

Figure 3. Comparison of Po across Treatment Groups and Timepoints in Males…...…151

Figure 4. Comparison of Pt across Treatment Groups and Timepoints in Males………151

Figure 5. Comparison of P30 Hz across Treatment Groups and Timepoints in Males…..152

Figure 6. Comparison of Po across Treatment Groups and Timepoints in Females..….152

Figure 7. Comparison of Pt across Treatment Groups and Timepoints in Females……153

Figure 8. Comparison of P30 Hz across Treatment Groups and Timepoints in Females..153

Table V. Body Weight at Implant and Explant, Defect Weight & Size, and LD Weight

and Lo for Males and Females………………………………………………………….154

Figure 9. Macroscopic Comparison of Selected Representative Tissues across Treatment

Groups for Males at 2mos………………………………………………………………157

Figure 10. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Males at 4mos……………………………..……………………158

x
Figure 11. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Males at 6mos……………………………………..……………159

Figure 12. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Females at 2mos………………………………………...………160

Figure 13. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Females at 4mos………………………………………...………161

Figure 14. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Females at 6mos………………………………………...………162

Table VI. Summary of Macroscopic Findings in Males…………………………….…163

Table VII. Summary of Macroscopic Findings in Females………………………...….164

Figure 15. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 2mos Male TEMR…………………………………..168

Figure 16. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 2mos Male BAM……………………..……………..170

Figure 17. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 2mos Male NR…………………..…………………..172

Figure 18. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 2mos Female TEMR……………………….………..174

Figure 19. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 2mos Female BAM……………………...………..…176

Figure 20. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 2mos Female NR………………………..………..…178

xi
Figure 21. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 4mos Male TEMR……………………………..……180

Figure 22. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 4mos Male BAM…………………..………..………182

Figure 23. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 4mos Male NR……………..…………………..……184

Figure 24. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 4mos Female TEMR……………………….…..……186

Figure 25. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 4mos Female BAM…………………………….....…188

Figure 26. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 4mos Female NR…………………………..……..…190

Figure 27. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 6mos Male TEMR…………………………………..192

Figure 28. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 6mos Male BAM………………………..…………..194

Figure 29. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 6mos Male NR………………………………………196

Figure 30. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 6mos Female TEMR………….……………………..198

Figure 31. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 6mos Male TEMR…………………………………..200

xii
Figure 32. Hematoxylin and Eosin and Masson’s Trichrome Staining of Tissues from

Selected Representative Tissues: 6mos Male TEMR……………………………..……202

Table VIII. Summary of Key Histological Findings for TEMR and BAM Treated

Tissues in Males…………………...……………………………………………………204

Table IX. Summary of Key Histological Findings for TEMR and BAM Treated Tissues

in Females……………………………………………….……………………………...206

Supplemental Figure 1. Macroscopic Images of all 2mos Male TEMR Tissues..……215

Supplemental Figure 2. Macroscopic Images of all 2mos Male BAM Tissues………216

Supplemental Figure 3. Macroscopic Images of all 2mos Male NR Tissues…....……217

Supplemental Figure 4. Macroscopic Images of all 4mos Male TEMR Tissues..……218

Supplemental Figure 5. Macroscopic Images of all 4mos Male BAM Tissues....……219

Supplemental Figure 6. Macroscopic Images of all 4mos Male NR Tissues..………..220

Supplemental Figure 7. Macroscopic Images of all 6mos Male TEMR Tissues..……221

Supplemental Figure 8. Macroscopic Images of all 6mos Male BAM Tissues............222

Supplemental Figure 9. Macroscopic Images of all 6mos Male NR Tissues..…….….223

Supplemental Figure 10. Macroscopic Images of all 2mos Female TEMR Tissues….224

Supplemental Figure 11. Macroscopic Images of all 2mos Female BAM Tissues…...225

Supplemental Figure 12. Macroscopic Images of all 2mos Female NR Tissues….….226

Supplemental Figure 13. Macroscopic Images of all 4mos Female TEMR Tissues….227

Supplemental Figure 14. Macroscopic Images of all 4mos Female BAM Tissues…...228

Supplemental Figure 15. Macroscopic Images of all 4mos Female NR Tissues……..229

Supplemental Figure 16. Macroscopic Images of all 6mos Female TEMR Tissues….230

Supplemental Figure 17. Macroscopic Images of all 6mos Female BAM Tissues…...231

xiii
Supplemental Figure 18. Macroscopic Images of all 6mos Female NR Tissues……..232

CHAPTER V: Summary, Conclusions, and Future Directions

Table I. Experimental Groups for Optimization of Keratin Hydrogel Cellularity……..253

Table II. Experimental Groups for In Vivo Keratin Hydrogel Coupled Growth Factor

Dose Response Experiment………………………………………………………….....256

Table III. Experimental Groups for Mechanical Testing of Test Articles in Series with

Rat LD Muscle…......…………………………………………………………………...258

Table IV. Experimental Groups for Study of Laminate and Multi-Seeded TEMR……260

APPENDIX 1: Implantation of In Vitro Tissue Engineered Muscle Repair Constructs and

Bladder Acellular Matrix Partially Restore In Vivo Skeletal Muscle Function in a Rat Model

of Volumetric Muscle Loss Injury

Table I. TA muscle in vivo functional capacity 12 weeks post-injury……………..…293

Table II. Summary of BAM scaffold and TEMR construct in vivo performance…….294

Figure 1 TA muscle VML injury repair with BAM scaffold and TEMR constructs…295

Figure 2. In vivo isometric torque of the anterior crural muscles before and after TA

muscle synergist ablation and VML…………………………………………....296

Figure 3. TEMR improves functional recovery of rat TA muscle with VML injury…297

Figure 4. TA muscle gross morphology 12 weeks after VML and treatment…….......298

Figure 5. TA muscle tissue morphology following transplantation of BAM and TEMR

constructs……………………………………………………………….………299

xiv
Figure 6. TA muscle fiber generation and inflammation following transplantation of

BAM and TEMR constructs……………………………………………………300

APPENDIX 3. Growth Factor Release from Keratin Hydrogel Formulation Used in

Mouse LD and Rat TA Models (Unpublished Data from KeraNetics, LLC)

Figure 1. Release of IGF-1 and bFGF from Blends of KOS:KTN……………...........351

xv
LIST OF ABBREVIATIONS

AA: Antibiotic Antimycotic

ADSC: Adipose Derived Stem Cell

BAM: Bladder Acellular Matrix

BMMSC: Bone Marrow Mesenchymal Stem Cell

BMP: Bone Morphogenic Protein

DMEM: Dulbecco’s Modified Eagle Medium

ECM: Extracellular Matrix

EDS: Glutamic Acid Aspartic Acid Serine

FBS: Fetal Bovine Serum

FGF: Fibroblast Growth Factor (bFGF, FGF-2: Basic Fibroblast Growth Factor)

GAG: Glycosaminoglycan

GIF: Gamma Interferon

GMP: Good Manufacturing Process

HGF: Hepatocyte Growth Factor

HS: Horse Serum

HSPG: Heparan Sulfate Proteoglycans

IGF: Insulin-like Growth factor

IL: Interleukin

IND: Investigational New Drug

KAP: Keratin Associated Protein

KIF: Keratin Intermediate Filament

xvi
KOS: Keratose

KTN: Kerateine

LD: Latissimus Dorsi

LDV: Leucine Aspartic Acid Valine

LIF: Leukemia Inhibitory Factor

M1: Type 1 Macrophage

M2: Type 2 Macrophage

MHC: Myosin Heavy Chain

MMP: Matrix Metalloproteinase

MPC: Muscle Progenitor Cell

NGF: Nerve Growth Factor

NR: No Repair

OCT: Optimum Cutting Temperature

Po: Maximum Isometric Contraction Force

PAA: Peracetic Acid

PBS: Phosphate Buffered Saline

PCSA: Physiological Cross Sectional Area

PFA: Paraformaldehyde

QSR: Quality Service Regulation

TA: Tibialis Anterior

TE: Tissue Engineered

TGA: Thioglycolic Acid

TEMR: Tissue Engineered Muscle Repair

xvii
TGF: Transforming Growth Factor

TNF-α: Tissue Necrosis Factor alpha

VEGF: Vascular Endothelial Growth Factor

VML: Volumetric Muscle Loss

xviii
ABSTRACT

Volumetric muscle loss (VML) is defined as the irrecoverable loss of function in

skeletal muscle as a result of tissue loss. Herein, cell and growth factor loaded keratin

hydrogels and Tissue Engineered Muscle Repair (TEMR) constructs consisting of cell

seeded, cyclic stretched bladder acellular matrix (BAM), were investigated in VML

injuries in which the overriding hypothesis was that these technologies would improve

functional recovery and tissue morphology in otherwise irrecoverable injuries.

In the presented studies, investigation of keratin hydrogels in mouse latissimus

dorsi (LD) and rat tibialis anterior (TA) injuries determined that keratin combined with

insulin like growth factor-1, IGF-1, and basic fibroblast growth factor, bFGF, (KN+I+b)

resulted in the highest recovered contraction force. In the mouse, improvement was

significantly greater than that of all groups besides keratin alone and keratin combined with

muscle progenitor cells and IGF-1. In the rat, these findings were different from no repair

and BAM, but not from keratin alone or KN+b. Histological findings in both studies

indicated keratin combined with both growth factors, KN+I+b, resulted in improved tissue

quality with evidence of new muscle tissue formation.

In the investigation of TEMR in a novel model of VML with bio-relevance to

secondary revision of cleft lip, constructs were introduced into a surgically created defect

in the rat LD which was ~4 times larger than our previously largest model. To determine

the effectiveness of TEMR in the new model, in vitro functional analysis as well as

morphological and histological analysis were conducted on whole excised muscles at 2, 4,

6 months after implant. Results indicated that while the greatest improvement in function

was seen in TEMR treated tissue at 2 months, the findings were not significantly different

xix
from BAM treated and untreated tissues and no difference was seen at later timepoints.

Interestingly, morphological and histological findings indicated that TEMR resulted in

improved vascularity and attenuated inflammation compared with BAM treated tissues.

Investigation of these technologies indicated their capacity to improve muscle

function and form and contributed to the overall goal of our work to develop a platform of

TE skeletal muscle therapies capable of treating a diversity of VML presentations.

xx
CHAPTER I

Introduction

Hannah Besser Baker

Department of Biomedical Engineering

Wake Forest University School of Medicine, Winston-Salem, NC 27157

Affiliations: Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences,

Winston-Salem, NC 27157; Wake Forest Institute for Regenerative Medicine,

Winston-Salem, NC 27101

Note: This chapter represents the majority of a manuscript in preparation for submission

as a review of tissue engineering approaches to treating Volumetric Muscle Loss injuries.

This chapter was composed by Hannah Besser Baker with editorial guidance from

George J. Christ, Ph.D.

1
Clinical Significance

Extremity and craniofacial traumatic injuries are common among civilians and

active military personnel alike. In a report detailing the resources devoted to injuries on the

battlefield, extremity trauma ranked as the most common injury, having the longest

inpatient period, and accounting for 65% of the total disability costs calculated (1). High

energy explosions are responsible for as much as 81% of war conflict extremity injuries (1,

2). In civilians traumatic injuries are typically a result of gunshot wounds and motor

vehicle accidents, while on the battlefield, explosions are the major cause. In addition to

gunshot wounds, civilians may face volumetric muscle loss as a result of several other

indications. Some include the removal of cancerous tissue, congenital defects such as cleft

lip or palate, and muscle-neurological indications such as Bell’s palsy and Moebius

Syndrome (3, 4). Among the most prevalent of non-traumatic VML indications is cleft lip

which occurs alone or with cleft palate in 10.5/10,000 live births in the United States (5).

VML varies greatly in presentation with a wide range of complexity and severity,

especially when several tissues are involved. For example, a blast injury to the extremities,

head, or neck may result in burns, bone fracture, and can cause a variety of muscular

injuries that can lead to loss of muscle function and amputation of the limbs (6, 7).

Standard Treatment

When a significant portion of muscle has been lost, surgical management using

muscle flap transfer is the standard treatment (7-10). As a result of the various conditions

surrounding volumetric muscle loss, the native structure of the muscle, including the shape,

the vascularity, and the innervation, must be considered when choosing a repair strategy;

2
not just one type of graft is advisable for all situations. For example, in reconstruction of a

large craniofacial muscle defect the latissimus dorsi myocutaneous flap may be preferred

for its greater size. However, in the case of facial reanimation the neurovascular transfer of

a gracilis free flap may be preferred due to its particular innervation. Moreover, in the case

of lower extremity trauma, the flap which most adequately fills the space and has the most

similar vasculature is chosen (3, 9, 11).

While microsurgical technique has made major strides in reducing the donor site

morbidity and increasing the aesthetic quality of muscle flap grafts for traumatic VML and

orofacial defects, such setbacks remain present. Flap transfers have high risk of

complication and, due to the complexity of the procedure and lengthy hospitalization time,

are also costly (12, 13). For muscular defects such as cleft lip, primary surgical revision

involves repositioning of the skin and muscular tissue of the nose and nasal floor, however

this treatment suffers from graft tissue deficiency, scar formation, and stigmatic secondary

deformities (5, 14, 15). In addition, revision of secondary cleft lip/palate deformities often

requires an additional muscular graft and thus further donor site morbidity (15).

Research Considerations

The limitations of conventional treatments for VML have spurred tissue

engineering research endeavors for alternative therapies. It is important to note that unlike

minor injuries such as contusions and over exertion injuries and severe trauma injuries such

as compartment syndrome and ischemia-reperfusion injuries, VML injuries to skeletal

muscle pose an exceptional challenge due to the physical loss of muscle and the

surrounding matrix. Significant loss of tissue coupled with the absence of proper

3
scaffolding for regeneration results in injuries that cannot properly heal through the natural

regenerative process (16). Without intervention, the remodeling phase of the healing

process results in mostly non-functional scar tissue (17-21).

Skeletal Muscle Background

Skeletal Muscle Structure and Function

Comprising almost half of the human body’s mass, skeletal muscle enables

coordinated motion to the body through attachments to the skeleton (19). Muscle consists

of bundles of muscle fibers of which there are three types: type I which are slow twitch

fibers that do not fatigue easily, type IIA fibers that are fast twitch fibers that are resistant

to fatigue, and type IIX which are fast twitch and not resistant to fatigue. These fibers exist

as groups of bundled muscle fibers surrounded by connective tissue and interwoven with

the nerves and blood vessels that are vital to proper muscle function. The connective tissue

layers extend beyond the length of the muscle tissue to form tendons. A single muscle is

covered tendon to tendon with an outer protective sheath of connective tissue, the

epimysium, which holds together several bundles of muscle fibers called fascicles. Each

fascicle is surrounded by a connective tissue layer called perimysium. The individual fibers

within fascicles are multinucleated muscle cells, myocytes, which are each surrounded by

a plasma membrane, several layers of extracellular matrix which are referred to as the

sarcolemma, as well as an additional outer connective tissue layer called endomysium.

Below the sarcolemma are three additional layers of extracellular matrix proteins which

make up the basal lamina. Between the basal lamina and the sarcolemma there exists a

4
quiescent population of satellite cells that will regenerate tissue after injury (Figure 1) (19,

22).

Contained within the sarcolemma are the cellular organelles including nuclei,

which reside just below the sarcolemma in mature muscle fibers, the Golgi apparatus,

mitochondria, and the sarcoplasmic reticulum. Each myocyte contains several myofibrils

which are bundles of contractile proteins, myofilaments. Myofilaments consist of thick

filaments, made mostly of the protein myosin, and thin filaments, made mostly of the

protein actin. Actin and myosin overlap to form the basic contractile unit of muscle, the

sarcomere.

The sarcoplasmic reticulum consists of branches that surround each myofibril and

form terminal cisterna that store calcium for muscle contraction. At the midpoint of the

overlap between actin and myosin filaments, the terminal cisternae abut invaginations

called transverse tubules that travel from the surface of the sarcolemma deep into the

muscle cells along the sarcoplasmic reticulum (23).

The motor nerves which drive muscle function have axons that branch many times

into terminal axons that each interact with only one myocyte at the motor end plate of the

neuromuscular junction. A group of myocytes innervated by the same nerve-cell axon are

called a motor unit. An action potential propagated down a motor nerve will cause release

of acetylcholine at the presynaptic axon into the synaptic cleft. The acetylcholine binds

receptors on an adjacent post synaptic region of the sarcolemma which causes

depolarization of the sarcolemma and t-tubules.

5
Figure 1. Skeletal Muscle Form and Histology of Immature and Mature

Myofibers. Muscle exists as bundled bundles of fibers surrounded by connective

tissue and interwoven with nerves and blood vessels. The epimysium is a protective

sheath surrounding the whole muscle. The perimysium surrounds bundles of fibers

called fascicles. The endomysium surrounds individual fibers consisting of Myocytes

and satellite cells. Upon injury satellite cells are activated to become myoblasts which

differentiate into immature myotubes which become mature fibers of which the nuclei

are present at the periphery of fibers. (Figure from Huard, 2002 used with permission)

6
Activated voltage gated dihydropyridine receptors on the t-tubule will induce a

conformational change in colocalized ryanodine receptors on the sarcoplasmic reticulum

which causes a transient release of calcium from the terminal cisternae into the cytoplasm.

The released calcium ions interact with the protein troponin-C on actin filaments resulting

in a conformational change that allows the actin to bind myosin and contraction of the

sarcomere to occur. The contraction ends when acetylcholinesterases break down

acetylcholine, calcium is recycled to the sarcoplasmic reticulum, and troponin conforms to

prevent actin-myosin interaction (23).

Muscle can contract in three ways: eccentric, isometric, and concentric. In eccentric

contraction the muscle contracts with less force than the applied load and lengthens. During

isometric contraction, the muscle contracts with a force equal to the load and there is no

change in length. In concentric contraction the muscle contracts with a force greater than

the load and shortens (23).

Skeletal Muscle Injury Pathology

Skeletal muscle trauma is variable in pathology as well as severity, however the

manifestation of distinct trauma injuries are similar at the cellular level. The major

processes of skeletal muscle damage and healing have been well documented (17-20, 24).

These phases overlap with one another, but can be distinguished as destruction, repair, and

remodeling (Figure 2) (17).

7
Figure 2. Skeletal Muscle Injury and Repair in VML. Injury resulted in

disruption of the basal lamina (a). Invading macrophages clear the debris (b).

Activated satellite cells migrate to the injury site and differentiate into myoblasts

while fibroblasts begin to create scar tissue (c). The myoblasts fuse to form

myotubes at the ends of the injured fibers (d). In VML the fibroblast production of

scar tissue exceeds the regenerative capacity of muscle and a scar tissue gap is

formed within the defect (e). The resulting scar formation will cause denervation

for previously innervated tissue (f). (Figure from Turner, 2012 used with

permission)

8
Destruction of Damaged Tissue: Necrosis and Phagocytosis

Mechanical disruption of the sarcolemma and basal lamina results in an influx of

extracellular calcium activating calcium-dependent proteases, such as calpains, which

dismantle myofibers and the cytoskeleton. This process is localized to the injured fibers

through the formation of a contraction band-hematoma in the void. Further destruction on

either side of the contraction band arises as circulating inflammatory cells are recruited and

activated by cytokines secreted by the injured tissue. Additional inflammatory cells are

also released and activated from within the muscle and disrupted blood vessels. The first

inflammatory cells to congregate around the damaged fibers are neutrophils, present

between two and 24 hours after injury. These are followed by an initial wave of phagocytic

macrophages which peaks around 48 hours. Thereafter a second wave of macrophages

arrives, peaking around four days and remaining for as long as several weeks. The early

arriving cells begin to degrade the necrotized tissue and release cytokines and growth

factors which amplify the recruitment of inflammatory cells and promote the release of

satellite cells from nearby intact basal lamina. An abundance of research supports the

theory of subpopulations of macrophages playing distinct roles in muscle repair (25-29).

Early arriving macrophages (type 1 macrophages) are associated with the degradation of

necrotized tissue through phagocytosis and with the release of pro-inflammatory factors.

Later arriving macrophages (type 2 macrophages) are considered to be responsible for

regeneration of tissue by inhibiting degradation through the secretion of anti-inflammatory

proteins and promoting the fusion of satellite cells to myofibers.

9
Repair of the Injured Myofibers

Satellite cells are primarily responsible for repairing injured muscle (30). These

cells are either physically released upon disruption of the basal lamina or migrate from the

healthy tissue to damaged areas. Growth factors, chemokines, and cytokines secreted by

muscle and inflammatory cells and released from stores within the extracellular matrix

control the activation, proliferation, and differentiation of the satellite cells. Upon

activation, cells will rapidly proliferate. Proliferating cells divide into both an

undifferentiated cell population to repopulate the store of quiescent satellite cells as well

as muscle precursor cells. The precursor cells differentiate into myoblasts and fuse to intact

fibers or to one another to form multinucleated myotubes which will also fuse to uninjured

fibers. Finally growing myotubes will mature into myofibers in which the nuclei move

from a central to a peripheral location (31, 32).

Remodeling of the Tissue

Newly formed myofibers will begin to fill the injury site, eventually contacting the

contraction band. By this time the contraction band has been reorganized by invading

fibroblasts. Initially, the inflammatory cells break down the hematoma which is replaced

by a network blood derived fibrin and fibronectin. The cross-linked network provides a site

for invading fibroblasts to adhere and subsequently secrete extracellular matrix proteins.

With time the contraction band is able to provide the injury site with the necessary

mechanical integrity to withstand contractions, but this reorganization makes it difficult for

the muscle fiber stumps on either side to penetrate and reunite. If the damage to the muscle

is too severe, as in volumetric muscle loss, the fibroblast ECM of the band will have created

10
an impenetrable scar. The muscle fiber stumps will attempt to reunite by branching and

even traveling outside of the basal lamina in an effort to push through the scar tissue, but

ultimately will form a musculotendinous junction with the scar tissue. In cases of less

severe damage in which the basal lamina remains intact, the regenerated myofibers will

extend from either side of the injury until they reunite. Muscle that regenerates in this

fashion is morphologically and histologically indistinguishable from undamaged tissue

(31, 32).

Chemical and Physical Cues Guiding Regeneration

After muscle tissue has been damaged, there are intricate interactions between cells

and the extracellular matrix and the milieu of secreted factors which play large roles in the

overall regenerative process. These central regenerative mechanisms are important

considerations in development of a tissue engineered treatment for VML.

Secreted Factors

Substantial effort has been put into isolating the factors that regulate the major

processes of muscle injury repair and regeneration. Several authors have thoroughly

reviewed the source and mechanisms of a wide variety of these factors, only the key factors

will be summarized here (17-21, 24, 33). A table has been included for reference (Table I).

11
Factor Source Role References
Allen, 1995;
Activation of satellite cells,
Bischoff, 1997
Myocytes, recruitment of MPCs to
HGF Hayashi, 2004;
MPCs injury site, inhibition of
Tatsumi, 1998;
differentiation
Tatsumi, 2004
Allen, 1989;
Activation and
Myocytes, Bischoff, 1997
proliferation of satellite
macrophages, Charge, 2004;
FGF cells, inhibition of
endothelial Florini, 1996
differentiation, potential
cells, platelets Sheehan, 1994;
influence on IGF-1
Thompson, 1989
Promotes differentiation,
Allen, 1989;
MPCs, in presence of mitogen
IGF-1 Florini, 1996
macrophages such as FGF promotes
Hayashi, 2004
proliferation
Allen, 1989;
Inhibits myoblast Bischoff, 1997
Macrophages,
proliferation and Charge, 2004;
vascular
TGF-β1 differentiation, promotes Florini, 1989
endothelial
differentiation of MPCs Kirk, 2000;
cells, platelets
into myofibroblasts Li, 2002; Li, 2004
Massague, 1986
Promote myoblast Arnold, 2007;
TNF-α and Type 1
proliferation, impair Charge, 2004
IL-1β macrophages
differentiation Ciciliot, 2010
Promote proliferation of Charge, 2004;
LIF Myocytes
MPCs Wang, 2008
Promote transition from
Differentiating
IL-4 M1 to M2 phenotype in Arnold, 2007
myoblasts
macrophages
Induce M2 phenotype,
Type 2 recruitment of myoblasts, Arnold, 2007;
IL-10
macrophages promotion of Deng, 2012
differentiation
Table I. Summary of Key Growth Factors and Trophic Factors involved in

Skeletal Muscle Repair.

12
HGF and FGF: Activators of Satellite Cells and Proliferation, Inhibitors of

Differentiation

Hepatocyte growth factor, HGF, is secreted by uninjured skeletal muscle cells and

muscle progenitor cells and is stored in the extracellular matrix. It has been shown to be

the major cytokine responsible for the activation of quiescent satellite cells both in vitro

and in vivo (34-37). HGF is untethered from the extracellular matrix upon mechanical

stretch or local injury and acts in a paracrine-autocrine manner by binding to the c-met

receptors on nearby satellite cells (34, 35). Studies have demonstrated the presence of the

activated form of HGF at early time points after injury surrounding myocytes and myotubes

in vivo (37, 38). Additionally, these investigations further demonstrated the established

effects of HGF as well as its chemotactic effects on nearby satellite cells (24, 38). When

added to in vitro cultures of myocytes in differentiation media, increased dosage of HGF

resulted in increased cell proliferation whereas the formation of myotubes decreased (24,

34-37, 39, 40).

Several studies have shown that a handful of cytokines in the Fibroblast Growth

Factor family, FGFs, have also been implicated in satellite cell activation and proliferation

as well as inhibition of MPC differentiation (24, 38, 41-43). Also similar to HGF, FGFs

act directly on satellite cells via FGF receptors and are stored in the basement membrane

and connective tissue surrounding myofibers through attachment to proteoglycans (44).

Unlike HGF, several cell types and blood borne platelets present in the muscle injury

environment have been found to produce FGFs including macrophages and vascular

endothelial cells (38, 43). In experiments in which in vitro cultures of muscle derived cells

13
were treated with a combination of growth factors, it was demonstrated that FGFs may

influence the activity of IGFs; however this mechanism remains unclear (43, 45).

IGF

Insulin-like growth factors I and II, IGF-I and IGF-II, are well known regulators of

regeneration in many tissues (24). IGFs are produced by myocytes as well as invading

macrophages and are present in the active form during the early and middle time points of

regeneration (37). Both IGFs have been shown to promote the contradictory activities of

proliferation and differentiation of muscle precursor cells; however, the mechanism

surrounding the differential activity is debated (37, 45). It has been suggested that in the

presence of a mitogen such as FGF or HGF, IGF-1 will also act as a mitogen and in the

absence of a mitogen IGF-1 will promote differentiation (43).

TGF-β1

Transforming growth factor beta 1, TGF-β1, is a key negative regulator during

regeneration. TGF-β1 is secreted by invading immune cells, endothelial cells, and platelets

(38, 46). It is well documented as acting to inhibit myoblast proliferation and differentiation

(24, 38, 43, 47-49). Alternatively, TGF-β gradients have been shown to recruit adjacent

activated satellite cells (38). While TGF- β is known to inhibit the differentiation of satellite

cells into myoblasts, it has been demonstrated to cause the differentiation of activated

muscle precursor cells into myofibroblasts (38, 43, 46, 50). Therefore, it is no coincidence

that studies have shown an increased expression of TGF- β1 to be associated with increased

extracellular matrix production and scar formation (17).

14
Additional Relevant Trophic Factors

Several other secreted factors isolated from regenerating skeletal muscle have been

proven to play a role in repair and remodeling events. Macrophages contribute greatly to

these processes and have been deemed essential for proper regeneration to take place (26,

27). Early arriving inflammatory or type I macrophages in culture with satellite cells

promote proliferation and inhibit differentiation (18, 24, 27). Type I macrophages secrete

the inflammatory cytokine tumor necrosis factor alpha, TNF-α, and interlukin-1β, IL-1β,

which promote myoblast proliferation and impair differentiation (51). Leukemia inhibitory

factor, LIF, of the IL-6 family of cytokines is secreted by muscle cells and has been shown

to increase proliferation of MPCs in culture (24, 51). Differentiating myoblasts secrete IL-

4 which in culture with type I macrophages, pro-inflammatory M1 population of

macrophages, alters their phenotype to that of a later arriving, anti-inflammatory or type II

macrophage, M2 population (27). Anti-inflammatory factors secreted by the M2 phenotype

include interlukin-10 and TGF-β which are respectively associated with inducing the M2

phenotype and with regulating the recruitment and differentiation of myoblasts (27, 52).

The Extracellular Matrix

The extracellular matrix (ECM) of skeletal muscle consisting of the basal lamina

and the sarcolemma physically and chemically guides the activation, proliferation, and

differentiation of satellite cells released after an injury (53). These properties are derived

from the proportions and arrangement of constituent proteins and polysaccharides. The

ECM is made up of collagens I and IV, laminins, fibronectin, and the glycosaminoglycans

(GAGs): heparan sulfate, chondroitin sulfate, and hyaluronic acid (54). The strength and

15
structure of the sarcolemma or stromal matrix results from a network collagen I fibers

decorated with hyaluronan and GAGs. The basement membrane is primarily made up of

the non-fibrillar collagen IV interwoven with fibronectin and laminin (33).

Cell integrin attachment sites on collagen I promote myoblast cell binding and

collagens I and IV as well as laminin promote myoblast proliferation. Components of the

stromal and basement matrix: Fibronectin and hyaluronan, are known to promote the

differentiation of myoblasts to myocytes (55). Heparan sulfate proteoglycans, HSPG, play

a very important role in the regulation of growth factors. Specifically, the growth factors

HGF and FGF which play roles in satellite cell activation and proliferation are stored in an

inactive state while tethered to HSPGs in the ECM (39, 40).

It has been shown that the ECM provides physical cues which can guide the differentiation

of stem cells according to stiffness (56, 57). In addition, the specific ultrastructural

properties of the remaining intact basal lamina guide alignment of the fusing myocytes and

new myofibers (31).

Mechanical Cues

Together, the ECM and the secreted factors enable the satellite cells to repair an

injury. A key mechanism enabling this collaboration is the effect of mechanical stiffness

on regenerating muscle tissue. Myocytes and especially fibroblasts sense and react to the

stiffness of their environment through mechanoreceptors (40, 57, 58). For muscle cells and

fibroblasts mechanical sensing directly involves ECM components. As the surrounding

intact muscle fibers contract and pull on the ECM surrounding the damaged tissue, the

filaments extending from the muscle cells and fibroblasts which are attached to the ECM

16
are physically pulled (40). For fibroblasts this results in increased secretion of scar tissue

proteins, while for muscle cells this promotes alignment and fusion (33, 50). In addition,

injury and further stretch on the ECM result in the release of growth factors bound to GAGs

which regulate the activation, proliferation, and differentiation of myoblasts (36, 39). This

is thought to begin with the secretion of matrix metalloproteinases, MMPs, by muscle cells

which stimulates nitric oxide synthesis. Nitric oxide mediates the release of HGF and IGF

from the ECM, two growth factors responsible for the proliferation and differentiation of

myoblasts (38-40).

Tissue Engineering Approaches to Volumetric Muscle Loss

Animal Models

To study treatments aimed to restore structure and function in VML, animal models

which adequately recapitulate the injury pathology are necessary. To mimic the human

condition, a proper animal model of volumetric muscle loss injury should entail the

physical removal of muscle tissue which results in a permanent loss of function. Further,

investigators must employ physiologically relevant assays of muscle form and function to

indicate that irrecoverable damage has been achieved. Several animal models exist which

have given insight into the regenerative process of skeletal muscle injuries, without causing

the permanent damage seen in VML. These models have however, successfully mimicked

the short term damage common to sports injuries and more importantly have captured the

general process of regeneration associated with all muscle injures (24). Methods used to

mimic and study sports related muscle damage have included rodent eccentric contraction

injuries and thermal injuries which produce large functional deficits at early time points,

17
but tend to regain almost all function within four weeks (59-64). Other models of muscle

injury have entailed the use myotoxins which disrupt membranes, cause cell lysis, and

inhibit neuro-transduction. The application of these toxins enabled the study of muscle

damage and repair mechanisms, but similarly to the sports injury models the toxin induced

injures fully recovered within three weeks or less (24, 47, 65, 66). While appropriate for

investigating repair and regeneration, these models resulted in full functional recovery

within weeks after the injury and are therefore not comparable to a VML injury. Several

groups have indeed developed animal models of VML in which there is permanent tissue

and functional loss. Surgical resection of tissue from muscles is one approach which results

in a functional deficit proportional to the amount of tissue removed. Groups have created

defects in this manner primarily in rodent and dog hindlimbs and abdominal walls (67-74).

Beyond the use of histological analysis, whole muscle force production capacity is a widely

employed physiologic measurement for studying skeletal muscle conditions. Whole

muscle peak tetanic force may be evaluated through nerve stimulation in vivo or electrical

or chemical stimulation in vitro through the use of a force transducer attached to the active

moment arm in vivo (the foot in the case of the hindlimb) or attached to either end of an

isolated muscle in vitro. When used with proper controls, such devices have enabled

researchers to quantitatively assess the extent of damage and recovery present for a given

VML model and investigated therapy (59, 61, 63, 64, 67, 74-76). Physiologically relevant

analyses such as functional testing serve to corroborate and give merit to histological

evidence. Realistic models of skeletal muscle injury coupled with appropriate assays will

serve to minimize translational distance and will provide an honest approximation of the

clinical application of a given technology.

18
Growth Factors

As previously described, many growth factors and cytokines influence the

regeneration of skeletal muscle after injury. These factors, usually secreted by cells

localized to the injury site, may be introduced artificially as human recombinant proteins.

Direct injection of IGF-1, basic-FGF (bFGF or FGF-2), and to a small extent nerve growth

factor, NGF, into a lacerated mouse hindlimb have enhanced the number and diameter of

newly formed muscle fibers and improved the ex vivo whole muscle force generation (77,

78). The exact role of the FGFs, such as bFGF, in vivo is debated as bFGF as well as FGFs

-4, and -6 have been shown to improve healing in some studies and to have no effect in

others (24, 79-82). Several groups have investigated the impact of LIF on regenerating

muscle in vivo in models of induced muscular dystrophy as well as traumatic injuries (24,

83, 84). In a rodent crush injury model, injections of LIF to the injury site increased the

size but not the number of regenerating myofibers (83). Perhaps a more feasible approach

to muscle regeneration using growth factors is the idea of limiting fibrosis at the wound

site (19). In preventing excessive scar tissue formation, the approach is to inhibit the

activity of TGF-β. TGF- β3, gamma interferon or GIF, and the molecule decorin have been

shown successful in inhibiting scar tissue formation in vivo (14, 77, 85). In a rodent

laceration model, injections of decorin alone improved muscle contraction force over

injections of IGF and decorin and IGF together; however IGF combined with decorin gave

the greatest new myofiber number and diameter (77). In a similar study, GIF also improved

force generation recovery and decreased scar tissue formation (85). The use of growth

factors for treatment of VML suffers due to the complex physical, spatial, and temporal

regulation and from promiscuity of growth factors among cells present in the injury site.

19
Nonetheless, the use of growth factors for treatment of VML has shown positive results,

with perhaps the most promising endeavors surrounding the use of antifibrotic factors.

Cellular Therapies

As it is well established that satellite cells are the major players in the natural repair

process, several researchers have investigated injectable muscle derived and purified

satellite cell therapies for skeletal muscle injuries (86-88). However, other investigators

have demonstrated that non-muscle stem cells are capable of differentiating into muscle

progenitor cells and promoting regeneration in skeletal muscle injuries (89-94). In such

studies, investigators have undertaken the use of primary muscle cells as well as adipose

derived stem cells (ADSCs) and bone marrow mesenchymal stem cells (BMMSCs) among

others for treatment of cryo-injury, laceration, crush injury, and loss of function due to

nerve denervation-reinnervation and cardiotoxin injection. In a study in which primary

myoblasts were injected into severely cryo-damaged mouse soleus muscles, the cell treated

muscles had significantly greater twitch and tetany tensions ex vivo (87). Similarly in a

rabbit tibialis anterior denervation-reinnervation model, primary isolated myoblasts

enabled improved in situ max contraction force at two months post injury, but at 14 months

there was no difference between treated and non-treated muscle contraction force (88).

Labeled ADSCs injected into a cardiotoxin damaged tibialis anterior muscles in rabbits

were found to be present in regenerating tissues fifteen days later and resulted in improved

muscle contraction force at two months post injection when compared with untreated

animals with damaged tissues. However, use of the freshly isolated ADSCs resulted in

adipose tissue development along the injection site (89). In a laceration injury in the rat

20
soleus, an injection of isolated ADSCs and matrigel resulted in improved ex vivo tetanic

contraction force relative to matrigel alone at two weeks post injury; however at four weeks

the contraction forces for the treatments were not different (92). Bone marrow

mesenchymal stem cells, have been investigated in a variety of regenerative medicine

applications, including their role in skeletal muscle injury repair (4). In crush injury model

in the rat soleus, direct injection of BMMSCs resulted in increased twitch and tetanic in

situ contraction force relative to untreated injured muscles (91, 93). Cell therapy shows

promise for the injuries described in these studies such as laceration cryo-damage, however

such cell therapies have not been investigated with success in larger VML injury models

in which the muscle tissue is no longer present. Further, direct cell transplant therapies

suffer from issues surrounding viability after transplant (95, 96)

Naturally Derived Scaffolds

As muscle progenitor cells along with other key cell types have been identified as

necessary components in skeletal muscle healing, the use of a variety of scaffolds alone to

attract cells to the injury site has been investigated. Several groups have researched the use

of decellularized extracellular matrices for use in treating skeletal muscle injuries. One

such study found that repair of a rat tendon injury using the BAM increased the presence

of multipotent progenitor cells when compared with control tissue, showcasing the ability

to act as a cellular chemoattractant (97). It has also been shown that after decellularization

such ECM scaffolds retain the protein structure, growth factors, and glycosaminoglycans

known to be involved with guiding muscle repair (98-101). Further, acellular ECM

scaffolds have also been shown to promote a favorable immune response as was the case

21
when BAM and an acellular body wall matrix were used to repair a body wall defect in rats

and the majority of invading macrophage populations were the favorable type two variety

at several early and later time points after implant (25). This finding also correlated to a

greater remodeling of the implant.

Several other studies have demonstrated varied success of various acellular ECM

scaffolds in repairing an abdominal wall defects. In studies using small intestine submucosa

and acellular skeletal muscle matrix in animal models of abdominal wall defects there was

evidence of replacement of the scaffolds with host tissue and the presence of functional

skeletal muscle within the scaffold (68, 102, 103). However such findings are contradicted

by others such as one case in which a diaphragm acellular matrix implant in an abdominal

wall defect in rabbits led to fibrosis and did not enable muscle formation within the graft

site (70). Similarly varied results have been found using acellular matrices in extremity

muscles. In studies using acellular ECM in hindlimb defects in rats and dogs, results

showed evidence of moderate cell infiltration within the scaffolds but without functional

improvement (69, 73). Another study evidenced the opposite phenomena: using acellular

skeletal muscle matrix in a rat hindlimb defect resulted in fibrotic tissue formation with a

lack of muscle cell presence within the scaffold, however there was functional

improvement when compared with no treatment. The author has attributed this to

“functional fibrosis,” in which the fibrotic tissue bears weight and prevents overload injury

to the surrounding muscle (104). In a study using porcine BAM to treat defects in mouse

and human quadriceps there was similar improvement in function with only minimal

evidence of muscle formation within the implants (105).

22
Other naturally derived scaffolds such as silk fibroin, keratin, alginate, and chitosan

have been investigated in several applications including skeletal muscle regeneration due

to their capacity to be manipulated into various structures and to act as drug delivery

vehicles (106-108). In one study silk fibroin-chitosan blend scaffold was found to

outperform an acellular matrix when used for abdominal wall repair in guinea pigs. The

blended scaffold better integrated with host tissue and retained a greater tensile strength

when compared with human acellular dermal matrix (109).

Other successful use of tunable natural scaffolds has included the use of hydrogels

as growth factor carriers. In one case alginate beads loaded with IGF-1 and or VEGF were

implanted in a limb ischemia model in rodents and resulted in increased muscle fiber

regeneration and angiogenesis respectfully (and increased angiogenesis and fiber

regeneration when used together) (110). It is apparent that naturally derived scaffolds alone

and coupled with growth factors show promise in treating skeletal muscle injuries. While

ECM scaffolds stand to promote cellular infiltration at the injury site and improve muscle

function, these scaffolds have a varied capacity for regenerating muscle tissue within the

scaffold and in the worst cases resulted in non-functional, fibrotic tissue. On the other hand,

synthesized natural materials such as alginate, silk, and keratin offer the potential to deliver

drugs or growth factors to the injury site to further improve recruitment and infiltration of

cells and new tissue formation, but these have not yet shown efficacy in VML models.

23
Combination Therapies: Scaffolds, Cells, and Preconditioning

More complex strategies to treat VML injuries have involved the use of a

combinations of scaffolds, cells, growth factors, and preconditioning. As with the use of

naturally derived scaffolds alone, investigators have tested a variety of ECM as well as

other materials in conjunction with different cell types in several skeletal muscle injury

models (111, 112). Several authors have used natural scaffolds seeded with primary

isolations of skeletal muscle cells with success (67, 74, 76, 113-116). In a notable study

using a photo-cross linking hyaluronan based hydrogel loaded with freshly isolated satellite

cells, muscle progenitor cells, or freshly isolated muscle fibers in a mouse tibialis anterior

defect, the gel and satellite cell combination was found to enable a much greater repair of

muscle tissue and functional recovery compared with use of the gel alone or the gels with

MPCs or fibers (113). Fibrin constructs, as gels and microthreads, loaded with MPCs in

rodent defects were also shown to improve neo tissue formation, reduce collagen

deposition, and improve functional recovery compared with non-treated injuries (114,

115).

Other investigators have utilized growth factors either directly or through genetic

modification of cells in combination with scaffolds and MPCs for treatment of skeletal

muscle injury. Alginate loaded with MPCs, IGF-1, and vascular endothelial growth factor

(VEGF) improved functional recovery, cell engraftment, and minimized scar formation in

a combined ischemia and toxin injury in mice (116). However, another study using alginate

loaded with either MPCs or MPCs made to over express FGF-2 in a crush injury in rats

found that while FGF-2 over expressing cells in alginate evidenced modulation of

24
apoptosis and proliferation of injured tissue, neither treatment resulted in significant

improvements to muscle function compared with no treatment (117).

In addition to the use of growth factors in combination with cells and natural

scaffolds, another strategy to improve the success of tissue engineered constructs for

skeletal muscle entails preconditioning scaffolds using mechanical or electrical stimuli.

Use of bioreactors to provide these cues to cell seeded scaffolds in vitro aims to enhance

the differentiation and alignment of cells on the construct to improve the ability of the

scaffold to restore function in vivo (118). Use of cyclic stretch alone or coupled with

electrical preconditioning in vitro has been found to promote contractile protein synthesis,

to increase myotube formation, diameter, and alignment as well as to improve the

excitability and force production of the cells on the scaffold (119-122). Further, when

preconditioned allogeneic cell seeded BAM scaffolds were used in a mouse model of VML

injury, functional recovery was greater than that enabled by static cultured cell seeded

scaffolds (67, 74). While combination therapies have demonstrated the ability to promote

the restoration of form and function in skeletal muscle injury models, it is worth noting that

the regulatory pathway for these complex technologies remains an obstacle for

investigators (123).

Keratin Hydrogels and Tissue Engineered Muscle Repair Constructs

Further Development of a Platform of Technologies for VML

Tissue engineering approaches to treatment of VML are diverse and unlimited

when one considers the possibilities enabled by combination therapies. To match the

variety and complexity of skeletal muscle injuries and pathologies, a multitude of treatment

25
options are necessary. In establishing a platform of such treatments for VML presentations,

cell and growth factor loaded keratin hydrogels and cell seeded and preconditioned BAM

are to be investigated as is described in this proposal (Figure 3). To gain a better

understanding of these technologies and their potential for use in treating VML, necessary

background and foundational research shall be reviewed.

Figure 3. Platform of Technologies for TE Skeletal Muscle. All technologies are

cell delivery capable while silk and keratin can be altered to achieve a desired

degradation rate and can be modified to deliver drugs. The geometry of each therapy

facilitates its use. TEMR for muscles with sheet like architecture, Silk for solid

geometry injuries, and keratin hydrogels for complex geometries. In this proposal,

studies to investigate keratin hydrogels and TEMR for VML are outlined

26
Keratin Structure and Isolation

In human hair, trichocytic keratins are intermediate filaments which make up the

cortex and cuticle of hair fibers. They can be classified into three types: alpha, beta, and

gamma keratins of which the alpha and gamma components have been investigated (Figure

4a). Beta keratins make up the cuticle of the hair fiber and are difficult to isolate and have

structural properties which have not yet been found useful (124). Within the cortex, alpha

keratins assemble together into fibrous structures referred to as keratin intermediate

filaments (KIF) which make up 50-60% of the material and provide structure (Figure 4b).

Gamma keratins provide a supporting matrix and are referred to as keratin associated

proteins (KAPs). Gamma keratins make up 20-30% of the cortex. Alpha keratins exist as

heterodimers of type I (acidic) and type II (neutral-basic) keratins which further assemble

into 10nm intermediate filaments. Type I and type II keratins have a central alpha-helical

structure and a non-helical end domain but differ in amino acid composition and molecular

weight. Type I keratins have a higher molecular weight (40-60kDa) and a lower sulfur

content while type II keratins have a lower molecular weight (10-25kDa) and a higher

sulfur content (125-128).

27
Figure 4. Keratin Structure. Keratin intermediate filaments make up the bulk of hair

fibers (A). Acidic type I and basic type II keratins form helical dimers which

polymerize to form tetramers that make up protofibrils that further polymerize to form

intermediate fibers (B). Oxidation extraction of keratin converts cysteine bonds to

sulfonic acid while reductive extraction breaks the bonds but does not alter the

cysteine groups allowing the bonds to reform (C). The resulting proteins are referred

to as keratose and kerateine respectively and have distinct properties. (Keratin hair

illustration from hairkeratins.com, secondary to tertiary structure image from Sigma,

keratose and kerateine image from manuscript under review used with permission

from KeraNetics, LLC (Saul et al)).

28
Cortical keratins can be extracted by first chemically disrupting disulfide crosslinks

holding the fibers together followed by the use of denaturing solvents to solubilize the

proteins. The alpha and gamma fractions may then be purified using filtration and dialysis

(124). Reducing agents used for extraction will result in non-cross linked cysteine residues

for which the resulting keratins are referred to as kerateines (KTN). When oxidizing agents

are used in the extraction process the cysteine disulfide bonds are broken and converted to

sulfonic acid groups and the resulting extraction is referred to as keratose (KOS) (129,

130). Differences in the cysteine residue chemistry between KOS and KTN result in

different physical behaviors. Unpublished experiments carried out by collaborators using

keratin gels have demonstrated that KTN gels undergo a longer degradation time, have an

order of magnitude lower ratio of elastic to viscous moduli, and have a slower release of

loaded growth factors when compared with KOS, due to the reformation of disulfide bonds

in the free cysteine groups (Figure 4c)*. Specifically, KTN will form disulfide cross links

which result in a slower degradation rate and longer release profile of growth factors as

well as resulting in a stiffer less easily flowing gel (Figure 5)*. As such, the two materials

can be combined in specific ratios to achieve a desired degradation and drug or growth

factor release period.

*Denotes unpublished data used with permission of collaborators at KeraNetics, LLC.

29
Figure 5. Growth Factor Release from 20% Keratin Gels. 20% keratose gels (A)

showed a quicker release of growth factors than 20% kerateine gels (B). The release of

growth factors from both gels was first order, dependent on the degradation of the gel

(A, B).

30
Keratin Hydrogels for Use in Regenerative Medicines

Keratins have several useful properties for use in regenerative medicine

applications. Extracted alpha keratins have been found to contain the cell recognition

amino acid sequences leucine-aspartic acid-valine (LDV) and glutamic acid-aspartic acid-

serine (EDS) (131, 132). Keratins are capable of adhering and activating platelets which

led investigators to believe keratins have receptors for the β1 and β3 integrin present on

platelets (133). Keratins have also been indicated in the promotion of a favorable immune

response. In an in vitro study in which a monocytic cell line was seeded onto keratin

macrophages exhibited the type 2 anti-inflammatory, pro-remodeling phenotype.

Macrophages were found to secrete more anti-inflammatory cytokines, and a decreased

amount of inflammatory cytokines (134). The favorable interactions of keratin biomaterials

with cells in addition to the capacity to be manipulated into a variety of constructs and the

ability to act as a drug delivery vehicle has led many investigators to utilize keratin in a

variety of regenerative medicine applications (126).

Keratin hydrogels in particular have been successful in several applications,

including bone and nerve repair, treatment of cardiac infarctions, and use in promoting

hemostasis (124, 133, 135-140). Encouraged by these successes, keratin hydrogels with

and without the addition of cells and growth factors will be investigated in a rodent model

of VML for use as a skeletal muscle injury treatment as explained in this proposal.

31
Development of a Tissue Engineered Muscle Repair Construct

As reviewed previously, combination products involving the use of ECM scaffolds

and cells have great potential for skeletal muscle tissue engineering. One such scaffold,

which will be referred to as a Tissue Engineered Muscle Repair (TEMR) construct consists

of a porcine derived bladder acellular matrix (BAM) scaffold seeded with primary muscle

progenitor cells and subjected to cyclic stretch preconditioning (Figure 6) (67). Initial

studies carried out by Moon et al sought to determine the effect of cyclic stretch

preconditioning on a bioengineered muscle scaffold which in this study was a human

skeletal muscle cell seeded BAM scaffold. In this work, it was determined that use of a

stretch regimen for one week in vitro resulted in enhanced tissue organization and cellular

alignment. Further, at one week after a subcutaneous implant onto the LD of athymic mice,

retrieved constructs stained positive the muscle markers α-actin and myosin heavy chain,

MHC (122). With the framework in place: a stretch protocol and an ECM scaffold material

to pursue, TEMR development was underway.

32
Figure 6. TEMR Technology. Primary isolated myoblasts (A) are seeded onto BAM

scaffolds (B). The cell seeded scaffolds are subjected to cyclic stretch in a bioreactor

(C) which increases the number and size of multinucleated myofibers (D). The

scaffolds are then ready to implant (E).

33
In the first major study utilizing TEMR, BAM scaffolds alone were compared with

the cell seeded, preconditioned, TEMR, constructs for their ability to restore function to a

VML injury in the mouse LD (67). Two months after implant into the defect, explanted

TEMR scaffold treated muscles subjected to electrical stimuli had a maximal contraction

of ~72% of the contralateral control muscles, while the BAM and no repair, NR, groups

were at ~46% and ~45% (Figure 7d). Immunohistochemistry indicated the presence of new

muscle tissue at the interface and within the scaffold as well as the presence of

neurovascular bundles (Figure 7e, 7a-c). In a follow up study investigating the effects of

the addition of MPCs during the week of bioreactor preconditioning also using the mouse

LD defect model, TEMR constructs with and without the addition of MPCs gave similarly

favorable functional and morphologic outcomes. TEMR treated tissues examined at the

interface of the implant and the native tissue stained positive for the force transmission

proteins desmin and myosin the excitation-contraction coupling proteins ryanodine

receptor 1 and junctiphilin 1 (Figure 8) (74).

34
Figure 7. Data from Study Using TEMR in Mouse LD (from Machingal, 2011

used with permission). Neurovascular bundles could be seen within the site of the

TEMR implant (A) with positive staining for vasculature (von Willebrand’s factor, B)

and neurofilaments (NF-200, C). The formation of new muscle tissue within the

implant region was further evidenced by positive staining for desmin filaments (E).

Maximum isometric muscle contraction forces are shown at two months post-surgery

(D) for native tissue (blue), NR (red), TEMR treated (green) and BAM treated (purple)

samples.

35
Figure 8. Muscle Regeneration in a Mouse LD VML model (Corona, 2012 used

with permission). All histology shown was conducted on TEMR treated LD muscles.

Images were taken at the interface (A-E) and within the scaffold region (F-J). Muscles

were stained with Masson’s trichrome (A, F), for the functional proteins desmin (B,G)

and myosin (C, H) and the excitation-contraction coupling proteins ryanodine recptor-

1 (D, I) and junctophilin-1 (E, J).

36
The TEMR scaffold (one seeding, original protocol) was later employed in a VML

model in the rat TA (76). Interestingly, in this system the TEMR treatment gave two

distinct results referred to as low and high responders. While the high responders gave a

functional recovery significantly greater than that enabled by BAM or no repair, the low

responders resulted in a loss of function (Figure 9i). Similar to previous results from

Machingal et al, high responders yielded tissues with evidence of new muscle formation

and less of an inflammatory response when compared with NR, BAM, and TEMR low

responders (Figure 9ii-iii). Taken together, the results of these TEMR studies indicate the

benefits of this technology in VML models while highlighting some potential pitfalls. For

example, the resulting subset of TEMR low responders in the study carried out by Corona

et al in the rat model should be a forethought to planning future studies with this treatment.

One theory is that the cellular density exposed to the injury site was compromised when

applying the TEMR technology in the larger VML injury model.

Research using TEMR constructs has showcased the ability of the constructs to

integrate with the host tissue and enable functional recovery in an otherwise irrecoverable

defect in two VML models with distinct geometries. These successes have inspired its use

for VML indications outside of trauma. As covered earlier there are many VML

presentations which include traumatic injury, but also acquired diseases which affect the

musculature and congenital birth defects involving muscle such as cleft lip and cleft palate.

In the study proposed herein, TEMR constructs will be investigated for application in a

VML model which has bio-relevance to a cleft lip secondary revision.

37
Figure 9. Data from TEMR Study in Rat TA (from Corona, 2014 used with

permission). Maximum contraction force relative to control muscles is shown for NR,

BAM, and negative and positive TEMR groups at 4 weeks in top row, 8 weeks in the

middle row, and for 12 weeks in the bottom row (i). A Masson’s trichrome stain was

conducted (ii) on longitudinal sections (A-E) and cross sections (F-J) for control

muscles (A, F), NR (B, G), BAM (C, H), TEMR high responders (D, I) and TEMR

low responders (E, J). Muscle samples were stained for myosin (MF-20, A-C) and

macrophages (CD-68, D-F) (iii). BAM samples are shown in A and D, TEMR high

responders in B and E, and TEMR low responders in C and F.

38
Summary

The repair and regeneration of skeletal muscle after an injury is a complex process

involving multiple cell types, factors, and physiologic cues (17-20, 24). VML indications

are characterized by their ability to overwhelm the native capacity of skeletal muscle to

repair and remodel tissue after injury (7). TE therapies offer a means of enabling repair and

regeneration for VML (19, 21, 86, 141-143). When examining potential treatments for

skeletal muscle repair or remodeling in animal models, it is of key importance that the

chosen models accurately mimic the clinical indication for the investigated treatment. In

the case of VML injury, this means induced muscle injuries must be irrecoverable without

intervention (7). For treatment of muscle injury, investigators have examined the

effectiveness of cell therapies, naturally derived scaffolds, growth factors and

combinations of the three as well with accompanying bioreactor preconditioning with a

range of results (141-143). Among the most exciting interventions were combination

therapies utilizing natural scaffolds (67, 74, 76, 113, 116, 117, 144). In developing a

platform of technologies capable of treating the vast presentation of VML, two exceptional

combination therapies using natural scaffolds will be further investigated: cell and growth

factor loaded keratin hydrogels and a cell seeded and preconditioned ECM scaffold

(TEMR).

39
Literature Cited

1. Masini, B.D., Waterman, S.M., Wenke, J.C., Owens, B.D., Hsu, J.R., andFicke, J.R.

Resource Utilization and Disability Outcome Assessment of Combat Casualties From

Operation Iraqi Freedom and Operation Enduring Freedom. Journal of Orthopaedic

Trauma 23, 261, 2009.

2. Owens, B.D., Kragh, J.F., Jr., Wenke, J.C., Macaitis, J., Wade, C.E., andHolcomb, J.B.

Combat wounds in operation Iraqi freedom and operation enduring freedom. Journal of

Trauma-Injury Infection and Critical Care 642008.

3. Pohlenz, P., Klatt, J., Schoen, G., Blessmann, M., Li, L., andSchmelzle, R.

Microvascular free flaps in head and neck surgery: complications and outcome of 1000

flaps. International Journal of Oral and Maxillofacial Surgery 412012.

4. Bianchi, B., Copelli, C., Ferrari, S., Ferri, A., andSesenna, E. Free flaps: Outcomes and

complications in head and neck reconstructions. Journal of Cranio-Maxillofacial Surgery

37, 438, 2009.

5. Canfield, M.A., Honein, M.A., Yuskiv, N., Xing, J., Mai, C.T., Collins, J.S., Devine,

O., Petrini, J., Ramadhani, T.A., Hobbs, C.A., Kirby, R.S., andNatl Birth Defects

Prevention, N. National estimates and race/ethnic-specific variation of selected birth

defects in the United States, 1999-2001. Birth Defects Research Part a-Clinical and

Molecular Teratology 76, 747, 2006.

6. Brown, K.V., Ramasamy, A., Tai, N., MacLeod, J., Midwinter, M., andClasper, J.C.

Complications of Extremity Vascular Injuries in Conflict. Journal of Trauma-Injury

Infection and Critical Care 66, S145, 2009.

40
7. Grogan, B.F., Hsu, J.R., andSkeletal Trauma Res, C. Volumetric Muscle Loss. Journal

of the American Academy of Orthopaedic Surgeons 19, S35, 2011.

8. Lawson, R., andLevin, L.S. Principles of free tissue transfer in orthopaedic practice.

Journal of the American Academy of Orthopaedic Surgeons 15, 290, 2007.

9. Heller, L., andLevin, L.S. Lower extremity microsurgical reconstruction. Plastic and

Reconstructive Surgery 108, 1029, 2001.

10. Seal, A., andStevanovic, M. Free Functional Muscle Transfer for the Upper Extremity.

Clinics in Plastic Surgery 38, 561, 2011.

11. Bianchi, B., Copelli, C., Ferrari, S., Ferri, A., andSesenna, E. Successful salvage

surgery after treatment failures with cross graft and free muscle transplant in facial

reanimation. Journal of Cranio-Maxillofacial Surgery 40, 185, 2012.

12. Kroll, S.S., Evans, G.R.D., Goldberg, D., Wang, B.G., Reece, G.P., Miller, M.J., Robb,

G.L., Baldwin, B.J., andSchusterman, M.A. A comparison of resource costs for head and

neck reconstruction with free and pectoralis major flaps. Plastic and Reconstructive

Surgery 991997.

13. Patel, R.S., McCluskey, S.A., Goldstein, D.P., Minkovich, L., Irish, J.C., Brown, D.H.,

Gullane, P.J., Lipa, J.E., andGilbert, R.W. CLINICOPATHOLOGIC AND

THERAPEUTIC RISK FACTORS FOR PERIOPERATIVE COMPLICATIONS AND

PROLONGED HOSPITAL STAY IN FREE FLAP RECONSTRUCTION OF THE

HEAD AND NECK. Head and Neck-Journal for the Sciences and Specialties of the Head

and Neck 32, 1345, 2010.

14. Hosokawa, R., Nonaka, K., Morifuji, M., Shum, L., andOhishi, M. TGF-beta decreases

type I collagen and scarring after labioplasty. Journal of Dental Research 82, 558, 2003.

41
15. Chen, Y.R., andYeow, V.K.L. Cleft lip scar camouflage using dermal micrografts.

Plastic and Reconstructive Surgery 103, 1250, 1999.

16. Jarvinen, T.A.H., Jarvinen, T.L.N., Kaariainen, M., Aarimaa, V., Vaittinen, S., Kalimo,

H., andJarvinen, M. Muscle injuries: optimising recovery. Best Practice & Research in

Clinical Rheumatology 21, 317, 2007.

17. Jarvinen, T., Jarvinen, T.L.N., Kaariainen, M., Kalimo, A., andJarvinen, M. Muscle

injuries - Biology and treatment. American Journal of Sports Medicine 33, 745, 2005.

18. Ciciliot, S., andSchiaffino, S. Regeneration of Mammalian Skeletal Muscle: Basic

Mechanisms and Clinical Implications. Current Pharmaceutical Design 16, 906, 2010.

19. Huard, J., Li, Y., andFu, F.H. Current concepts review - Muscle injuries and repair:

Current trends in research. Journal of Bone and Joint Surgery-American Volume 84A, 822,

2002.

20. Grefte, S., Kuijpers-Jagtman, A.M., Torensma, R., andVon den Hoff, J.W. Skeletal

muscle development and regeneration. Stem Cells and Development 16, 857, 2007.

21. Turner, N.J., andBadylak, S.F. Regeneration of skeletal muscle. Cell and Tissue

Research 3472012.

22. Zammit, P.S., Heslop, L., Hudon, V., Rosenblatt, J.D., Tajbakhsh, S., Buckingham,

M.E., Beauchamp, J.R., andPartridge, T.A. Kinetics of myoblast proliferation show that

resident satellite cells are competent to fully regenerate skeletal muscle fibers.

Experimental Cell Research 2812002.

23. Hall, J.E., andGuyton, A.C. Guyton and Hall Textbook of Medical Physiology.

Twelfth ed. Philadelphia, PA: Saunders Elsevier; 2010.

42
24. Charge, S.B.P., andRudnicki, M.A. Cellular and molecular regulation of muscle

regeneration. Physiological Reviews 84, 209, 2004.

25. Brown, B.N., Valentin, J.E., Stewart-Akers, A.M., McCabe, G.P., andBadylak, S.F.

Macrophage phenotype and remodeling outcomes in response to biologic scaffolds with

and without a cellular component. Biomaterials 30, 1482, 2009.

26. Brunelli, S., andRovere-Querini, P. The immune system and the repair of skeletal

muscle. Pharmacological Research 58, 117, 2008.

27. Arnold, L., Henry, A., Poron, F., Baba-Amer, Y., van Rooijen, N., Plonquet, A.,

Gherardi, R.K., andChazaud, B. Inflammatory monocytes recruited after skeletal muscle

injury switch into antiinflammatory macrophages to support myogenesis. Journal of

Experimental Medicine 204, 1057, 2007.

28. Stpierre, B.A., andTidball, J.G. DIFFERENTIAL RESPONSE OF MACROPHAGE

SUBPOPULATIONS TO SOLEUS MUSCLE RELOADING AFTER RAT HINDLIMB

SUSPENSION. Journal of Applied Physiology 77, 290, 1994.

29. Mantovani, A., Sica, A., Sozzani, S., Allavena, P., Vecchi, A., andLocati, M. The

chemokine system in diverse forms of macrophage activation and polarization. Trends in

Immunology 25, 677, 2004.

30. Hawke, T.J., andGarry, D.J. Myogenic satellite cells: physiology to molecular biology.

Journal of Applied Physiology 91, 534, 2001.

31. Hurme, T., Kalimo, H., Lehto, M., andJarvinen, M. HEALING OF SKELETAL-

MUSCLE INJURY - AN ULTRASTRUCTURAL AND IMMUNOHISTOCHEMICAL

STUDY. Medicine and Science in Sports and Exercise 23, 801, 1991.

43
32. Bodinefowler, S. SKELETAL-MUSCLE REGENERATION AFTER INJURY - AN

OVERVIEW. Journal of Voice 8, 53, 1994.

33. Gillies, A.R., andLieber, R.L. STRUCTURE AND FUNCTION OF THE SKELETAL

MUSCLE EXTRACELLULAR MATRIX. Muscle & Nerve 44, 318, 2011.

34. Allen, R.E., Sheehan, S.M., Taylor, R.G., Kendall, T.L., andRice, G.M.

HEPATOCYTE GROWTH-FACTOR ACTIVATES QUIESCENT SKELETAL-

MUSCLE SATELLITE CELLS IN-VITRO. Journal of Cellular Physiology 165, 307,

1995.

35. Tatsumi, R., Anderson, J.E., Nevoret, C.J., Halevy, O., andAllen, R.E. HGF/SF is

present in normal adult skeletal muscle and is capable of activating satellite cells.

Developmental Biology 194, 114, 1998.

36. Tatsumi, R., andAllen, R.E. Active hepatocyte growth factor is present in skeletal

muscle extracellular matrix. Muscle & Nerve 30, 654, 2004.

37. Hayashi, S., Aso, H., Watanabe, K., Nara, H., Rose, M.T., Ohwada, S., andYamaguchi,

T. Sequence of IGF-I, IGF-II, and HGF expression in regenerating skeletal muscle.

Histochemistry and Cell Biology 122, 427, 2004.

38. Bischoff, R. Chemotaxis of skeletal muscle satellite cells. Developmental Dynamics

208, 505, 1997.

39. Yamada, M., Tatsumi, R., Kikuiri, T., Okamoto, S., Nonoshita, S., Mizunoya, W.,

Ikeuchi, Y., Shimokawa, H., Sunagawa, K., andAllen, R.E. Matrix metalloproteinases are

involved in mechanical stretch-induced activation of skeletal muscle satellite cells. Muscle

& Nerve 34, 313, 2006.

44
40. Tatsumi, R. Mechano-biology of skeletal muscle hypertrophy and regeneration:

Possible mechanism of stretch-induced activation of resident myogenic stem cells. Animal

Science Journal 81, 11, 2010.

41. Sheehan, S.M., andAllen, R.E. Skeletal muscle satellite cell proliferation in response

to members of the fibroblast growth factor family and hepatocyte growth factor. Journal of

Cellular Physiology 1811999.

42. Thompson, S.H., Boxhorn, L.K., Kong, W.Y., andAllen, R.E. TRENBOLONE

ALTERS THE RESPONSIVENESS OF SKELETAL-MUSCLE SATELLITE CELLS TO

FIBROBLAST GROWTH-FACTOR AND INSULIN-LIKE GROWTH FACTOR-I.

Endocrinology 124, 2110, 1989.

43. Allen, R.E., andBoxhorn, L.K. REGULATION OF SKELETAL-MUSCLE

SATELLITE CELL-PROLIFERATION AND DIFFERENTIATION BY

TRANSFORMING GROWTH FACTOR-BETA, INSULIN-LIKE GROWTH FACTOR-

I, AND FIBROBLAST GROWTH-FACTOR. Journal of Cellular Physiology 138, 311,

1989.

44. Cornelison, D.D.W., Filla, M.S., Stanley, H.M., Rapraeger, A.C., andOlwin, B.B.

Syndecan-3 and syndecan-4 specifically mark skeletal muscle satellite cells and are

implicated in satellite cell maintenance and muscle regeneration. Developmental Biology

239, 79, 2001.

45. Florini, J.R., Ewton, D.Z., andCoolican, S.A. Growth hormone and the insulin-like

growth factor system in myogenesis. Endocrine Reviews 17, 481, 1996.

46. Li, Y., Foster, W., Deasy, B.M., Chan, Y.S., Prisk, V., Tang, Y., Cummins, J.,

andHuard, J. Transforming growth factor-beta 1 induces the differentiation of myogenic

45
cells into fibrotic cells in injured skeletal muscle - A key event in muscle fibrogenesis.

American Journal of Pathology 1642004.

47. Kirk, S., Oldham, J., Kambadur, R., Sharma, M., Dobbie, P., andBass, J. Myostatin

regulation during skeletal muscle regeneration. Journal of Cellular Physiology 184, 356,

2000.

48. Florini, J.R., andMagri, K.A. EFFECTS OF GROWTH-FACTORS ON MYOGENIC

DIFFERENTIATION. American Journal of Physiology 256, C701, 1989.

49. Massague, J., Cheifetz, S., Endo, T., andNadalginard, B. TYPE BETA-

TRANSFORMING GROWTH-FACTOR IS AN INHIBITOR OF MYOGENIC

DIFFERENTIATION. Proceedings of the National Academy of Sciences of the United

States of America 831986.

50. Li, Y., andHuard, J. Differentiation of muscle-derived cells into myofibroblasts in

injured skeletal muscle. American Journal of Pathology 161, 895, 2002.

51. Wang, X., Wu, H.T., Zhang, Z.X., Liu, S.H., Yang, J., Chen, X.P., Fan, M., andWang,

X.M. Effects of interleukin-6, leukemia inhibitory factor, and ciliary neurotrophic factor

on the proliferation and differentiation of adult human myoblasts. Cellular and Molecular

Neurobiology 28, 113, 2008.

52. Deng, B., Wehling-Henricks, M., Villalta, S.A., Wang, Y., andTidball, J.G. IL-10

Triggers Changes in Macrophage Phenotype That Promote Muscle Growth and

Regeneration. Journal of Immunology 189, 3669, 2012.

53. Osses, N., andBrandan, E. ECM is required for skeletal muscle differentiation

independently of muscle regulatory factor expression. American Journal of Physiology-

Cell Physiology 282, C383, 2002.

46
54. Kuraitis, D., Giordano, C., Ruel, M., Musaro, A., andSuuronen, E.J. Exploiting

extracellular matrix-stem cell interactions: A review of natural materials for therapeutic

muscle regeneration. Biomaterials 33, 428, 2012.

55. Calve, S., Odelberg, S.J., andSimon, H.G. A transitional extracellular matrix instructs

cell behavior during muscle regeneration. Developmental Biology 344, 259, 2010.

56. Reilly, G.C., andEngler, A.J. Intrinsic extracellular matrix properties regulate stem cell

differentiation. Journal of Biomechanics 43, 55, 2010.

57. Engler, A.J., Griffin, M.A., Sen, S., Bonnetnann, C.G., Sweeney, H.L., andDischer,

D.E. Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological

implications for soft or stiff microenvironments. Journal of Cell Biology 166, 877, 2004.

58. Calve, S., andSimon, H.G. Biochemical and mechanical environment cooperatively

regulate skeletal muscle regeneration. Faseb Journal 26, 2538, 2012.

59. Ingalls, C.P., Warren, G.L., andArmstrong, R.B. Dissociation of force production from

MHC and actin contents in muscles injured by eccentric contractions. Journal of Muscle

Research and Cell Motility 19, 215, 1998.

60. Warren, G.L., Hulderman, T., Jensen, N., McKinstry, M., Mishra, M., Luster, M.I.,

andSimeonova, P.P. Physiological role of tumor necrosis factor alpha in traumatic muscle

injury. Faseb Journal 16, 1630, 2002.

61. Warren, G.L., O'Farrell, L., Summan, M., Hulderman, T., Mishra, D., Luster, M.I.,

Kuziel, W.A., andSimeonova, P.P. Role of CC chemokines in skeletal muscle functional

restoration after injury. American Journal of Physiology-Cell Physiology 286, C1031,

2004.

47
62. Lowe, D.A., Warren, G.L., Ingalls, C.P., Boorstein, D.B., andArmstrong, R.B.

MUSCLE FUNCTION AND PROTEIN-METABOLISM AFTER INITIATION OF

ECCENTRIC CONTRACTION-INDUCED INJURY. Journal of Applied Physiology 79,

1260, 1995.

63. Corona, B.T., Hamilton, S.L., andIngalls, C.P. EFFECT OF PRIOR EXERCISE ON

THERMAL SENSITIVITY OF MALIGNANT HYPERTHERMIA-SUSCEPTIBLE

MUSCLE. Muscle & Nerve 42, 270, 2010.

64. Corona, B.T., Balog, E.M., Doyle, J.A., Rupp, J.C., Luke, R.C., andIngalls, C.P.

Junctophilin damage contributes to early strength deficits and EC coupling failure after

eccentric contractions. American Journal of Physiology-Cell Physiology 298, C365, 2010.

65. Hallcrag.Ec. RAPID DEGENERATION AND REGENERATION OF A WHOLE

SKELETAL-MUSCLE FOLLOWING TREATMENT WITH BUPIVACAINE

(MARCAIN). Experimental Neurology 43, 349, 1974.

66. Dalbis, A., Couteaux, R., Janmot, C., Roulet, A., andMira, J.C. REGENERATION

AFTER CARDIOTOXIN INJURY OF INNERVATED AND DENERVATED SLOW

AND FAST MUSCLES OF MAMMALS - MYOSIN ISOFORM ANALYSIS. European

Journal of Biochemistry 174, 103, 1988.

67. Machingal, M.A., Corona, B.T., Walters, T.J., Kesireddy, V., Koval, C.N.,

Dannahower, A., Zhao, W.X., Yoo, J.J., andChrist, G.J. A Tissue-Engineered Muscle

Repair Construct for Functional Restoration of an Irrecoverable Muscle Injury in a Murine

Model. Tissue Engineering Part A 17, 2291, 2011.

68. Valentin, J.E., Turner, N.J., Gilbert, T.W., andBadylak, S.F. Functional skeletal muscle

formation with a biologic scaffold. Biomaterials 31, 7475, 2010.

48
69. Merritt, E.K., Hammers, D.W., Tierney, M., Suggs, L.J., Walters, T.J., andFarrar, R.P.

Functional Assessment of Skeletal Muscle Regeneration Utilizing Homologous

Extracellular Matrix as Scaffolding. Tissue Engineering Part A 16, 1395, 2010.

70. Gamba, P.G., Conconi, M.T., Lo Piccolo, R., Zara, G., Spinazzi, R., andParnigotto,

P.P. Experimental abdominal wall defect repaired with acellular matrix. Pediatric Surgery

International 18, 327, 2002.

71. Burns, N.K., Jaffari, M.V., Rios, C.N., Mathur, A.B., andButler, C.E. Non-Cross-

Linked Porcine Acellular Dermal Matrices for Abdominal Wall Reconstruction. Plastic

and Reconstructive Surgery 125, 167, 2010.

72. Turner, N.J., Yates, A.J., Weber, D.J., Qureshi, I.R., Stolz, D.B., Gilbert, T.W.,

andBadylak, S.F. Xenogeneic Extracellular Matrix as an Inductive Scaffold for

Regeneration of a Functioning Musculotendinous Junction. Tissue Engineering Part A 16,

3309, 2010.

73. Turner, N.J., Badylak, J.S., Weber, D.J., andBadylak, S.F. Biologic Scaffold

Remodeling in a Dog Model of Complex Musculoskeletal Injury. Journal of Surgical

Research 176, 490, 2012.

74. Corona, B.T., Machingal, M.A., Criswell, T., Vadhavkar, M., Dannahower, A.C.,

Bergman, C., Zhao, W.X., andChrist, G.J. Further Development of a Tissue Engineered

Muscle Repair Construct In Vitro for Enhanced Functional Recovery Following

Implantation In Vivo in a Murine Model of Volumetric Muscle Loss Injury. Tissue

Engineering Part A 18, 1213, 2012.

49
75. Criswell, T.L., Corona, B.T., Ward, C.L., Miller, M., Patel, M., Wang, Z., Christ, G.J.,

andSoker, S. Compression-Induced Muscle Injury in Rats That Mimics Compartment

Syndrome in Humans. American Journal of Pathology 180, 787, 2012.

76. Corona, B.T., Ward, C.L., Baker, H.B., Walters, T.J., andChrist, G.J. Implantation of

In Vitro Tissue Engineered Muscle Repair Constructs and Bladder Acellular Matrices

Partially Restore In Vivo Skeletal Muscle Function in a Rat Model of Volumetric Muscle

Loss Injury. Tissue Engineering Part A 20, 705, 2014.

77. Sato, K., Li, Y., Foster, W., Fukushima, K., Badlani, N., Adachi, N., Usas, A., Fu,

F.H., andHuard, J. Improvement of muscle healing through enhancement of muscle

regeneration and prevention of fibrosis. Muscle & Nerve 28, 365, 2003.

78. Menetrey, J., Kasemkijwattana, C., Day, C.S., Bosch, P., Vogt, M., Fu, F.H.,

Moreland, M.S., andHuard, J. Growth factors improve muscle healing in vivo. Journal of

Bone and Joint Surgery-British Volume 82B2000.

79. Doukas, J., Blease, K., Craig, D., Ma, C.L., Chandler, L.A., Sosnowski, B.A.,

andPierce, G.F. Delivery of FGF genes to wound repair cells enhances arteriogenesis and

myogenesis in skeletal muscle. Molecular Therapy 5, 517, 2002.

80. Floss, T., Arnold, H.H., andBraun, T. A role for FGF-6 in skeletal muscle regeneration.

Genes & Development 11, 2040, 1997.

81. Fiore, F., Sebille, A., andBirnbaum, D. Skeletal muscle regeneration is not impaired

in Fgf6 -/- mutant mice. Biochemical and Biophysical Research Communications 272, 138,

2000.

50
82. Mitchell, C.A., McGeachie, J.K., andGrounds, M.D. The exogenous administration of

basic fibroblast growth factor to regenerating skeletal muscle in mice does not enhance the

process of regeneration. Growth Factors 13, 37, 1996.

83. Barnard, W., Bower, J., Brown, M.A., Murphy, M., andAustin, L. LEUKEMIA

INHIBITORY FACTOR (LIF) INFUSION STIMULATES SKELETAL-MUSCLE

REGENERATION AFTER INJURY - INJURED MUSCLE EXPRESSES LIF

MESSENGER-RNA. Journal of the Neurological Sciences 123, 108, 1994.

84. White, J.D., Bower, J.J., Kurek, J.B., andAustin, L. Leukemia inhibitory factor

enhances regeneration in skeletal muscles after myoblast transplantation. Muscle & Nerve

24, 695, 2001.

85. Foster, W., Li, Y., Usas, A., Somogyi, G., andHuard, J. Gamma interferon as an

antifibrosis agent in skeletal muscle. Journal of Orthopaedic Research 21, 798, 2003.

86. Gharaibeh, B., Chun-Lansinger, Y., Hagen, T., Ingham, S.J.M., Wright, V., Fu, F.,

andHuard, J. Biological Approaches to Improve Skeletal Muscle Healing After Injury and

Disease. Birth Defects Research Part C-Embryo Today-Reviews 96, 82, 2012.

87. Irintchev, A., Langer, M., Zweyer, M., Theisen, R., andWernig, A. Functional

improvement of damaged adult mouse muscle by implantation of primary myoblasts.

Journal of Physiology-London 500, 775, 1997.

88. Coulet, B., Lacombe, F., Lazerges, C., Daussin, P.A., Rossano, B., Micallef, J.P.,

Chammas, M., Reyne, Y., andBacou, F. Short- or long-term effects of adult myoblast

transfer on properties of reinnervated skeletal muscles. Muscle & Nerve 33, 254, 2006.

89. Bacou, F., el Andalousi, R.B., Daussin, P.A., Micallef, J.P., Levin, J.M., Chammas,

M., Casteilla, L., Reyne, Y., andNougues, J. Transplantation of adipose tissue-derived

51
stromal cells increases mass and functional capacity of damaged skeletal muscle. Cell

Transplantation 13, 103, 2004.

90. Matziolis, G., Winkler, T., Schaser, K., Wiemann, M., Krocker, D., Tuischer, J., Perka,

C., andDuda, G.N. Autologous bone marrow-derived cells enhance muscle strength

following skeletal muscle crush injury in rats. Tissue Engineering 12, 361, 2006.

91. Winkler, T., von Roth, P., Matziolis, G., Mehta, M., Perka, C., andDuda, G.N. Dose-

Response Relationship of Mesenchymal Stem Cell Transplantation and Functional

Regeneration After Severe Skeletal Muscle Injury in Rats. Tissue Engineering Part A 15,

487, 2009.

92. Pecanha, R., Bagno, L., Ribeiro, M.B., Ferreira, A.B.R., Moraes, M.O., Zapata-Sudo,

G., Kasai-Brunswick, T.H., Campos-de-Carvalho, A.C., Goldenberg, R.C.D.,

andWerneck-de-Castro, J.P.S. Adipose-Derived Stem-Cell Treatment of Skeletal Muscle

Injury. Journal of Bone and Joint Surgery-American Volume 94A, 609, 2012.

93. von Roth, P., Duda, G.N., Radojewski, P., Preininger, B., Perka, C., andWinkler, T.

Mesenchymal Stem Cell Therapy Following Muscle Trauma Leads to Improved Muscular

Regeneration in Both Male and Female Rats. Gender Medicine 9, 129, 2012.

94. Ferrari, G., Cusella-De Angelis, G., Coletta, M., Paolucci, E., Stornaiuolo, A., Cossu,

G., andMavilio, F. Muscle regeneration by bone marrow derived myogenic progenitors.

Science 279, 1528, 1998.

95. Beauchamp, J.R., Morgan, J.E., Pagel, C.N., andPartridge, T.A. Dynamics of myoblast

transplantation reveal a discrete minority of precursors with stem cell-like properties as the

myogenic source. Journal of Cell Biology 144, 1113, 1999.

52
96. Qu, Z.Q., Balkir, L., van Deutekom, J.C.T., Robbins, P.D., Pruchnic, R., andHuard, J.

Development of approaches to improve cell survival in myoblast transfer therapy. Journal

of Cell Biology 142, 1257, 1998.

97. Beattie, A.J., Gilbert, T.W., Guyot, J.P., Yates, A.J., andBadylak, S.F. Chemoattraction

of Progenitor Cells by Remodeling Extracellular Matrix Scaffolds. Tissue Engineering Part

A 15, 1119, 2009.

98. Farhat, W.A., Chen, J., Haig, J., Antoon, R., Litman, J., Sherman, C., Derwin, K.,

andYeger, H. Porcine bladder acellular matrix (ACM): protein expression, mechanical

properties. Biomedical Materials 3, 8, 2008.

99. Teodori, L., Costa, A., Marzio, R., Perniconi, B., Coletti, D., Adamo, S., Gupta, B.,

andTarnok, A. Native extracellular matrix: a new scaffolding platform for repair of

damaged muscle. Frontiers in physiology 5, 218, 2014.

100. Brzoska, E., Ciemerych, M.A., Przewozniak, M., andZimowska, M. REGULATION

OF MUSCLE STEM CELLS ACTIVATION: THE ROLE OF GROWTH FACTORS

AND EXTRACELLULAR MATRIX. In: Litwack G., ed. Vitamins and Hormones: Stem

Cell Regulators. San Diego: Elsevier Academic Press Inc; 2012. pp. 239.

101. Hinds, S., Bian, W.N., Dennis, R.G., andBursac, N. The role of extracellular matrix

composition in structure and function of bioengineered skeletal muscle. Biomaterials 32,

3575, 2011.

102. Badylak, S., Kokini, K., Tullius, B., Simmons-Byrd, A., andMorff, R. Morphologic

study of small intestinal submucosa as a body wall repair device. Journal of Surgical

Research 103, 190, 2002.

53
103. Wolf, M.T., Daly, K.A., Reing, J.E., andBadylak, S.F. Biologic scaffold composed

of skeletal muscle extracellular matrix. Biomaterials 33, 2916, 2012.

104. Corona, B.T., Wu, X., Ward, C.L., McDaniel, J.S., Rathbone, C.R., andWalters, T.J.

The promotion of a functional fibrosis in skeletal muscle with volumetric muscle loss

injury following the transplantation of muscle-ECM. Biomaterials 34, 3324, 2013.

105. Sicari, B.M., Rubin, J.P., Dearth, C.L., Wolf, M.T., Ambrosio, F., Boninger, M.,

Turner, N.J., Weber, D.J., Simpson, T.W., Wyse, A., Brown, E.H.P., Dziki, J.L., Fisher,

L.E., Brown, S., andBadylak, S.F. An Acellular Biologic Scaffold Promotes Skeletal

Muscle Formation in Mice and Humans with Volumetric Muscle Loss. Science

Translational Medicine 6, 10, 2014.

106. Altman, G.H., Diaz, F., Jakuba, C., Calabro, T., Horan, R.L., Chen, J.S., Lu, H.,

Richmond, J., andKaplan, D.L. Silk-based biomaterials. Biomaterials 24, 401, 2003.

107. Hoffman, A.S. Hydrogels for biomedical applications. Advanced Drug Delivery

Reviews 54, 3, 2002.

108. van Blitterswijk, C.e.a. Tissue Engineering. 1st ed. San Diego, CA: Elsevier; 2008.

109. Gobin, A.S., Butler, C.E., andMathur, A.B. Repair and regeneration of the abdominal

wall musculofascial defect using silk fibroin-chitosan blend. Tissue Engineering 12, 3383,

2006.

110. Borselli, C., Storrie, H., Benesch-Lee, F., Shvartsman, D., Cezar, C., Lichtman, J.W.,

Vandenburgh, H.H., andMooney, D.J. Functional muscle regeneration with combined

delivery of angiogenesis and myogenesis factors. Proceedings of the National Academy of

Sciences of the United States of America 107, 3287, 2010.

54
111. Corona, B.T., Wu, X., andWalters, T.J. Treatment of volumetric muscle loss injury

with muscle-derived ECM and BMSCs promotes functional recovery in rat tibialis anterior

muscle. Journal of Tissue Engineering and Regenerative Medicine 6, 117, 2012.

112. Merritt, E.K., Cannon, M.V., Hammers, D.W., Le, L.N., Gokhale, R., Sarathy, A.,

Song, T.J., Tierney, M.T., Suggs, L.J., Walters, T.J., andFarrar, R.P. Repair of Traumatic

Skeletal Muscle Injury with Bone-Marrow-Derived Mesenchymal Stem Cells Seeded on

Extracellular Matrix. Tissue Engineering Part A 16, 2871, 2010.

113. Rossi, C.A., Flaibani, M., Blaauw, B., Pozzobon, M., Figallo, E., Reggiani, C.,

Vitiello, L., Elvassore, N., andDe Coppi, P. In vivo tissue engineering of functional skeletal

muscle by freshly isolated satellite cells embedded in a photopolymerizable hydrogel.

Faseb Journal 252011.

114. Page, R.L., Malcuit, C., Vilner, L., Vojtic, I., Shaw, S., Hedblom, E., Hu, J., Pins,

G.D., Rolle, M.W., andDominko, T. Restoration of Skeletal Muscle Defects with Adult

Human Cells Delivered on Fibrin Microthreads. Tissue Engineering Part A 172011.

115. Beier, J.P., Stern-Straeter, J., Foerster, V.T., Kneser, U., Stark, G.B., andBach, A.D.

Tissue engineering of injectable muscle: Three-dimensional myoblast-fibrin injection in

the syngeneic rat animal model. Plastic and Reconstructive Surgery 118, 1113, 2006.

116. Wang, L., Cao, L., Shansky, J., Wang, Z., Mooney, D., andVandenburgh, H.

Minimally Invasive Approach to the Repair of Injured Skeletal Muscle With a Shape-

memory Scaffold. Molecular therapy : the journal of the American Society of Gene

Therapy 22, 1441, 2014.

117. Stratos, I., Madry, H., Rotter, R., Weimer, A., Graff, J., Cucchiarini, M., Mittlmeier,

T., andVollmar, B. Fibroblast Growth Factor-2-Overexpressing Myoblasts Encapsulated

55
in Alginate Spheres Increase Proliferation, Reduce Apoptosis, Induce Adipogenesis, and

Enhance Regeneration Following Skeletal Muscle Injury in Rats. Tissue Engineering Part

A 17, 2867, 2011.

118. Rangarajan, S., Madden, L., andBursac, N. Use of Flow, Electrical, and Mechanical

Stimulation to Promote Engineering of Striated Muscles. Annals of Biomedical

Engineering 42, 1391, 2014.

119. Donnelly, K., Khodabukus, A., Philp, A., Deldicque, L., Dennis, R.G., andBaar, K.

A Novel Bioreactor for Stimulating Skeletal Muscle In Vitro. Tissue Engineering Part C-

Methods 16, 711, 2010.

120. Candiani, G., Riboldi, S.A., Sadr, N., Lorenzoni, S., Neuenschwander, P.,

Montevecchi, F.M., andMantero, S. Cyclic mechanical stimulation favors myosin heavy

chain accumulation in engineered skeletal muscle constructs. Journal of Applied

Biomaterials & Biomechanics 8, 68, 2010.

121. Powell, C.A., Smiley, B.L., Mills, J., andVandenburgh, H.H. Mechanical stimulation

improves tissue-engineered human skeletal muscle. American Journal of Physiology-Cell

Physiology 283, C1557, 2002.

122. Moon, D.G., Christ, G., Stitzel, J.D., Atala, A., andYoo, J.J. Cyclic mechanical

preconditioning improves engineered muscle contraction. Tissue Engineering Part A 14,

473, 2008.

123. Levine, D.W., Mondano, L., andHalpin, M. FDA Regulatory Pathways for Knee

Cartilage Repair Products. Sports Medicine and Arthroscopy Review 16, 202, 2008.

56
124. Sierpinski, P., Garrett, J., Ma, J., Apel, P., Klorig, D., Smith, T., Koman, L.A., Atala,

A., andVan Dyke, M. The use of keratin biomaterials derived from human hair for the

promotion of rapid regeneration of peripheral nerves. Biomaterials 29, 118, 2008.

125. Moll, R., Divo, M., andLangbein, L. The human keratins: biology and pathology.

Histochemistry and Cell Biology 129, 705, 2008.

126. Rouse, J.G., andVan Dyke, M.E. A Review of Keratin-Based Biomaterials for

Biomedical Applications. Materials 3, 999, 2010.

127. Yu, J.L., Yu, D.W., Checkla, D.M., Freedberg, I.M., andBertolino, A.P. HUMAN

HAIR KERATINS. Journal of Investigative Dermatology 101, S56, 1993.

128. Fraser, R.D.B., Macrae, T.P., Parry, D.A.D., andSuzuki, E. INTERMEDIATE

FILAMENTS IN ALPHA-KERATINS. Proceedings of the National Academy of Sciences

of the United States of America 83, 1179, 1986.

129. Richter, J.R., de Guzman, R.C., Greengauz-Roberts, O.K., andVan Dyke, M.

Structure-property relationships of meta-kerateine biomaterials derived from human hair.

Acta Biomaterialia 8, 274, 2012.

130. de Guzman, R.C., Merrill, M.R., Richter, J.R., Hamzi, R.I., Greengauz-Roberts, O.K.,

andVan Dyke, M.E. Mechanical and biological properties of keratose biomaterials.

Biomaterials 32, 8205, 2011.

131. Verma, V., Verma, P., Ray, P., andRay, A.R. Preparation of scaffolds from human

hair proteins for tissue-engineering applications. Biomedical Materials 3, 12, 2008.

132. Tachibana, A., Furuta, Y., Takeshima, H., Tanabe, T., andYamauchi, K. Fabrication

of wool keratin sponge scaffolds for long-term cell cultivation. Journal of Biotechnology

93, 165, 2002.

57
133. Burnett, L.R., Rahmany, M.B., Richter, J.R., Aboushwareb, T.A., Eberli, D., Ward,

C.L., Orlando, G., Hantgan, R.R., andVan Dyke, M.E. Hemostatic properties and the role

of cell receptor recognition in human hair keratin protein hydrogels. Biomaterials 34, 2632,

2013.

134. Fearing, B.V., andVan Dyke, M.E. In vitro response of macrophage polarization to a

keratin biomaterial. Acta Biomaterialia 10, 3136, 2014.

135. .

136. Hill, P.S., Apel, P.J., Barnwell, J., Smith, T., Koman, L.A., Atala, A., andVan Dyke,

M. Repair of Peripheral Nerve Defects in Rabbits Using Keratin Hydrogel Scaffolds.

Tissue Engineering Part A 17, 1499, 2011.

137. Shen, D., Wang, X., Zhang, L., Zhao, X., Li, J., Cheng, K., andZhang, J. The

amelioration of cardiac dysfunction after myocardial infarction by the injection of keratin

biomaterials derived from human hair. Biomaterials 32, 9290, 2011.

138. Lin, Y.-C., Ramadan, M., Van Dyke, M., Kokai, L.E., Philips, B.J., Rubin, J.P.,

andMarra, K.G. Keratin Gel Filler for Peripheral Nerve Repair in a Rodent Sciatic Nerve

Injury Model. Plastic and Reconstructive Surgery 129, 67, 2012.

139. Pace, L.A., Plate, J.F., Mannava, S., Barnwell, J.C., Koman, L.A., Li, Z., Smith, T.L.,

andVan Dyke, M. A Human Hair Keratin Hydrogel Scaffold Enhances Median Nerve

Regeneration in Nonhuman Primates: An Electrophysiological and Histological Study.

Tissue Engineering Part A 20, 507, 2014.

140. Kowalczewski, C.J., Tombyln, S., Wasnick, D.C., Hughes, M.R., Ellenburg, M.D.,

Callahan, M.F., Smith, T.L., Van Dyke, M.E., Burnett, L.R., andSaul, J.M. Reduction of

58
ectopic bone growth in critically-sized rat mandible defects by delivery of rhBMP-2 from

kerateine biomaterials. Biomaterials 35, 3220, 2014.

141. Bach, A.D., Beier, J.P., Stern-Staeter, J., andHorch, R.E. Skeletal muscle tissue

engineering. Journal of Cellular and Molecular Medicine 8, 413, 2004.

142. Quintero, A.J., Wright, V.J., Fu, F.H., andHuard, J. Stem Cells for the Treatment of

Skeletal Muscle Injury. Clinics in Sports Medicine 28, 1, 2009.

143. Levenberg, S., Rouwkema, J., Macdonald, M., Garfein, E.S., Kohane, D.S., Darland,

D.C., Marini, R., van Blitterswijk, C.A., Mulligan, R.C., D'Amore, P.A., andLanger, R.

Engineering vascularized skeletal muscle tissue. Nature Biotechnology 23, 879, 2005.

144. Kamelger, F.S., Marksteiner, R., Margreiter, E., Klima, G., Wechselberger, G.,

Hering, S., andPiza, H. A comparative study of three different biomaterials in the

engineering of skeletal muscle using a rat animal model. Biomaterials 25, 1649, 2004.

59
CHAPTER II

Cell and Growth Factor Loaded Keratin Hydrogels for Treatment of Volumetric Muscle

Loss in a Mouse Model

Hannah B. Bakera,b; Juliana A. Passipieria,c; Mevan Siriwardanea; Mary D. Ellenburgd;

Justin M. Saule; Luke Burnettd; Seth Tomblynd; George J. Christa,b,c,f

Affiliations:
a
Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-

Salem, NC 27157, USA


b
VT-WFU School of Biomedical Engineering and Sciences, Winston-Salem, NC 27157
c
Biomedical Engineering Department, University of Virginia, Charlottesville, VA, 22908,

USA
d
KeraNetics, LLC, Suite 168, 391 Technology Way, Winston-Salem, NC 27101
e
Department of Chemical and Paper Engineering, Miami University, Oxford, OH 45056,

USA
f
Orthopedics Department, University of Virginia, Charlottesville, VA, 22908, USA

Note: This manuscript was prepared by Hannah B. Baker and has not yet been submitted.

She (primarily) and Dr. Juliana A. Passipieri (secondarily) were responsible for the

majority of the work described.

60
Introduction

Skeletal muscle injuries and disorders which result in permanent loss of function

have been defined as Volumetric Muscle Loss injuries by Grogan et al (1). Such injuries

are common among active military personnel wherein musculoskeletal trauma makes up

60-70% of all injuries (2). In addition to traumatic injuries, VML can also arise as a result

of excision of cancerous tissue, congenital birth defects such as cleft lip and cleft palate,

and as a result of acquired diseases such as Bell’s palsy (3-6) . Presently, the gold standard

of treatment for VML is an autologous or cadaveric muscle graft. While these interventions

have become increasingly successful with advances in microsurgery, flap transfer

procedures still suffer from donor-host compatibility in the case of cadaveric tissue and

from donor site morbidity when using autologous tissues (7-9). Current efforts in tissue

engineering research seek to develop of a variety of therapies which overcome these issues

and are capable of meeting the needs of the wide variety of VML presentations.

Researchers have investigated several biomaterial scaffolds including synthetic and

biologically derived materials for use in skeletal muscle indications. Biologic materials are

of particular interest in tissue engineering applications due to their availability and

favorable structural and chemical characteristics (10-12). Investigators have applied a

variety of biologic materials alone or in concert with cells, drugs, or factors in several

different animal models of muscle injury (13, 14). Among these approaches, extracellular

matrix (ECM) derived materials have shown great promise for use in tissue engineering

applications and a number of ECM constructs have been FDA approved for several

indications including soft tissue repair, rotator cuff reinforcement, and superficial wound

healing (10). Prior work in our group has focused on the use of such an ECM scaffold

61
coupled with cells and physiologic preconditioning in rodent models of VML. In studies

using this approach, we observed a significant improvement in functional recovery (15-

17). In building a platform of technologies capable of treating a wide variety of VML

indications, our group has begun to explore other biologic scaffold materials including

keratin hydrogels, the subject of this study.

Keratin biomaterials have a host of properties favorable for regenerative medicine

applications and pertinent to skeletal muscle repair. Isolated keratin proteins allow cell

attachment via known cell recognition sequences (18, 19) and reductively isolated keratins

(kerateines) have been found to promote platelet adhesion and coagulation (20). Keratin

hydrogels are capable of achieving sustained release of drugs and growth factors (21-23).

Furthermore, keratin biomaterials have been shown to induce differentiation of monocytes

into the favorable pro-regeneration type-2 population of macrophage (24). As it would

follow, these properties of keratin biomaterials have afforded positive results in a wide

range of regenerative medicine applications including burn healing, bone regeneration, and

nerve regeneration and have demonstrated potential for use in skeletal muscle repair

applications.

Trichocytic keratins which make up human hair are classified as alpha, beta, and

gamma wherein the alpha fraction makes up the majority of the hair fibers, the beta fraction

is present in the cortex of the fibers, and the globular gamma fraction provides the

supporting matrix around filaments (12). The keratins can be solubilized through disruption

of the disulfide bonds to open the cortex and denature the fibers into constitutive proteins

through either an oxidative or reductive process (25, 26). Oxidative extraction yields

keratins with altered cysteine residues that can no longer cross link with other keratins and

62
are referred to as keratose. Reductive isolation results in keratin referred to as kerateine in

which the cysteine group disulfide bonds are broken but the cysteine residues are unaltered

and able to reform disulfide bonds. As a result keratose has a faster degradation rate than

kerateine and the two can be combined to achieve a desired degradation rate.

In the current study, several properties of keratin hydrogels were utilized in

developing and employing a construct to promote muscle repair. Keratin hydrogels made

up of the kerateine and keratose were combined in a manner to achieve degradation within

8 weeks and sustained release of growth factors over this time. In addition, keratin

hydrogels served as a delivery vehicle for autologous skeletal muscle progenitor cells

(MPCs) to the site of injury. Insulin-like growth factor-1 (IGF-1) and basic fibroblast

growth factor (bFGF) were chosen for investigation based upon their known effects on the

skeletal muscle cell population. IGF-1 has been demonstrated to promote differentiation of

satellite cells and fusion of myoblasts while studies using bFGF indicated it promotes

satellite cell activation and proliferation (27-31).

The overriding hypothesis driving this study is that keratin hydrogels will promote

regeneration of skeletal muscle in VML by encouraging a favorable environment for tissue

repair and regeneration. It is further hypothesized that the addition of cells and growth

factors known to influence the skeletal muscle cell population will complement and

enhance the regenerative capacity of keratin alone. To investigate this hypothesis, keratin

hydrogels with or without the addition of a cellular component, growth factors, or a

combination will be examined in an established model of VML for their ability to enable

restoration of function and native morphology.

63
Methods

Keratin Extraction

Keratins were extracted using a patented process in a quality system

regulation/good manufacturing process, QSR/GMP, facility at KeraNetics, LLC. Briefly,

a cold solution of 0.5M thioglycolic acid (TGA) in sodium hydroxide (reductive

extraction for kerateine) or a 2% peracetic acid (PAA) solution (oxidative extraction for

keratose) were added to chopped human hair (chemicals were purchased from Sigma

Aldrich, St. Louis, MO; hair was purchased from World Response Group, Huntsville,

AL). For reductive extraction, the reducing solution was added to hair in a mixing tank

for 12 hours at 37 °C followed by two washes in 100mM Tris base (Sigma) and water.

The solution was then centrifuged, filtered, dialyzed against a 100kDa molecular weight

cutoff cellulose membrane, and neutralized to pH 7.4. For oxidative extraction with PAA,

the hair was handled in a similar manner. Both extractions were followed by acid

precipitation of the alpha fraction, centrifugation, re-dissolution in buffer, and dialyzed in

water against a 30 kDa nominal low molecular weight cutoff filter. The resulting

solutions were lyophilized, ground, and sterilized by 2Mrad gamma irradiation.

BAM Preparation

Briefly, porcine derived bladder was washed and trimmed to obtain the lamina

propria, which was placed in 0.05% trypsin (Hyclone, Logan, UT) for 1 h at 37 °C. The

bladder was then transferred to Dulbecco’s modified Eagle’s Medium, DMEM, solution

supplemented with 10% FBS and 1% antibiotic antimycotic, AA, (Hyclone) and kept

overnight at 4 °C. The preparation was then washed in a solution containing 1% triton-X

64
(Sigma) and 0.1% ammonium hydroxide (Fischer Scientific, Pittsburgh, PA) in de-ionized

water for 4 days at 4 °C. Finally, the bladder was then washed in deionized water for 3

days at 4 °C. The decellularized scaffold was further dissected to obtain a scaffold of 0.2–

0.4mm thickness. The scaffolds were then cut and draped onto custom made silicone molds

with a 4.5cm2 working area.

Cell Culture

Mouse MPCs were isolated from the tibialis anterior and soleus muscles of 4-8

week old C57/Bl6 mice purchased from Charles River Laboratories. Muscles were

sterilized in iodine and washed with PBS before transfer to a 0.2% w/v collagenase

(Worthington Biochemical, Lakewood, NJ) low glucose DMEM (Hyclone) solution where

they were finely minced and then allowed to further digest at 37 °C for 2 hours. The cell

slurry was plated on matrigel (BD Biosciences, San Jose, CA) coated tissue culture plastic

in myogenic media consisting of high glucose DMEM supplemented with 20% fetal bovine

FBS, 10% horse serum (HS), 1% chick embryo extract, and 1% AA (Hyclone). Three days

after plating the media was changed to seeding media consisting of low glucose DMEM

supplemented with 15% FBS and 1% AA (Hyclone). At 70-80% confluence cells were

passaged.

KOS:KTN Optimization and Cell Capacity

Prior to investigating the hydrogel in the mouse model, the ratio of KOS to KTN

which enabled a sustained release of both growth factors was determined via ELISA

analysis of solution surrounding growth factor loaded hydrogels (data shown in appendix:

65
3). The gels were prepared with selected ratios of KOS to KTN in which varied

concentrations (w/v) of keratin powder to sterile PBS were also examined. The results

indicated that a 70:30 blend of 15% KOS and 7% KTN resulted in the sustained release of

both IGF-1 and bFGF over the course of a month (later time points not examined).

Based on rheology data collected in an ongoing study (data not shown), the elastic

modulus of hydrogel which allows incorporation of cells and may still be extruded through

a surgical syringe is ~100Pa. The maximum number of cells which could be incorporated

into the optimized blend and still yield a hydrogel which was flowable in nature and

capable of being extruded through a surgical syringe was 1.7x106 MPCs/ml in the KOS

fraction. It may be assumed that the addition of this concentration of cells resulted in a

hydrogel which did not have an elastic modulus significantly greater than 100Pa. As such

it is worth noting that the concentration of cells used in this study was limited by the

viscosity of the hydrogel.

Hydrogel Preparation

For the growth factor containing groups, IGF-1 and/or bFGF (Peprotech, Rocky

Hill, NJ) were diluted in sterile water to yield a final concentration of 100 µg/mL of each

growth factor. Growth factor solutions were added to KTN powders to make a 7% w/v

hydrogel. KOS was added to either PBS or a serum free media cell suspension of

~1.65x106/mL passage 2 mouse MPCs to form a 15% w/v hydrogel. The KTN and KOS

hydrogels were then combined at a 70:30 KOS:KTN ratio by passing the gels between

coupled syringes.

66
Mouse Latissimus Dorsi VML Model and Hydrogel Implant

Eight week old female C57/BL6 mice were purchased from Charles River

Laboratories. The VML model was created by resecting ~50% of the area of the LD

muscle, ~16±3 mg, as was done in previous studies (15, 17). After creation of the defect

animals were put into one of the following treatment groups: no repair (NR), BAM, keratin

alone, and keratin with a combination of cells, IGF-1, and bFGF (groups listed in Table I,

surgery shown in Figure 1). For the BAM treatment, scaffolds were folded and secured

with 6-0 vicryl (Ethicon, Sommerville, NJ) at the proximal and distal ends of the defect.

For the NR and keratin groups, after creation of the defect, the fascia and mammary fat pad

were closed with 6-0 vicryl. For treatment with keratin hydrogels, 0.1-0.2ml of the

hydrogel was injected into the fascial pocket over the injury site. In all groups the skin was

then sutured with 5-0 prolene (Ethicon) and animals were allowed to recover. At 8 weeks

post implant, animals were sacrificed and the experimental and contralateral LD muscles

were explanted for functional and histological analysis.

67
TABLE I. TREATMENT GROUP COMPONENTS
Treatment Group Number of Components
(abbreviation) animals (n) Keratin MPCs IGF-1 bFGF
No Repair
9
(NR)
Bladder Acellular Matrix
8
(BAM)
Keratin
(KN)
6 +
Keratin+IGF-1
(KN+I)
8 + +
Keratin+bFGF
(KN+b)
8 + +
Keratin+IGF1+bFGF
(KN+I+b)
8 + + +
Keratin+MPC
(KN+M)
8 + +
Keratin+MPC+IGF-1
(KN+M+I)
8 + + +
Keratin+MPC+bFGF
(KN+M+ b)
9 + + +
Keratin+MPC+IGF-1+bFGF
(KN+M+I+b)
8 + + +

Table I. List of Groups, Components, and Number of Animals (n)

68
Figure 1. Surgical flow of VML Injury and Keratin Implant. Muscle is isolated

in (A) with proposed defect region outlined; defect is created and outlined in (B).

Fascia is partially closed and keratin hydrogel is injected into the fascial pocket

(C). Fascia and skin are then closed and the animal is allowed to recover.

Muscle Function Analysis

The entire LD muscle was isolated from the thoracolumbar fascia to the humeral

tendon from mice while under anesthesia and the tendon and facial ends were tied with 5-

0 silk suture (Ethicon) and transferred to ice cold Krebs-Ringer buffer solution (Sigma;

composition: pH 7.4; concentration in mM: 121.0 NaCl, 5.0 KCl, 0.5 MgCl2, 1.8 CaCl2,

24.0 NaHCO3, 0.4NaH2PO4, and 5.5 glucose). Muscles were transferred to individual

chambers of a DMT 750 tissue organ bath system (DMT, Ann Arbor, MI) filled with

Krebs-Ringer buffer at 35 °C bubbled with 95% O2 and 5% CO2. The muscles were

positioned between custom-made platinum electrodes with the proximal tendon attached

to a force transducer and the distal tendon to a fixed support. Direct muscle electrical

stimulation (0.2ms pulse at 30 V) was applied across the LD muscle using a Grass S88

stimulator (Grass, Warwick, RI). Real time display and recording of all force

measurements were performed on a PC with Power Lab/8sp (AD Instruments, Colorado

Springs, CO). Once the LD muscles were mounted in the organ bath, the muscles were

69
allowed to equilibrate for 5 min prior to determining optimal physiological muscle length

(Lo) via a series of twitch contractions. Force as a function of stimulation frequency (1–

250Hz) was measured at 37 °C during isometric contractions (750ms trains of 0.2ms

pulses), with 2 min between contractions. The maximal force of contraction, Po, was

normalized to an approximate physiological cross-sectional area (PCSA), which was

calculated using the following equation, where muscle density is 1.06 g/cm3:

𝑃𝑃𝑃𝑃𝑃𝑃𝑃𝑃 = (𝑤𝑤𝑤𝑤𝑤𝑤 𝑤𝑤𝑤𝑤𝑤𝑤𝑤𝑤ℎ𝑡𝑡 (𝑔𝑔)/ [𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚 𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑 (𝑔𝑔/𝑐𝑐𝑐𝑐3) 𝑥𝑥 𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚 𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙ℎ, 𝐿𝐿𝑜𝑜 (𝑐𝑐𝑐𝑐)]

Equation 1. Physiological Cross Sectional Area (PCSA) Calculation

Additionally the force-frequency curves were fit to the following dose response equation:
𝑓𝑓(𝑥𝑥) = 𝑚𝑚𝑚𝑚𝑚𝑚 − (𝑚𝑚𝑚𝑚𝑚𝑚 − min)/[1 + (𝑥𝑥 ⁄ 𝐸𝐸𝐸𝐸 50 )−𝑛𝑛 ]

Equation 2. Dose Response Formula

where x is the stimulation frequency in Hz, min is the lowest observed force generated,

equivalent to the twitch force or Pt, max is the largest observed force or Po, EC50 is the

stimulation frequency which yields half the amplitude of the maximum observed force (Po-

Pt), and n is the slope of the linear portion of the force-frequency curve.

Immunohistochemistry and Imaging

Hematoxylin and eosin and Masson’s trichrome stains were conducted by standard

techniques to determine the basic morphology of cells in and around the implant and to

observe any inflammatory response. Immunohistochemical staining was performed using

antibody to detect myosin (MF-20, 1:10) acquired from Developmental Studies Hybridoma

Bank, Iowa City, IA. Biotinylated anti-mouse IgG (MKB-2225, 1:250, Vector Laboratories

Inc.) secondary antibody was used to detect primary antibodies. The sections were next

70
treated with Avidin Biotin Complex Reagent (PK-7100, Vector Laboratories Inc.) and then

visualized using a NovaRED substrate kit (SK-4800, Vector Laboratories Inc.). Finally,

the sections were counterstained using Gill’s Hematoxylin (GHS280, Sigma-Aldrich).

Tissue sections without primary antibody were used as negative controls. Images were

captured and digitized (DM4000B Leica Upright Microscope).

Statistical Analysis

Functional data were statistically analyzed using one way ANOVA. When

significance was found (α set to 0.05) a post-hoc multiple comparison test was used to

compare group means (α set to 0.05) using a Fisher Least Significant Difference (LSD)

significance test. Statistical analysis was conducted using GraphPad Prism 6.0 for

Windows La Jolla, California.

Results

In Vitro Functional Assessment

In vitro force measurement was conducted as described at 8 weeks after implant on

whole LD operated and contralateral control muscles harvested from the ten treatment

groups referenced in Table I. Measured physical and functional parameters for each group

and parameter statistics are listed in Table II, charts of Po and specific Po are shown in

Figure 2.

71
72
Table II. Summary of Latissimus Dorsi Physical and Functional Measurement

Data. Values are mean ± standard error of the mean. Muscle mass was measured

after completion of functional testing. Lo is the optimal muscle length associated

with peak twitch response (Pt). PCSA was determined using the measured Lo and

wet weight using Equation 1. Peak twitch (Pt), contraction force at 80hz (P80), and

maximal isometric contraction force (Po) were measured by soliciting contractions

via direct electric stimuli (0.2ms, 30V) in the organ bath setup. Measured Po was

normalized to PCSA to determine specific force (Specific Po). The stimulation

frequency which yielded 50% of the absolute maximum amplitude (EC50) and the

slope of the linear portion of the force-frequency curves shown in Fig. 3 (n

coefficient) were determined by fitting the curves to function described by

Equation 2. Superscript denotations indicate statistically different group means (p

< 0.05) from: aUninjured, bNR, cBAM, dKN, eKN+IGF-1, fKN+ bFGF, gKN+IGF-
h
1+bFGF, KN+MPC, iKN+MPC+IGF-1, jKN+MPC+bFGF, kKN+MPC+IGF-

1+bFGF, and lall other groups.

73
74
Figure 2. Muscle Function Analysis at 2 Months Post-Surgery. Maximum isometric contraction force as a result of electrical

field stimulation (1-250hz, 0.2ms, 30V) (A) normalized to PCSA (B). For both measurements Uninjured n=46, NR n=9, BAM

n=8, KN n=6, KN+I n=8, KN+b n=8, KN+I+b n=8, KN+M n=8, KN+M+I n=8, KN+M+b n=9, KN+M+I+b n=8. Acellular

groups are colored green, cellularized are colored blue. Significant differences are denoted as follows: * p<0.05, ** p<0.01, ***

p<0.001, † different from all other groups p<0.001, # different from all other groups p<0.0001
The average created defect mass was 16.0±3 mg with no significant difference

between treatment groups. The injury created in this study resulted in a 58% reduction in

maximum isometric contraction force (Po), a 55% reduction in peak twitch force (Pt), and

a 64% reduction in measured submaximal tetanic contraction at 80hz (P80 Hz) in the No

Repair (NR) group relative to uninjured control muscles.

Among the treatment groups keratin+IGF-1+bFGF enabled the greatest recovery

of Po, Pt, P80 Hz, and had the highest specific Po. The Po values measured for KN+I+b treated

muscles were significantly greater than all treatment groups’ values excluding KN and

KN+M+I. Measured Pt values were also significantly greater than KN+b, KN+M, and

KN+M+I+b and specific Po values for KN+I+b were significantly greater than BAM and

KN+b values. In addition KN+M+I treatments also resulted in specific Po values which

were significantly higher than BAM and KN+b values.

Analysis of dose response curves fit to force-frequency data (selected curves shown

in Figure 3) indicated that EC50 values for all treatments were increased relative to control

values while curve slope values were not different between any groups (Table II). EC50 for

the KN+I+b group had the largest shift relative to uninjured values and was significantly

different from EC50 values for NR, BAM, KN+b, KN+M, and KN+M+I.

Muscle masses of BAM treated muscles were significantly greater than those of

any other group including uninjured tissues, equating to 176% of control values. KN+b

treated muscles were also significantly heavier than control tissues with values measuring

140% of uninjured tissue values. Similarly, BAM treated tissues had the longest optimal

muscle Lo of the treatment groups and this measurement was not significantly different

from uninjured muscles.

75
Figure 3. Selected Force-Frequency Curves with Fitted Dose-Response

Curves. Two months post-surgery contractions elicited via electrical stimuli from

1-250Hz is shown for Uninjured, KN+I+b, BAM, and NR muscles. Data has been

fit with dose response curves according to Equation 2. Curve fitting was used to

determine EC50 and Hill Slope values shown in Table II.

Morphology

Upon completion of functional analysis and physical measurements, experimental

and control tissues were placed next to one another in tissue culture plastic for

morphological comparison. Representative tissue samples from each treatment group are

shown in Figure 4. Among treatment groups, it was observed that BAM and KN+b treated

muscles were consistently larger in size and carried more fatty-connective tissue than the

other treatment groups’ tissue. While uninjured tissues had a distinct organized vasculature

compared to that seen in experimental muscles, a notable difference in vasculature

structures among the groups was not observable at the gross morphology level.

76
Figure 4. Representative Tissue Gross Morphology. Images shown were taken

following functional assessment carried out at 2 months post-surgery.

Representative samples from each group are shown.

77
Histology

All tissues were subjected to histological processing as described. Depicted in

Figures 5-7 are images of uninjured and experimental tissue sections stained with

hematoxylin and eosin, Masson’s Trichrome, and for the contractile protein myosin. Tissue

sections were examined at the interface between injured and uninjured tissue for the

presence of adipose and collagen deposition, and for evidence of reorganization and

regeneration of muscle tissue.

In NR tissues adipose and collagen deposition are present along the injury site with

mononuclear cells evenly distributed throughout the defect site. A small extent of new

muscle formation was evidenced with short and thin fibers dispersed along the region

between native tissue and the observed band of adipose and scar tissue. In BAM treated

muscles the injury-native tissue border resembles that seen in NR tissue with a much

greater volume of collagen present consistent with the presence of non-remodeled BAM

scaffold.

78
79
Figure 5. Mouse LD Cell and Tissue Morphology and Functional Protein

Expression in Uninjured, NR, and BAM Treated Tissues. Treatment groups

presented: uninjured control (A, D, G), NR (B, E, H), BAM (C, F, I). Cell and tissue

morphology was examined through hematoxylin and eosin staining (A-C) wherein

nuclei are stained blue-purple and cytoplasm and cellular proteins are stained pink

and Masson’s trichrome staining (D-F) in which tissue stains red, collagen stains

blue, and nuclei stain black. The presence of new muscle tissue formation within

the defect and treatment site was evaluated by staining for myosin (MF-20) shown

in brown (G-I). (*) denotes adipose tissue deposits; (#) denotes collagen

deposition/scar tissue; (‡) denotes native tissue; and new muscle formation is

indicated with a black arrow.

80
81
Figure 6. Mouse LD Morphology and Functional Protein Expression in KN,

KN+I, KN+b, and KN+I+b. Treatment groups presented: KN (A, E, I), KN+I (B,

F, J), and KN+b (C, G, K), and KN+I+b (D, H, L). Morphology was examined

through hematoxylin and eosin staining (A-D) wherein nuclei are stained blue-

purple and cytoplasm and cellular proteins are stained pink and Masson’s trichrome

staining (E-H) in which tissue stains red, collagen stains blue, and nuclei stain

black. The presence of new muscle tissue formation within the defect and treatment

site was evaluated by staining for myosin (MF-20) shown in brown (I-L). (*)

denotes adipose tissue deposits; (#) denotes collagen deposition/scar tissue; (‡)

denotes native tissue; and new muscle formation is indicated with a black arrow.

82
83
Figure 7. Mouse LD Morphology and Functional Protein Expression in

KN+M, KN+M+I, KN+M+b, and KN+M+I+b. Treatment groups presented:

KN+M (A, E, I), KN+M+I (B, F, J), KN+M+b (C, G, K), and KN+M+I+b (D, H,

L). Morphology was examined through hematoxylin and eosin staining (A-D)

wherein nuclei are stained blue-purple and cytoplasm and cellular proteins are

stained pink and Masson’s trichrome staining (E-H) in which tissue stains red,

collagen stains blue, and nuclei stain black. The presence of new muscle tissue

formation within the defect and treatment site was evaluated by staining for myosin

(MF-20) shown in brown (I-L). (*) denotes adipose tissue deposits; (#) denotes

collagen deposition/scar tissue; (‡) denotes native tissue; and new muscle

formation is indicated with a black arrow.

84
Cell morphology among the keratin treatment groups was varied with regard to the

presence of scar tissue, adipose and mononuclear cells, and neo-tissue formation.

Compared with BAM and NR groups, KN treated tissue showed much less collagen

presence, but a greater presence of adipose tissue and mononuclear cells which had formed

granulomas at the injury site. At the border of the defect site thin muscle fibers travelling

along the axis of the native tissue are evident with more continuous fibers present than in

the BAM and NR groups.

Unlike KN alone, KN+M tissue showed a large degree of scar tissue formation with

some adipose tissue deposition, a more disperse mononuclear cell presence, and a moderate

layer of organized but interrupted thin new muscle fibers. KN+I and KN+M+I+b had injury

borders that had larger layers of adipose tissue present with little to no scar tissue formation

and newly formed thin muscle fibers which traveled along the same axis as the native tissue

but did not infiltrate into the region of adipose tissue deposition- similar to that seen in

KN+M.

KN+I+b tissues had similar cell morphology to KN+M+I+b and KN+I tissues with

exception that muscle fibers could be observed up to and within the layer of adipose tissue

present. KN+b and KN+M+b tissues were similar in appearance to KN+M with a greater

presence of scar tissue as well as adipose deposition. Notably, KN+b tissues were observed

to have a greater extent of new muscle tissue formation, however the neo-tissues appeared

as rafts of muscle scattered and separated within the extensive fat and scar tissue present,

which also appeared to be interrupting the native tissue adjacent to the injury site.

85
Lastly, KN+M+I sections resembled the bFGF groups in that there was a large

presence of both fat and scar tissue, however in the KN+M+I tissues the deposition was

not as extensive or invasive as in the case of KN+b. In addition, the new muscle tissue

formation was more similar to that seen in KN+I+b in that it was visible within the scar

and fat deposition, but as an extension from the native tissue and not in rafts as in KN+b

tissues.

Discussion

In previous studies our group has developed two distinct models of VML injury in

rodents in which to examine the effectiveness of a BAM scaffold loaded with MPCs and

subjected to cyclic stretch preconditioning (15-17). With these established models it is

possible to evaluate additional technologies such as keratin hydrogels as potential

treatments for VML injuries. Keratin was a viable candidate for a muscle repair application

due to its known capacity to mitigate inflammation, to influence the coagulation cascade

and neurogenesis, and to act as a cell and drug delivery vehicle (18, 20, 23, 24, 32, 33).

Several studies have illustrated the importance of a cellular component in tissue

engineering technologies aimed to promote skeletal muscle repair and regeneration in

VML injuries (15-17, 34-40). Of these studies, those conducted by our group which

employed the same VML model used in the present study indicated that a cellular

component in addition to the scaffold used did not just improve function, but was necessary

for any functional improvement to occur (15, 17). Investigators have also shown that

growth factors alone or coupled with cell and scaffold based therapies can improve muscle

recovery after injury in vivo (37, 41, 42). These studies as well as in vitro investigations of

86
the effect of growth factors showcase the important role of IGF-1 and bFGF within

regenerating skeletal muscle (29-31). The culmination of these findings resulted in the

hypothesis for this study which is that keratin hydrogels will improve muscle repair and

regeneration in a VML injury model and that the regenerative capacity will be

complemented through the addition of a combination of MPCs, IGF-1, and bFGF. The

novelty of this work lies therein: this is the first known application of keratin hydrogels,

let alone the combination of keratin, MPCs, and growth factors, in an in vivo VML model.

The hypothesis of this study was addressed through use of an established injury

model and physiological and histochemical assays. The major goal of this work was to

determine the potential of keratin for use in such a model and to utilize the carrier capacity

of keratin hydrogels to further glean insight into the effectiveness of keratin in combination

with cells and growth factors. Furthermore, this study would allow for the study of the in

vivo interactions between keratin, cells, and growth factors in an active wound environment

and resulting effects on the repair of skeletal muscle.

Use of in vitro functional assessment indicated that the combination of keratin, IGF-

1, and bFGF resulted in the greatest recovery after injury according to metrics surrounding

contraction force (Po, Pt, P80 Hz, and Specific Po). Interestingly and unexpectedly, this

treatment yielded the greatest recovery of absolute and specific contraction force in the

absence of a cellular component. It is worth noting, however, that the functional results

observed for these measurements for KN+I+b treated muscles were not significantly

different from keratin alone or keratin coupled with cells and IGF-1. Further, the addition

of cells to the same combination keratin loaded with MPCs, IGF-1, and bFGF, KN+M+I+b,

resulted in a significantly lower Pt and Po than KN+I+b. This raises the question of how

87
the addition of cells influenced recovery. If the groups with cells are grouped together and

the same is done for the groups without cells, there is no significant difference between the

Po of the two groups (data not shown). In a previous study from our group in which cells

were coupled to an ECM scaffold, chemical and physical differentiation cues resulted in a

greater functional improvement at 2 months post-implant compared with the addition of

cells in the absence of differentiation cues (17). Similarly in the current study, cells were

combined with keratin and not cultured in a differentiation medium or subjected to

physiologic preconditioning which may account for why the addition of cells did not

impact functional recovery. Moreover and perhaps a more relevant comparison is the

concentration of cells applied in each model. In the previous studies utilizing this VML

model, implanted scaffolds carried ~6x106 MPCs (1x106 MPC/cm2 per side on a 1x3cm

construct) compared with ~1.2-2.3x105 MPCs in the injected volume of hydrogel used for

the cell groups in this study which constitutes a 26-50 fold difference.

Further investigation using cell morphology analysis gives insight into how the

cells and growth factors may have impacted the resulting functional outcomes. While the

cell morphology observed among the keratin treatment groups showed variation,

characteristic elements differed between groups which experienced a greater recovery of

function and those which did not. For example KN+I+b tissues showed evidence of new

tissue formation which extended from the native tissue well into the site where muscle was

removed which was unlike the poorly recovered groups such as BAM, KN+M, KN+I, and

KN+M+b where new muscle tissue was visible only in a thin layer along the border of the

injury, and was small in area relative to the fat and scar tissue layers present. In addition,

KN+I+b, KN+M+I, and KN treated tissues showed a thinner, more moderate level of

88
adipose and scar tissue deposition in comparison with lower performing groups such as

KN+b, KN+M+b, KN+M, and KN+M+I+b which had extensive fat and/or scar tissue

formation. For KN+b tissues, the scar and fat tissue appeared to separate new tissue

formation into smaller rafts of tissue and to disrupt surrounding native tissue which may

explain why the KN+b group had a lower P0 and Pt than NR muscles (Table II).

In the literature, there are several examples of studies in which bFGF and IGF-1

enhanced skeletal muscle repair in vivo; however, these growth factors have also been

shown to lead to increased connective and scar tissue formation representative of the

observations in this study (37, 41, 43). IGF-1 acts on a variety of cell types, promoting

proliferation in fibroblasts, myoblasts, and chondrocytes and differentiation also in

myoblasts and in other cell types including adipocytes (27, 44). In one study, sustained

release of IGF-1 from microspheres in a rat model resulted in adipose tissue formation

believed to be a result of IGF-1 promoted differentiation of infiltrating cells into mature

adipocytes (45). Thus, the increased level of lipid containing cells observed in the IGF-1

groups is potentially a result of IGF-1 mediated differentiation of invading cells with an

immature phenotype. Likewise, bFGF is secreted by and promotes proliferation in a

variety of cell types including fibroblasts (27). As such, a possible mechanism behind the

increased fibrosis seen in bFGF groups is the increased proliferation of invading fibroblasts

resulting in a greater ECM deposition and scar tissue formation. Based on the results of the

individual growth factors it would be difficult to predict the favorable results seen in this

study, however similar success has been seen in other studies. For example, in an in vitro

study combining IGF-1 and bFGF in culture of muscle derived cells, cell proliferation was

greater than use of either of the growth factors alone (31) and when applied to an in vivo

89
muscle laceration injury, IGF-1 and bFGF resulted in significantly improved twitch force

(42).

The addition of cells for treatment of skeletal muscle injury has been shown to

mitigate the inflammatory process which studies have indicated to be a result of interaction

with invading macrophages (46). In the present study, the concentration of cells may not

have been sufficient to result in a favorable attenuation of inflammation: herein cellularized

groups did not promote a greater functional recovery than the acellular groups and in nearly

all samples examined did not appear to cause a decrease in connective and fibrotic tissue

generation. In previous studies wherein the addition of cells led to decreased inflammation

and promotion of functional recovery, a greater number of cells was introduced by the

therapy (1-6x106+ in cited studies versus 1.2-2.3x105 MPCs used in this study)(15-17, 40).

As such, for the number of cells introduced in this study, the histological and functional

evidence suggests that this volume of cells may have been capable of recruiting

macrophages and fibroblasts to promote an inflammatory response, but may not have been

sufficient in promoting an anti-inflammatory progression of the immune cell population

necessary for promotion of muscle regeneration.

This study sought to investigate keratin hydrogels in combination with cells and

growth factors implicated in regulation of skeletal muscle growth and development. In

doing so, a previously developed murine model of VML was utilized to test the

combination of keratin hydrogels loaded with combinations of MPCs, IGF-1, and bFGF.

Functional analysis indicated that the combination of keratin and both growth factors

resulted the greatest recovery which was notably not significantly greater than that seen in

the application of keratin alone or keratin combined with MPCs and IGF-1. Histological

90
findings indicated that improved recovery was associated with a greater presence of neo-

tissue formation and a decreased lipid and fibrotic tissue formation. Further, the functional

and histological data indicated that the cell concentration used in this study was likely not

sufficient to result in improved functional recovery or a decrease in fibrosis and connective

tissue deposition. Further efforts to increase cell concentration while maintaining the

desired physical properties of the keratin hydrogel would be an insightful follow up to this

study. The results of this study further showcased the utility of keratin hydrogels in tissue

engineering and served to illustrate its potential as a treatment for VML injuries.

91
Literature Cited

1. Grogan, B.F., Hsu, J.R., and Skeletal Trauma Res, C. Volumetric Muscle Loss.

Journal of the American Academy of Orthopaedic Surgeons 19, S35, 2011.

2. Covey, D.C. Blast and fragment injuries of the musculoskeletal system. The Journal of

bone and joint surgery American volume 84-A, 1221, 2002.

3. Thiele, O.C., Seeberger, R., Engel, M., Freier, K., and Hoffmann, J. Development of

the clinical use of distant flaps for head and neck reconstruction. Journal of Cranio-

Maxillofacial Surgery 42, 79, 2014.

4. Muramatsu, K., Ihara, K., Doi, K., Yoshida, K., Iwanaga, R., Hashimoto, T., and

Taguchi, T. Functional neuro-vascularized muscle transfer for oncological reconstruction

of extremity sarcoma. Surgical Oncology-Oxford 21, 263, 2012.

5. Fisher, D.M., and Sommerlad, B.C. Cleft Lip, Cleft Palate, and Velopharyngeal

Insufficiency. Plastic and Reconstructive Surgery 128, 342E, 2011.

6. Baugh, R.F., Basura, G.J., Ishii, L.E., Schwartz, S.R., Drumheller, C.M., Burkholder,

R., Deckard, N.A., Dawson, C., Driscoll, C., Gillespie, M.B., Gurgel, R.K., Halperin, J.,

Khalid, A.N., Kumar, K.A., Micco, A., Munsell, D., Rosenbaum, S., and Vaughan, W.

Clinical Practice Guideline: Bell's Palsy. Otolaryngology-Head and Neck Surgery 149,

S1, 2013.

7. Bianchi, B., Copelli, C., Ferrari, S., Ferri, A., and Sesenna, E. Successful salvage

surgery after treatment failures with cross graft and free muscle transplant in facial

reanimation. Journal of Cranio-Maxillofacial Surgery 40, 185, 2012.

8. Lawson, R., and Levin, L.S. Principles of free tissue transfer in orthopaedic practice.

Journal of the American Academy of Orthopaedic Surgeons 15, 290, 2007.

92
9. Can, A., Orgill, D.P., Ulrich, J.O.D., and Mureau, M.A.M. The myocutaneous

trapezius flap revisited: A treatment algorithm for optimal surgical outcomes based on 43

flap reconstructions. Journal of Plastic Reconstructive and Aesthetic Surgery 67, 1669,

2014.

10. Brown, B.N., and Badylak, S.F. Extracellular matrix as an inductive scaffold for

functional tissue reconstruction. Translational Research 163, 268, 2014.

11. Altman, G.H., Diaz, F., Jakuba, C., Calabro, T., Horan, R.L., Chen, J.S., Lu, H.,

Richmond, J., and Kaplan, D.L. Silk-based biomaterials. Biomaterials 24, 401, 2003.

12. Rouse, J.G., and Van Dyke, M.E. A Review of Keratin-Based Biomaterials for

Biomedical Applications. Materials 3, 999, 2010.

13. Vigodarzere, G.C., and Mantero, S. Skeletal muscle tissue engineering: strategies for

volumetric constructs. Frontiers in Physiology 5, 13, 2014.

14. Turner, N.J., and Badylak, S.F. Regeneration of skeletal muscle. Cell and Tissue

Research 3472012.

15. Machingal, M.A., Corona, B.T., Walters, T.J., Kesireddy, V., Koval, C.N.,

Dannahower, A., Zhao, W.X., Yoo, J.J., and Christ, G.J. A Tissue-Engineered Muscle

Repair Construct for Functional Restoration of an Irrecoverable Muscle Injury in a

Murine Model. Tissue Engineering Part A 17, 2291, 2011.

16. Corona, B.T., Ward, C.L., Baker, H.B., Walters, T.J., and Christ, G.J. Implantation

of In Vitro Tissue Engineered Muscle Repair Constructs and Bladder Acellular Matrices

Partially Restore In Vivo Skeletal Muscle Function in a Rat Model of Volumetric Muscle

Loss Injury. Tissue Engineering Part A 20, 705, 2014.

93
17. Corona, B.T., Machingal, M.A., Criswell, T., Vadhavkar, M., Dannahower, A.C.,

Bergman, C., Zhao, W.X., and Christ, G.J. Further Development of a Tissue Engineered

Muscle Repair Construct In Vitro for Enhanced Functional Recovery Following

Implantation In Vivo in a Murine Model of Volumetric Muscle Loss Injury. Tissue

Engineering Part A 18, 1213, 2012.

18. Tachibana, A., Furuta, Y., Takeshima, H., Tanabe, T., and Yamauchi, K. Fabrication

of wool keratin sponge scaffolds for long-term cell cultivation. Journal of Biotechnology

93, 165, 2002.

19. Verma, V., Verma, P., Ray, P., and Ray, A.R. Preparation of scaffolds from human

hair proteins for tissue-engineering applications. Biomedical Materials 3, 12, 2008.

20. Burnett, L.R., Rahmany, M.B., Richter, J.R., Aboushwareb, T.A., Eberli, D., Ward,

C.L., Orlando, G., Hantgan, R.R., and Van Dyke, M.E. Hemostatic properties and the

role of cell receptor recognition in human hair keratin protein hydrogels. Biomaterials 34,

2632, 2013.

21. Peyton, C.C., Keys, T., Tomblyn, S., Burmeister, D., Beumer, J.H., Holleran, J.L.,

Sirintrapun, J., Washburn, S., and Hodges, S.J. Halofuginone infused keratin hydrogel

attenuates adhesions in a rodent cecal abrasion model. Journal of Surgical Research 178,

545, 2012.

22. de Guzman, R.C., Saul, J.M., Ellenburg, M.D., Merrill, M.R., Coan, H.B., Smith,

T.L., and Van Dyke, M.E. Bone regeneration with BMP-2 delivered from keratose

scaffolds. Biomaterials 34, 1644, 2013.

94
23. Saul, J.M., Ellenburg, M.D., de Guzman, R.C., and Van Dyke, M. Keratin hydrogels

support the sustained release of bioactive ciprofloxacin. Journal of Biomedical Materials

Research Part A 98A, 544, 2011.

24. Fearing, B.V., and Van Dyke, M.E. In vitro response of macrophage polarization to a

keratin biomaterial. Acta Biomaterialia 10, 3136, 2014.

25. Hill, P., Brantley, H., and Van Dyke, M. Some properties of keratin biomaterials:

Kerateines. Biomaterials 31, 585, 2010.

26. de Guzman, R.C., Merrill, M.R., Richter, J.R., Hamzi, R.I., Greengauz-Roberts,

O.K., and Van Dyke, M.E. Mechanical and biological properties of keratose biomaterials.

Biomaterials 32, 8205, 2011.

27. Charge, S.B.P., and Rudnicki, M.A. Cellular and molecular regulation of muscle

regeneration. Physiological Reviews 84, 209, 2004.

28. Bischoff, R. Chemotaxis of skeletal muscle satellite cells. Developmental Dynamics

208, 505, 1997.

29. Florini, J.R., Ewton, D.Z., and Coolican, S.A. Growth hormone and the insulin-like

growth factor system in myogenesis. Endocrine Reviews 17, 481, 1996.

30. Hayashi, S., Aso, H., Watanabe, K., Nara, H., Rose, M.T., Ohwada, S., and

Yamaguchi, T. Sequence of IGF-I, IGF-II, and HGF expression in regenerating skeletal

muscle. Histochemistry and Cell Biology 122, 427, 2004.

31. Allen, R.E., and Boxhorn, L.K. REGULATION OF SKELETAL-MUSCLE

SATELLITE CELL-PROLIFERATION AND DIFFERENTIATION BY

TRANSFORMING GROWTH FACTOR-BETA, INSULIN-LIKE GROWTH FACTOR-

95
I, AND FIBROBLAST GROWTH-FACTOR. Journal of Cellular Physiology 138, 311,

1989.

32. Pace, L.A., Plate, J.F., Mannava, S., Barnwell, J.C., Koman, L.A., Li, Z., Smith,

T.L., and Van Dyke, M. A Human Hair Keratin Hydrogel Scaffold Enhances Median

Nerve Regeneration in Nonhuman Primates: An Electrophysiological and Histological

Study. Tissue Engineering Part A 20, 507, 2014.

33. Kowalczewski, C.J., Tombyln, S., Wasnick, D.C., Hughes, M.R., Ellenburg, M.D.,

Callahan, M.F., Smith, T.L., Van Dyke, M.E., Burnett, L.R., and Saul, J.M. Reduction of

ectopic bone growth in critically-sized rat mandible defects by delivery of rhBMP-2 from

kerateine biomaterials. Biomaterials 35, 3220, 2014.

34. Corona, B.T., Wu, X., and Walters, T.J. Treatment of volumetric muscle loss injury

with muscle-derived ECM and BMSCs promotes functional recovery in rat tibialis

anterior muscle. Journal of Tissue Engineering and Regenerative Medicine 6, 117, 2012.

35. Merritt, E.K., Cannon, M.V., Hammers, D.W., Le, L.N., Gokhale, R., Sarathy, A.,

Song, T.J., Tierney, M.T., Suggs, L.J., Walters, T.J., and Farrar, R.P. Repair of Traumatic

Skeletal Muscle Injury with Bone-Marrow-Derived Mesenchymal Stem Cells Seeded on

Extracellular Matrix. Tissue Engineering Part A 16, 2871, 2010.

36. Rossi, C.A., Flaibani, M., Blaauw, B., Pozzobon, M., Figallo, E., Reggiani, C.,

Vitiello, L., Elvassore, N., and De Coppi, P. In vivo tissue engineering of functional

skeletal muscle by freshly isolated satellite cells embedded in a photopolymerizable

hydrogel. Faseb Journal 252011.

37. Wang, L., Cao, L., Shansky, J., Wang, Z., Mooney, D., and Vandenburgh, H.

Minimally Invasive Approach to the Repair of Injured Skeletal Muscle With a Shape-

96
memory Scaffold. Molecular therapy : the journal of the American Society of Gene

Therapy 22, 1441, 2014.

38. Page, R.L., Malcuit, C., Vilner, L., Vojtic, I., Shaw, S., Hedblom, E., Hu, J., Pins,

G.D., Rolle, M.W., and Dominko, T. Restoration of Skeletal Muscle Defects with Adult

Human Cells Delivered on Fibrin Microthreads. Tissue Engineering Part A 172011.

39. Beier, J.P., Stern-Straeter, J., Foerster, V.T., Kneser, U., Stark, G.B., and Bach, A.D.

Tissue engineering of injectable muscle: Three-dimensional myoblast-fibrin injection in

the syngeneic rat animal model. Plastic and Reconstructive Surgery 118, 1113, 2006.

40. Irintchev, A., Langer, M., Zweyer, M., Theisen, R., and Wernig, A. Functional

improvement of damaged adult mouse muscle by implantation of primary myoblasts.

Journal of Physiology-London 500, 775, 1997.

41. Sato, K., Li, Y., Foster, W., Fukushima, K., Badlani, N., Adachi, N., Usas, A., Fu,

F.H., and Huard, J. Improvement of muscle healing through enhancement of muscle

regeneration and prevention of fibrosis. Muscle & Nerve 28, 365, 2003.

42. Menetrey, J., Kasemkijwattana, C., Day, C.S., Bosch, P., Vogt, M., Fu, F.H.,

Moreland, M.S., and Huard, J. Growth factors improve muscle healing in vivo. Journal of

Bone and Joint Surgery-British Volume 82B2000.

43. Borselli, C., Storrie, H., Benesch-Lee, F., Shvartsman, D., Cezar, C., Lichtman, J.W.,

Vandenburgh, H.H., and Mooney, D.J. Functional muscle regeneration with combined

delivery of angiogenesis and myogenesis factors. Proceedings of the National Academy

of Sciences of the United States of America 107, 3287, 2010.

97
44. Jones, J.I., and Clemmons, D.R. INSULIN-LIKE GROWTH-FACTORS AND

THEIR BINDING-PROTEINS - BIOLOGICAL ACTIONS. Endocrine Reviews 16, 3,

1995.

45. Yuksel, E., Weinfeld, A.B., Cleek, R., Waugh, J.M., Jensen, J., Boutros, S., Shenaq,

S.M., and Spira, M. De novo adipose tissue generation through long-term, local delivery

of insulin and insulin-like growth factor-1 by PLGA/PEG microspheres in an in vivo rat

model: A novel concept and capability. Plastic and Reconstructive Surgery 105, 1721,

2000.

46. Arnold, L., Henry, A., Poron, F., Baba-Amer, Y., van Rooijen, N., Plonquet, A.,

Gherardi, R.K., and Chazaud, B. Inflammatory monocytes recruited after skeletal muscle

injury switch into antiinflammatory macrophages to support myogenesis. Journal of

Experimental Medicine 204, 1057, 2007.

98
CHAPTER III

Keratin Hydrogel Enhances In Vivo Skeletal Muscle Function in a Rat Model of

Volumetric Muscle Loss

Juliana A. Passipieria,b; Hannah B. Bakera,c; Mevan Siriwardanea; Mary D. Ellenburgd;

Justin M. Saule; Luke Burnettd; Seth Tomblynd; George J. Christa,b,c,f

Affiliations:
a
Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-

Salem, NC 27157, USA


b
Biomedical Engineering Department, University of Virginia, Charlottesville, VA, 22908,

USA
c
VT-WFU School of Biomedical Engineering and Sciences, Winston-Salem, NC 27157;
d
KeraNetics, LLC, Suite 168, 391 Technology Way, Winston-Salem, NC 27101;
e
Department of Chemical and Paper Engineering, Miami University, Oxford, OH 45056,

USA;
f
Orthopedics Department, University of Virginia, Charlottesville, VA, 22908, USA

Note: This manuscript was prepared by Dr. Juliana A. Passipieri and has not yet been

submitted. She (primarily) and Hannah B. Baker (secondarily) were responsible for the

majority of the work described.

99
Introduction

The natural progression of muscle injury involves a series of interdependent and

overlapping phases that include muscle degradation/inflammation, regeneration, and

remodeling (1). The muscle repair process is detailed reviewed in (2).

However, when the extent of the injury goes beyond the intrinsic regenerative

mechanism, the irrecoverable loss of tissue is referred to as volumetric muscle loss (VML)

(3). It can be the result of numerous congenital and/or acquired diseases, as well as

traumatic accidents, as those experienced by the military population during battlefield

injuries.

Currently, the survival rate of battlefield injuries is the highest in history (4) due to

significant improvements in medical training, tactical warfare, and advances in personal

armor. Consequently, there are an increasing number of wounded warriors with serious but

survivable injuries facing permanent disability and disfigurement. It is paramount that

every one of them has maximal potential for functional recovery.

Present therapeutic options are very limited and highly associated to local morbidity

and mortality (5). As a result, focus has turned to tissue-engineering technologies to

develop more effective treatments for large muscle injuries. A popular approach is the

implantation of growth factor-releasing biological scaffolds, which can alter the

macrophage phenotype response from scar formation to nascent muscle remodeling (6).

Acellular extracellular matrix (ECM) is an effective biological scaffold used to optimize

muscle repair, specifically when it is combined with a cellular component (e.g. muscle

progenitor cells – MPC) (7-10). However, every injury is unique and it is necessary to

explore a combination of treatments to efficiently and effectively restore muscle function.

100
Keratin-based biomaterials are at the forefront of tissue engineering applications

due to their tissue compatibility, degradability (11), non-antigenic, and non-immunogenic

properties (12). Medical and biotechnology applications of these biomaterials have been

presented in several studies showing their in vitro compatibility with different cell types

(13-15), and their contributions to bone, cardiac muscle and peripheral nerve regeneration

(16-20) as well as wound healing in vivo (21, 22).

Keratin, an intermediate filament protein that constitutes hair, wool, nails and other

epidermal appendageal structures (23), is processed via oxidation, creating a non-

covalently cross-link form known as keratose (KOS) or reduction, creating a disulfide

cross-linked form known as kerateine (KTN). As the mammalian metabolism is incapable

of synthetizing the enzyme responsible for keratin breakdown, the degradation rate of

keratin based-biomaterials is easily manipulated through the combination of different ratios

of the cross-linked and non cross-linked forms.

The easy manipulation of the degradation rate enables the controlled release of

physiologically relevant molecules (i.e. growth factor, antibiotics), potentially increasing

the clinical relevance of keratin-based biomaterials. Growth factors are essential in the

complex network of regulatory pathways responsible for triggering and maintaining

through cell proliferation, differentiation and growth, being intrinsically released from the

extracellular matrix surrounding injured tissue (24, 25) and by inflammatory cells during

the initial phase of tissue healing (26, 27).

Basic fibroblast growth factor (bFGF or FGF-2) is known to directly stimulate

muscle regeneration by activating satellite cell proliferation, enhancing satellite cell

recruitment to the injury site, and, due to its strong angiogenic effect (28, 29), by improving

101
overall blood perfusion at the injury site. Likewise, Insulin-like Growth Factor 1 (IGF-1)

also plays an important role in muscle maintenance and repair. IGF-1 influences muscle

metabolism, via muscle cell differentiation and proliferation (30) and it stimulates satellite

cell proliferation in rat skeletal muscle (31).

In this context, to investigate a novel skeletal muscle tissue engineering strategy,

we hypothesized whether keratin hydrogel would support muscle regeneration as well as

provide a suitable delivery platform for growth factors and ex-vivo grown cells. To address

this issue, we used a rat Tibialis Anterior (TA) VML injury model to evaluate the effects

of keratin hydrogel formulations with IGF-1 and/or bFGF and/or MPC on muscle

regeneration.

Methods

Keratin hydrogel preparation

Keratins were extracted using a proprietary patent-pending process in a Quality

Systems Regulated facility at KeraNetics, LLC in a manner similar to those previously

published (32, 33). The lyophilized powders were weighed to produce a 15% keratose

(KOS) and 7% kerateine (KTN) weight to volume hydrogel. The KOS was hydrated with

either DPBS, if the gel did not contain cells, or was hydrated with cell culture media

(without fetal bovine serum) if the gel contained MPCs.

The KTN gel was hydrated with water, when growth factors were impregnated into

the gels. Individual gels are given approximately 30 minutes to spontaneously form

hydrogels at 37°C. The gels are then mixed together placing 70% KOS gel and 30% KTN

102
gel into one mixture. The concentration of growth factor used was 100µg/ml and the cell

concentration was 1.15 x 106 MPC/ml in the final product.

Treatment groups

Treatment groups and group sizes are listed in Table I and are as follows: Keratin

(n=18), Keratin + IGF (n=18), Keratin + bFGF (n=18), Keratin + IGF + bFGF (n=18),

Keratin + MPC (n=8), Keratin + MPC + IGF (n=8), Keratin + MPC + bFGF (n=8), Keratin

+ MPC + IGF + bFGF (n=8). Two negative control groups were included: no repair group

(NR, n=17), and BAM group (n=17) (Bladder Acellular Matrix, n=17), performed as

previously described (8).

Table I: Design of experimental groups submitted to VML injury model.

MPC
Group IGF-1 bFGF
Group Keratin 1.15x106 BAM
size (n) 100µg/mL 100µg/mL
cells/mL
No repair 17 - - - - -
BAM 17 - - - - +
Keratin 18 + - - - -
KN + I 18 + + - - -
KN+ b 18 + - + - -
KN + I + b 18 + + + - -
KN + M 8 + - - + -
KN + M + I 8 + + - + -
KN + M + b 8 + - + + -
KN + M + I + b 8 + + + + -

KN: keratin; I: IGF-1, insulin-like growth factor 1;b: bFGF, basic fibroblast growth
factor; MPC: muscle progenitor cell; BAM: bladder acellular matrix.

103
Muscle Progenitor Cell (MPC) isolation and culture

MPCs were isolated from the soleus and TA muscles of 4-6 weeks old male Lewis

rats (Charles River Laboratories) as previously described (8-10). Briefly, after sterilization

in iodine and consecutive washes in sterile PBS, muscles were minced into small pieces by

hand, and incubated in 0.2% collagenase (Worthington Biochemicals) solution prepared in

low glucose Dulbecco’s modified Eagle’s medium (DMEM; Hyclone) for 2 hours at 37

°C. Muscle tissue fragments were plated onto tissue culture dishes coated with Matrigel

(1:50 dilution, BD Biosciences) in myogenic medium containing DMEM high glucose

supplemented with 20% fetal bovine serum (FBS), 10% horse serum, 1% chicken embryo

extract, and 1% antibiotic/antimycotic (Hyclone). Cells were passaged at 70%–80%

confluence, cultured in low-glucose DMEM supplemented with 15% FBS and 1%

antibiotic/antimycotic, and impregnated in the keratin gel at second passage.

Animal care

This study was conducted in compliance with the Animal Welfare Act, the

Implementing Animal Welfare Regulations, and in accordance with the principles of the

Guide for the Care and Use of Laboratory Animals. The Wake Forest University Health

Sciences School of Medicine Animal Care and Use Committee approved all animal

procedures. Adult female Lewis rats (Charles River Laboratories) weighting 203.4 + 0.8 g,

at 11-13 weeks of age, were individually housed in a vivarium accredited by the American

Association for the Accreditation of Laboratory Animal Care, and provided with food and

water ad libitum.

104
Surgical Procedures

Surgical creation of volumetric muscle loss (VML) injury was performed in the TA

muscle as previously reported (8, 34). A longitudinal incision was made on the lateral

aspect of the left lower leg, followed by blunt separation of the skin from the underlying

fascia. Similarly, the fascia covering the anterior crural muscles was separated using blunt

dissection. The proximal and distal tendons of the Extensor Hallicus Longus (EHL) and

Extensor Digitorum Longus (EDL) muscles were then isolated and ablated. TA muscle

corresponds to 0.17% of the total body weight, as previously determined (8, 34). VML

injury was created by excising approximately 20% of the TA muscle weight at the middle

third of the muscle (Figure 1A).

105
Figure 1. Creation of VML in Rat TA and Injection of Keratin Hydrogel. Rat

hind limb is shown with anterior side facing forward. Anterior crural compartment

muscles are exposed by opening the skin and fascia and the EDL and EHL are

ablated tendon to tendon (not shown). The longitudinal middle third, ~1cm, is

notched 0.5cm across the width of the TA and ~0.25cm depth of muscle is removed

from the rectangular region (A). The surrounding fascia is closed, leaving one gap

for a needle to be inserted and ~200µl of hydrogel is injected into the fascia-muscle

pocket (B). The remaining fascial gap is closed, the skin is closed and the animal

is allowed to recover.

106
The BAM scaffold was sutured directly over the defect as previously described (8).

The fascia was closed with 6-0 vicryl sutures, and approximately 200µl of the hydrogel

were injected using a 19G needle over the muscle defect through the sutured fascia (Figure

1B).

The skin closure was performed with 5-0 prolene using interrupted sutures, and

skin glue was applied between the skin sutures to help prevent the incision from opening.

Ketoprofen (0.03 mg/kg; subcutaneously) was administered every twenty-four hours for

three days.

In vivo Functional Analysis

At 4, 8 and 12 weeks after surgery, rats were anesthetized (1.5 – 2.5% isoflurane)

and the left hind limb was aseptically prepared. The rat was placed supine on a heated

platform and the left knee was bent to a 90-degree angle and was secured using a stabilizing

rod. The left foot was taped to a footplate attached to the shaft of an Aurora Scientific

305C-LR-FP servomotor, which was controlled using a computer. Sterilized percutaneous

needle electrodes were inserted through the skin for stimulation of the left common

peroneal nerve. Electrical stimulus was applied using an Aurora Scientific stimulator with

a constant current SIU (Model 701C). Stimulation voltage and needle electrode placement

were optimized with a series of twitch contractions at 1 Hz. Contractile function of the

anterior crural muscles was assessed by measuring peak isometric tetanic torque derived

from the maximal response to a range of stimulation frequencies (10 - 200 Hz). After

functional testing, animals were allowed to recover on the heated platform and returned to

107
the vivarium. For terminal time points, animals were euthanized via CO2 inhalation and

the TA muscle harvested.

Gross morphology analysis of TA muscle

After animal euthanasia, skin on the hind limbs was removed and the fascia layer

was gently detached and removed. Once exposed, injured TA muscle and its contralateral

uninjured control were imaged, and then harvested. The weights of the samples were

measured, followed by preparation for histological analysis.

Histology and immunohistochemistry

TA muscles from all experimental groups were fixed in 4% paraformaldehyde

overnight at 4 °C. Five-micrometer-thick serial sections were cut from the paraffin

embedded blocks and submitted to Masson’s trichrome staining and Gill’s Hematoxylin-

Eosin staining following standard procedures. Histological analysis was performed in

longitudinal sections of the muscle at approximately 50 to 150 microns deep into the wound

bed. Images were captured and digitized (DM4000B Leica Upright Microscope; Leica

Microsystems) at varying magnifications.

Statistics

Numeric data are presented as Mean + SEM. Functional data were analyzed using

one-way or two-way ANOVA and Fisher’s PLSD post-hoc test using Prism (GraphPad

Software Inc, La Jolla, CA). Statistical significance was achieved at an alpha < 0.05.

108
Results

Creation of volumetric muscle loss (VML) injury in the rat tibialis anterior

None of the animals died during the surgical procedure and no post-implantation mortality

was recorded. The weight of the muscle excised from the animals in each treatment group

was similar, reflecting the creation of comparable VML injuries (NR: 73.3 ± 0.6 mg; BAM:

72.9 ± 0.7 mg; KN: 73.9 ± 0.6 mg; KN+I: 72.7 ± 1.1 mg; KN+b: 72.4 ± 0.9 mg; KN+I+b:

70.4 ± 1.0 mg; KN+M: 67.8 ± 0.5 mg; KN+M+I: 73.9 ± 1.4 mg; KN+M+b: 68.1 ± 0.83

mg; KN+M+I+b: 72.4 ± 0.98 mg).

In vivo Functional analysis after VML injury

Animal weights were similar among the groups at the time of baseline functional

analysis, surgery, and functional analysis at 4, 8 and 12 weeks post- surgery (Figure 2 A).

Additionally, the similarity in body weight reflects the lack of adverse reaction to the

various treatments. All data collected were normalized to body weight, in order to control

the effect of normal growth on peak isometric torque production over the course of the

study.

Mean values for the baseline (pre-injury) maximal isometric torque in response to

peroneal nerve stimulation are shown in Figure 2B for all animals and treatment groups.

The results demonstrate that the initial strength prior to VML injury is remarkably

consistent, and statistically indistinguishable, among animals in all treatment groups.

109
110
Figure 2. Contribution of Keratin Hydrogel Formulation to Muscle

Regeneration at 4 and 8 Weeks. A) Body weights of study animals reveal normal

healthy weight gain in all groups. B) Baseline contraction force resulting from

peroneal nerve stimulation and measured with a footplate force transducer. Peak

isometric torque measured at C) 4 weeks. D) 8 weeks. No significant advantage

resulted from the inclusion of cells in the injected hydrogel treatment Individual

responses are normalized to the respective group mean of the initial maximum pre-

injury isometric torque response and their body weight. Data are presented as Mean

+ SEM. *-Significantly different at the p<0.05 level using Fisher’s PLSD test; after

performing a Two-Way ANOVA for matched samples. Group sample sizes are

listed in parentheses.

111
For the first phase of the study, seven to eight animals were randomized into 10

different treatments. The mean isometric torque values normalized to pre-injury baseline

force and body weight at 4 and 8 weeks after surgery are shown in Figure 2C and Figure

2D, respectively. A two-way ANOVA was used to compare functional outcomes among

the different treatment groups over time. Statistical analysis revealed that most treatment

groups showed improvement relative to the negative control BAM group, but overall, no

significant advantage resulted from the inclusion of cells in the injected hydrogel treatment.

Moreover, as illustrated, at 4 weeks (Figure 2C) and 8 weeks post-VML injury (Figure 2D

and Table II), the groups treated with only keratin or keratin containing growth factor(s)

actually showed significant improvement over the groups that had cells embedded within

the keratin hydrogel.

112
Table II. In Vivo Functional Analysis of TA Muscle at 8 Weeks Post-Injury. Values are mean ± SEM. Values denoted with

the same letter ate not significantly different (p>0.05), while values without a like letter denotation are significantly different

113
(p<0.05). # Deficit calculated from unoperated animals. BAM: bladder acellular matrix; b: basic fibroblast growth factor;

I: insulin like growth factor type 1; M: muscle progenitor cells; K: keratin; TA: tibialis anterior; wt: weight.
Animals were euthanized at 8 weeks after surgery and an independent second phase

of the study was conducted. Nine to ten animals were randomly assigned to one of 6

treatment groups (i.e., the 4 groups containing cells were eliminated), and the studies were

extended to a 12-week time point to ensure that the extent of functional recovery was more

completely evaluated for these groups (as indicated in (8)).

Placement/implantation of keratin hydrogels (with the exception of KN+I) at the

site of VML injury in the TA was associated with significant and sustained increases in

functional recovery, relative to No Repair (NR) and/or implantation of BAM alone, at all

time points post-VML injury (Figure 3). Moreover, at 12 weeks post-VML injury, the peak

isometric torque response to peroneal nerve stimulation in all keratin treatment groups

(again, except for KN+I) was significantly greater than that observed for both the NR and

BAM treatment groups (Figure 3C and Table III).

Analysis of TA muscle wet weight showed significant reduction of muscle mass in

all experimental groups relative to their contralateral control. However, after 12 weeks, the

TA wet weight of the samples in the KN+b and KN+I+b groups were improved compared

to the other experimental groups (Figure 3D).

114
115
Figure 3. Keratin Hydrogel Promotes Continuous Functional Recovery over

Time. Peak isometric torque production for the treatment groups that combined

formulations of Keratin and growth factors at 4 weeks (A), 8 weeks (B) and 12

weeks (C) are presented. Implantation of all keratin-based treatment groups, except

keratin + IGF, resulted in significant and sustained functional recovery relative to

BAM and/or NR. Wet weight measurements of the injured TA muscle and their

contralateral control are shown in (D). Individual responses are normalized to the

respective group mean of the initial maximum pre-injury isometric torque response

and their body weight. Data is presented as Mean + SEM. *-Significantly different

at the p<0.05 level using Fisher’s PLSD test; after performing a Two-Way ANOVA

for matched samples. Group sample sizes are listed in parentheses.

116
Table III. In Vivo Functional Analysis of TA Muscle at 12 Weeks Post-Injury. Values are mean ± SEM. Values denoted

117
with the same letter ate not significantly different (p>0.05), while values without a like letter denotation are significantly different

(p<0.05). # Deficit calculated from unoperated animals. BAM: bladder acellular matrix; b: basic fibroblast growth factor; I:

insulin like growth factor type 1; K: keratin; TA: tibialis anterior; wt: weight.
Gross Morphology and histological Analysis of TA muscle

Macroscopically, the implants were well tolerated by the recipient animals, with no

episodes infection or seroma. Significant gross morphological modifications were evident

at 12 weeks after the surgical procedure (Figure 4 i-vi). The wound bed was nearly

indistinguishable in the KN (Figure 4 iv), KN+b (Figure 4 v), and KN+I+b (Figure 4 vi)

groups, where signs of regeneration were evident by the presence of viable tissue at the

injury site close to the native muscle, reduced fibrosis and inflammatory infiltrate (Figure

4 I-X).

Corroborating the functional data, the KN+I group showed signs of atrophy at the

injury site (Figure 4 iii), especially when compared to the other keratin treated groups.

Nevertheless, new tissue formation was still observed at the injury site (Figure 4 M-P).

The BAM implant was well integrated with the surrounding wound bed (Figure 4

ii) and filled the void created by the surgical excision of the muscle. However,

histologically, the lack of newly formed tissue as well as increased collagen matrix

formation and inflammatory cell infiltration were evident and corroborated with

diminished functional recovery (Figure 4 E-H).

Different from every group, the wound bed present in the NR group created was

very prominent (Figure 4 i) and the lack of muscle or fibrosis formation at the initial

150mm inside the wound bed confirmed the insufficient regeneration (Figure 4 A-D).

118
119
Figure 4. Gross Morphology and Histological Analysis of TA after Keratin

Based Hydrogel Treatment. Distinguishable differences in the gross appearance

of each group were evident 12 weeks after VML injury. Injury site was apparent in

the NR (i), and, in less extent, in the KN + I group (iv). New tissue formation

completed most of the wound bed in the KN (iii), KN + b (v), and KN + I + b (vi).

BAM implant was incorporated into the wound bed in the BAM group (ii).

Hematoxylin- Eosin (red/pink = cell cytoplasm; violet = nuclei) and Mason’s

Thrichrome (red = tissue; blue = collagen; black = nuclei) staining of longitudinal

section from NR (A-D), BAM (E-H), KN (I-L), KN + I (M-P), KN + b (Q-T) and

KN + I + b (U-X) groups. Arrows denote regions with new muscle fiber formation.

Stars denote regions with native muscle.

120
Discussion

Skeletal muscle has a robust capacity for regeneration after injury, based on the

proliferation and differentiation of resident quiescent satellite cells (35). However, there

are no commercially available products to treat irreversible muscle injuries that result from

traumatic accidents, or acquired and/or congenital diseases.

Our group recently showed the efficacy of TEMR (tissue engineered muscle repair)

construct implantation to restore function in a VML injury model in the TA muscle. The

TEMR constructs were created by seeding muscle progenitor cells (MPCs) onto a bladder

acellular matrix (BAM) and subjecting it to cyclic mechanical preconditioning (10%

stretch) in a bioreactor prior to implantation. In fact, the presence of MPCs in the construct

played a crucial role in the regeneration process as shown by the greater functional recovery

on the TEMR group compared to the group implanted with unseeded BAM constructs (8).

Despite the encouraging outcome from the TEMR technology studies, a

combination of different tissue engineered approaches are necessary to treat the ample

variety of traumatic injuries and diseases that compromise soft tissue.

Keratin hydrogel is a malleable biological scaffold, with future clinical applications

promoting minimally invasive surgical procedures. Additionally, its material properties

(e.g. degradation rate) can be easily tailored to promote sustainable release of biological

compounds (20, 33) that closely influence tissue regeneration (e.g. growth factors).

IGF-1 and bFGF are locally produced molecules that play crucial roles in the

complex signaling cascade that stimulates recruitment, proliferation and differentiation of

satellite cells (36, 37). The incorporation of these growth factors into keratin hydrogel

allows for their controlled release over time, avoiding loss of biological activity due to

121
diffusion and/or enzymatic inactivation (38).

Herein, keratin hydrogels were a 70% KOS/30% KTN combination, as used in

previous studies (32, 33), and contained a combination of IGF-1 and/or bFGF (Table 1).

Functional recovery of the TA muscle after VML injury for all treatment groups is

represented in Figure 3. The maximal isometric torque in the NR and BAM groups

corroborates with data previously published (8, 34), indicating precision in the

reproduction of the TA injury model. Hydrogel implantation mediated restoration of

significant functional capacity that continued to improve over the course of 12 weeks, when

the functional recovery observed by keratin alone was indistinguishable from other

effective keratin hydrogels containing growth factors (Keratin + bFGF and Keratin + IGF

+ bFGF).

These observations highlight the efficacy of the keratin biomaterial as a potentially

important platform for treatment of VML injury. To put these observations into

perspective, it should be noted that the synergists (EDL and EHL) are removed at the time

of the surgical VML injury, resulting in a permanent ≈20% functional deficit (see (8) for

details). This means that after removing approximately 20% of the TA muscle, the total

functional defect is ≈50%, of which only 30% is recoverable. Thus, the maximal functional

recovery possible is 0.8 of the pre-injury baseline maximal isometric torque response. Both

NR and BAM treatment groups exhibited maximal torque responses of ≤0.6, while the

keratin hydrogel treatments (Keratin alone, Keratin + bFGF and Keratin+ bFGF + IGF)

exhibited values of ≥0.7, ranging as high as 0.74. This corresponds to ≈90% functional

recovery. These are robust and reproducible functional recoveries on a substantial number

of animals.

122
Qualitative morphological and histological analysis corroborates with functional

data. The TA muscle treated with keratin-based hydrogels had distinguishable morphology

from the untreated group (NR) (Figure 4), and evidence of new muscle fiber formation and

diminished fibrosis related to the BAM treated group (Figure 5).

Interestingly, keratin-only implantation promoted morphological and functional

improvement even in the absence of growth factor (IGF-1 and bFGF). It is possible that

during muscle regeneration, keratin biomaterial modulates cell proliferation, cell adhesion,

and gene expression in a similar manner as seen in nerve repair (16, 32, 39, 40). However,

the data from this study does not provide sufficient basis to formulate an explanation for

the molecular mechanics behind the functional and morphological improvement in the

experimental groups. Further investigation regarding the influence of the keratin-based

hydrogel over skeletal muscle regeneration is warranted.

The contribution of controlled time-released growth factors to muscle healing have

been extensively discussed (reviewed in (41)). However, throughout the literature, there is

clear evidence that the addition of cells component to tissue-engineered scaffolds is also

crucial to enhance functional recovery in a VML injury model (8-10, 42-44). Synthetic and

biological hydrogels have been used as a cell delivery system in a variety of injury models

in vivo (45-55). Through hydrogel implantation, myoblasts formed new muscle fibers in a

skeletal muscle injury in one study (45), while bone marrow mesenchymal stem cells were

shown to improve cardiac remodeling after myocardial infarction in another (54). This

study has shown that addition of MPCs (at the concentration used herein) used to the

keratin formulation did not significantly improve muscle regeneration.

123
Based on our experience with the keratin hydrogel, the number of MPC implanted

was consistent with the hydrogel’s limitations regarding viscosity. The number of MPCs

is remarkably smaller compared to the number of cells used in the above-mentioned

studies. Our previous work has revealed that implantation of too few myoblasts in this

VML injury setting may actually be associated with diminished functional regeneration

(8). The data are consistent with the supposition that before hydrogels can be used in

muscle repair, further optimization of keratin hydrogel properties are required to

accommodate the delivery of a higher cell yield.

Conclusion

We have shown for the first time that keratin hydrogel, successfully used for bone

and nerve regeneration, promotes significant functional improvement of severely damaged

skeletal muscle. The recovery observed after keratin implantation is achieved through a

minimally invasive transplantation procedure. This procedure has apparent equivalence to

the efficacy of materials used for cell delivery in the same animal model, but with the

advantage of not requiring a cellular component, a xenogeneic scaffold (BAM), or

bioreactor preconditioning.

124
Literature Cited

1. Li, Y., Foster, W., Deasy, B.M., Chan, Y., Prisk, V., Tang, Y., Cummins, J., and

Huard, J. Transforming growth factor-beta1 induces the differentiation of myogenic cells

into fibrotic cells in injured skeletal muscle: a key event in muscle fibrogenesis. The

American journal of pathology 164, 1007, 2004.

2. Jarvinen, T.A., Jarvinen, T.L., Kaariainen, M., Kalimo, H., and Jarvinen, M. Muscle

injuries: biology and treatment. The American journal of sports medicine 33, 745, 2005.

3. Grogan, B.F., Hsu, J.R., and Skeletal Trauma Research, C. Volumetric muscle loss.

The Journal of the American Academy of Orthopaedic Surgeons 19 Suppl 1, S35, 2011.

4. Holcomb, J.B., Stansbury, L.G., Champion, H.R., Wade, C., and Bellamy, R.F.

Understanding combat casualty care statistics. The Journal of trauma 60, 397, 2006.

5. Lin, C.H., Lin, Y.T., Yeh, J.T., and Chen, C.T. Free functioning muscle transfer for

lower extremity posttraumatic composite structure and functional defect. Plastic and

reconstructive surgery 119, 2118, 2007.

6. Badylak, S.F., Brown, B.N., Gilbert, T.W., Daly, K.A., Huber, A., and Turner, N.J.

Biologic scaffolds for constructive tissue remodeling. Biomaterials 32, 316, 2011.

7. Corona, B.T., Wu, X., Ward, C.L., McDaniel, J.S., Rathbone, C.R., and Walters, T.J.

The promotion of a functional fibrosis in skeletal muscle with volumetric muscle loss

injury following the transplantation of muscle-ECM. Biomaterials 34, 3324, 2013.

8. Corona, B.T., Ward, C.L., Baker, H.B., Walters, T.J., and Christ, G.J. Implantation of

in vitro tissue engineered muscle repair constructs and bladder acellular matrices partially

restore in vivo skeletal muscle function in a rat model of volumetric muscle loss injury.

Tissue engineering Part A 20, 705, 2014.

125
9. Machingal, M.A., Corona, B.T., Walters, T.J., Kesireddy, V., Koval, C.N.,

Dannahower, A., Zhao, W., Yoo, J.J., and Christ, G.J. A tissue-engineered muscle repair

construct for functional restoration of an irrecoverable muscle injury in a murine model.

Tissue engineering Part A 17, 2291, 2011.

10. Corona, B.T., Machingal, M.A., Criswell, T., Vadhavkar, M., Dannahower, A.C.,

Bergman, C., Zhao, W., and Christ, G.J. Further development of a tissue engineered

muscle repair construct in vitro for enhanced functional recovery following implantation

in vivo in a murine model of volumetric muscle loss injury. Tissue engineering Part A 18,

1213, 2012.

11. de Guzman, R.C., Merrill, M.R., Richter, J.R., Hamzi, R.I., Greengauz-Roberts,

O.K., and Van Dyke, M.E. Mechanical and biological properties of keratose biomaterials.

Biomaterials 32, 8205, 2011.

12. Kirfel, J., Magin, T.M., and Reichelt, J. Keratins: a structural scaffold with emerging

functions. Cellular and molecular life sciences : CMLS 60, 56, 2003.

13. Tachibana, A., Kaneko, S., Tanabe, T., and Yamauchi, K. Rapid fabrication of

keratin-hydroxyapatite hybrid sponges toward osteoblast cultivation and differentiation.

Biomaterials 26, 297, 2005.

14. Yamauchi, K., Maniwa, M., and Mori, T. Cultivation of fibroblast cells on keratin-

coated substrata. Journal of biomaterials science Polymer edition 9, 259, 1998.

15. Reichl, S. Films based on human hair keratin as substrates for cell culture and tissue

engineering. Biomaterials 30, 6854, 2009.

126
16. Hill, P.S., Apel, P.J., Barnwell, J., Smith, T., Koman, L.A., Atala, A., and Van Dyke,

M. Repair of peripheral nerve defects in rabbits using keratin hydrogel scaffolds. Tissue

engineering Part A 17, 1499, 2011.

17. Dias, G.J., Peplow, P.V., McLaughlin, A., Teixeira, F., and Kelly, R.J.

Biocompatibility and osseointegration of reconstituted keratin in an ovine model. Journal

of biomedical materials research Part A 92, 513, 2010.

18. Shen, D., Wang, X., Zhang, L., Zhao, X., Li, J., Cheng, K., and Zhang, J. The

amelioration of cardiac dysfunction after myocardial infarction by the injection of keratin

biomaterials derived from human hair. Biomaterials 32, 9290, 2011.

19. Pace, L.A., Plate, J.F., Smith, T.L., and Van Dyke, M.E. The effect of human hair

keratin hydrogel on early cellular response to sciatic nerve injury in a rat model.

Biomaterials 34, 5907, 2013.

20. de Guzman, R.C., Saul, J.M., Ellenburg, M.D., Merrill, M.R., Coan, H.B., Smith,

T.L., and Van Dyke, M.E. Bone regeneration with BMP-2 delivered from keratose

scaffolds. Biomaterials 34, 1644, 2013.

21. Pechter, P.M., Gil, J., Valdes, J., Tomic-Canic, M., Pastar, I., Stojadinovic, O.,

Kirsner, R.S., and Davis, S.C. Keratin dressings speed epithelialization of deep partial-

thickness wounds. Wound repair and regeneration : official publication of the Wound

Healing Society [and] the European Tissue Repair Society 20, 236, 2012.

22. Poranki, D., Whitener, W., Howse, S., Mesen, T., Howse, E., Burnell, J., Greengauz-

Roberts, O., Molnar, J., and Van Dyke, M. Evaluation of skin regeneration after burns in

vivo and rescue of cells after thermal stress in vitro following treatment with a keratin

biomaterial. Journal of biomaterials applications 29, 26, 2013.

127
23. Crewther WG, F.R., Lennox FG, Lindley H. The chemistry of keratins. In: Anfinsen

CB A.M., Edsall JT, Richards FM, ed. Advances in protein

chemistry. New York: Academic Press; 1965. pp. 191.

24. Clarke, M.S., Khakee, R., and McNeil, P.L. Loss of cytoplasmic basic fibroblast

growth factor from physiologically wounded myofibers of normal and dystrophic muscle.

Journal of cell science 106 ( Pt 1), 121, 1993.

25. DiMario, J., Buffinger, N., Yamada, S., and Strohman, R.C. Fibroblast growth factor

in the extracellular matrix of dystrophic (mdx) mouse muscle. Science 244, 688, 1989.

26. Collins, C.A., and Partridge, T.A. Self-renewal of the adult skeletal muscle satellite

cell. Cell cycle 4, 1338, 2005.

27. Hill, M., Wernig, A., and Goldspink, G. Muscle satellite (stem) cell activation during

local tissue injury and repair. Journal of anatomy 203, 89, 2003.

28. Lafreniere, J.F., Mills, P., Tremblay, J.P., and El Fahime, E. Growth factors improve

the in vivo migration of human skeletal myoblasts by modulating their endogenous

proteolytic activity. Transplantation 77, 1741, 2004.

29. Efthimiadou, A., Asimakopoulos, B., Nikolettos, N., Giatromanolaki, A., Sivridis,

E., Lialiaris, T.S., Papachristou, D.N., and Kontoleon, E. The angiogenetic effect of

intramuscular administration of b-FGF and a-FGF on cardiac muscle: the influence of

exercise on muscle angiogenesis. Journal of sports sciences 24, 849, 2006.

30. Monier, S., and Le Marchand-Brustel, Y. Effects of insulin and IGF-I on RNA

synthesis in isolated soleus muscle. Molecular and cellular endocrinology 37, 109, 1984.

128
31. Dodson, M.V., Allen, R.E., and Hossner, K.L. Ovine somatomedin, multiplication-

stimulating activity, and insulin promote skeletal muscle satellite cell proliferation in

vitro. Endocrinology 117, 2357, 1985.

32. Sierpinski, P., Garrett, J., Ma, J., Apel, P., Klorig, D., Smith, T., Koman, L.A., Atala,

A., and Van Dyke, M. The use of keratin biomaterials derived from human hair for the

promotion of rapid regeneration of peripheral nerves. Biomaterials 29, 118, 2008.

33. Saul, J.M., Ellenburg, M.D., de Guzman, R.C., and Van Dyke, M. Keratin hydrogels

support the sustained release of bioactive ciprofloxacin. Journal of biomedical materials

research Part A 98, 544, 2011.

34. Wu, X., Corona, B.T., Chen, X., and Walters, T.J. A standardized rat model of

volumetric muscle loss injury for the development of tissue engineering therapies.

BioResearch open access 1, 280, 2012.

35. Collins, C.A., Olsen, I., Zammit, P.S., Heslop, L., Petrie, A., Partridge, T.A., and

Morgan, J.E. Stem cell function, self-renewal, and behavioral heterogeneity of cells from

the adult muscle satellite cell niche. Cell 122, 289, 2005.

36. Musaro, A., Giacinti, C., Borsellino, G., Dobrowolny, G., Pelosi, L., Cairns, L.,

Ottolenghi, S., Cossu, G., Bernardi, G., Battistini, L., Molinaro, M., and Rosenthal, N.

Stem cell-mediated muscle regeneration is enhanced by local isoform of insulin-like

growth factor 1. Proceedings of the National Academy of Sciences of the United States of

America 101, 1206, 2004.

37. Yablonka-Reuveni, Z., Seger, R., and Rivera, A.J. Fibroblast growth factor promotes

recruitment of skeletal muscle satellite cells in young and old rats. The journal of

129
histochemistry and cytochemistry : official journal of the Histochemistry Society 47, 23,

1999.

38. Andreopoulos, F.M., and Persaud, I. Delivery of basic fibroblast growth factor

(bFGF) from photoresponsive hydrogel scaffolds. Biomaterials 27, 2468, 2006.

39. Apel, P.J., Garrett, J.P., Sierpinski, P., Ma, J., Atala, A., Smith, T.L., Koman, L.A.,

and Van Dyke, M.E. Peripheral nerve regeneration using a keratin-based scaffold: long-

term functional and histological outcomes in a mouse model. The Journal of hand surgery

33, 1541, 2008.

40. Pace, L.A., Plate, J.F., Mannava, S., Barnwell, J.C., Koman, L.A., Li, Z., Smith,

T.L., and Van Dyke, M. A human hair keratin hydrogel scaffold enhances median nerve

regeneration in nonhuman primates: an electrophysiological and histological study.

Tissue engineering Part A 20, 507, 2014.

41. Lee, K., Silva, E.A., and Mooney, D.J. Growth factor delivery-based tissue

engineering: general approaches and a review of recent developments. Journal of the

Royal Society, Interface / the Royal Society 8, 153, 2011.

42. Ayele, T., Zuki, A.B., Noorjahan, B.M., and Noordin, M.M. Tissue engineering

approach to repair abdominal wall defects using cell-seeded bovine tunica vaginalis in a

rabbit model. Journal of materials science Materials in medicine 21, 1721, 2010.

43. Merritt, E.K., Cannon, M.V., Hammers, D.W., Le, L.N., Gokhale, R., Sarathy, A.,

Song, T.J., Tierney, M.T., Suggs, L.J., Walters, T.J., and Farrar, R.P. Repair of traumatic

skeletal muscle injury with bone-marrow-derived mesenchymal stem cells seeded on

extracellular matrix. Tissue engineering Part A 16, 2871, 2010.

130
44. Moon du, G., Christ, G., Stitzel, J.D., Atala, A., and Yoo, J.J. Cyclic mechanical

preconditioning improves engineered muscle contraction. Tissue engineering Part A 14,

473, 2008.

45. Hagiwara, K., Chen, G., Kawazoe, N., Tabata, Y., and Komuro, H. Promotion of

muscle regeneration by myoblast transplantation combined with the controlled and

sustained release of bFGFcpr. Journal of tissue engineering and regenerative medicine

2013.

46. Dosier, C.R., Uhrig, B.A., Willett, N.J., Krishnan, L., Li, M.T., Stevens, H.Y.,

Schwartz, Z., Boyan, B.D., and Guldberg, R.E. Effect of cell origin and timing of

delivery for stem cell-based bone tissue engineering using biologically functionalized

hydrogels. Tissue engineering Part A 21, 156, 2015.

47. Chen, C.H., Chang, M.Y., Wang, S.S., and Hsieh, P.C. Injection of autologous bone

marrow cells in hyaluronan hydrogel improves cardiac performance after infarction in

pigs. American journal of physiology Heart and circulatory physiology 306, H1078,

2014.

48. Rossi, C.A., Flaibani, M., Blaauw, B., Pozzobon, M., Figallo, E., Reggiani, C.,

Vitiello, L., Elvassore, N., and De Coppi, P. In vivo tissue engineering of functional

skeletal muscle by freshly isolated satellite cells embedded in a photopolymerizable

hydrogel. FASEB journal : official publication of the Federation of American Societies

for Experimental Biology 25, 2296, 2011.

49. Seebach, E., Freischmidt, H., Holschbach, J., Fellenberg, J., and Richter, W.

Mesenchymal stroma cells trigger early attraction of M1 macrophages and endothelial

131
cells into fibrin hydrogels, stimulating long bone healing without long-term engraftment.

Acta biomaterialia 10, 4730, 2014.

50. Lee, S.H., Kim, I.G., Jung, A.R., Shrestha, K.R., Lee, J.H., Park, K.D., Chung, B.H.,

Kim, S.W., Kim, K.H., and Lee, J.Y. Combined effects of brain-derived neurotrophic

factor immobilized poly-lactic-co-glycolic acid membrane with human adipose-derived

stem cells and basic fibroblast growth factor hydrogel on recovery of erectile

dysfunction. Tissue engineering Part A 20, 2446, 2014.

51. Soler-Botija, C., Bago, J.R., Llucia-Valldeperas, A., Valles-Lluch, A., Castells-Sala,

C., Martinez-Ramos, C., Fernandez-Muinos, T., Chachques, J.C., Pradas, M.M., Semino,

C.E., and Bayes-Genis, A. Engineered 3D bioimplants using elastomeric scaffold, self-

assembling peptide hydrogel, and adipose tissue-derived progenitor cells for cardiac

regeneration. American journal of translational research 6, 291, 2014.

52. Huang, J., Wang, S., Wei, C., Xu, Y., Wang, Y., Jin, J., andTeng, G. In vivo

differentiation of adipose-derived stem cells in an injectable poloxamer-octapeptide

hybrid hydrogel. Tissue & cell 43, 344, 2011.

53. Mulyasasmita, W., Cai, L., Dewi, R.E., Jha, A., Ullmann, S.D., Luong, R.H., Huang,

N.F., and Heilshorn, S.C. Avidity-controlled hydrogels for injectable co-delivery of

induced pluripotent stem cell-derived endothelial cells and growth factors. Journal of

controlled release : official journal of the Controlled Release Society 191, 71, 2014.

54. Mathieu, E., Lamirault, G., Toquet, C., Lhommet, P., Rederstorff, E., Sourice, S.,

Biteau, K., Hulin, P., Forest, V., Weiss, P., Guicheux, J., and Lemarchand, P.

Intramyocardial delivery of mesenchymal stem cell-seeded hydrogel preserves cardiac

132
function and attenuates ventricular remodeling after myocardial infarction. PloS one 7,

e51991, 2012.

55. Fuoco, C., Sangalli, E., Vono, R., Testa, S., Sacchetti, B., Latronico, M.V.,

Bernardini, S., Madeddu, P., Cesareni, G., Seliktar, D., Rizzi, R., Bearzi, C., Cannata,

S.M., Spinetti, G., and Gargioli, C. 3D hydrogel environment rejuvenates aged pericytes

for skeletal muscle tissue engineering. Frontiers in physiology 5, 203, 2014.

133
CHAPTER IV

Investigation of Tissue Engineered Muscle Repair Constructs for Use in Cleft Lip

Secondary Revision Procedures in a Rat Model

Hannah Besser Baker

Department of Biomedical Engineering

Wake Forest University School of Medicine, Winston-Salem, NC 27157

Affiliations: Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences,

Winston-Salem, NC 27157; Wake Forest Institute for Regenerative Medicine,

Winston-Salem, NC 27101

Note: This chapter was composed by Hannah Besser Baker

134
Introduction

Volumetric Muscle Loss (VML) has been defined by Grogan et al as the

irrecoverable loss of function (1). This definition came about through the study of traumatic

injuries observed in active military personnel wherein musculoskeletal traumas make up

as much as 70% of reported injuries (2). It is important to consider that in addition to

traumatic injuries, several other skeletal muscle pathologies and indications are associated

with irrecoverable loss of function. Such conditions include acquired diseases of the

musculature i.e. Bell’s palsy, surgical resection of tissue concurrent with cancer treatment,

and congenital birth defects such as cleft lip and cleft palate (CL/CP) (3, 4). Among these

presentations, cleft lip and cleft palate (CL/CP) are extremely common- occurring in 1 in

1,000 live births the United States (5). Current strategies for repair of CL/CP largely

surround the use of neighboring tissues to act as flaps to bridge tissue voids (6-8).

Advancements in surgical repair strategy have continued to improve patient outcomes after

CL/CP revision; however, secondary revision is typically required (9-12). Present standard

procedures for correction of the most frequent secondary lip and nasal deformities utilize

regional tissue flaps. These procedures can be complicated by tissue deficiency as well as

the quality of the available tissue which can be impaired as a result of the primary repair

procedure (9, 10, 13, 14). Tissue engineered grafts offer an alternative to tissue flaps and

the often associated need to implement tissue expansion devices or regimens to provide

sufficient tissue area for use in secondary revisions (8, 10, 12, 13).

The crucial concern in secondary as well as primary revision of CL/CP is

establishment of continuity and function in the orbicularis oris muscle (OOM) (8, 9, 11,

15, 16). As such, tissue engineering strategies should provide additional graft material

135
which facilitates favorable OOM form and function. Numerous investigators have shown

the utility of decellularized extracellular matrix based technologies in a variety of tissue

engineering applications including promotion of skeletal muscle regeneration in animal

models of VML (17-27). Among these studies, our group has illustrated the capacity of a

porcine bladder acellular matrix (BAM) seeded with autologous muscle cells and subjected

to axial stretch preconditioning to confer recovery of muscle function and intrinsic

morphology in two distinct rodent models of VML (18, 25, 28).

To investigate an established technology for use as a graft in a (CL/CP) secondary

revision procedure in an in vivo study, an appropriate animal model would accommodate

a graft size and surgical application akin to revision procedures with local tissue flaps.

Investigators have indicated the use of rat latissimus dorsi (LD) for studying muscle repair

and VML and have highlighted similarities in architecture between rat LD and human

craniofacial muscles such as the OOM (21, 29). We have recently developed a model in

the rat LD with a defect area larger in size than used in our previous studies. The new

model allows surgical incorporation of a test article by methods which have also been

applied when using autologous tissue flaps in CL/CP revision.

We hypothesize that Tissue Engineered Muscle Repair (TEMR) constructs

developed in previous studies will enable return of both function and form by promotion

of favorable integration with the host tissue and development of new muscle within a VML

model with bio-relevance to cleft lip secondary revision. Moreover we hypothesize that the

addition of a cellular component and stretch preconditioning will improve the recovery in

the TEMR repaired muscles relative to those treated with BAM alone.

136
To evaluate the proposed hypotheses, the newly developed VML model was

employed to investigate both TEMR and BAM constructs in both male and female rats. To

capture the full potential of muscle repair and to investigate potential long term outcomes

this study examined functional and morphological recovery at 2, 4 and 6 months after

injury.

Methods

BAM Preparation

BAM scaffolds were prepared as previously described (25, 28). Briefly, the bladder

was washed and trimmed to obtain the lamina propria, which was placed in 0.05% trypsin

(Hyclone, Logan, UT) for 1 hour at 37 °C. The bladder was then transferred to Dulbecco’s

modified Eagle’s Medium, DMEM, solution supplemented with 10% FBS and 1%

antibiotic antimycotic, AA, (Hyclone) and kept overnight at 4 °C. The preparation was then

washed in a solution containing 1% triton-X (Sigma) and 0.1% ammonium hydroxide

(Fischer Scientific, Pittsburgh, PA) in de-ionized water for 4 days at 4 °C. Finally, the

bladder was then washed in deionized water for 3 days at 4 °C. The decellularized scaffold

was further dissected to obtain a scaffold of 0.2–0.4mm thickness. The scaffolds were then

cut and draped onto custom made silicone molds with a 5.4cm2 working area.

Cell Isolation and Culture

Rat MPCs were isolated from the tibialis anterior and soleus muscles of 4 week old

(male or female depending on the sex of the recipient rats) Lewis Rats purchased from

Charles River Laboratories. Muscles were sterilized in iodine and washed with PBS before

137
transfer to a 0.2% w/v type 1 collagenase (Worthington Biochemical, Lakewood, NJ) low

glucose DMEM solution (Hyclone, Logan, UT) where they were finely minced and then

allowed to further digest at 37 °C for 2 hours. The cell slurry was plated on collagen coated

dishes for 24 hours (as a fibroblast reduction measure) in myogenic media consisting of

high glucose DMEM supplemented with 20% fetal bovine serum (FBS) (Hyclone), 10%

horse serum (HS), 1% chick embryo extract, and 1% antibiotic/antimitotic (AA) (Hyclone).

The following day, the slurry was moved to matrigel (BD) coated plates in myogenic

media. Three days after plating the cells on matrigel plates, the media was changed to

seeding media consisting of low glucose DMEM supplemented with 15% FBS and 1% AA

(Hyclone). At 70-80% confluence cells were passaged. At passage 2 cells were seeded onto

one side of BAM scaffolds at 1x106 cells/cm2, 5.4x106 MPCs/side. 24 hours later passage

2 cells were seeded onto the other side of the BAM scaffold at 1x106cells/cm2. Three days

after the first side was seeded, the scaffolds were switched to differentiation media

consisting of F12 DMEM, 2% HS, 1%AA (Hyclone). After 3 days scaffolds were given

fresh differentiation media.

Mechanical Stretch Preconditioning

After a total of 10 days of static culture on the scaffolds, scaffolds were transferred

to a bioreactor for stretch preconditioning which has been previously described (25). In

seeding media within the bioreactor, scaffolds were mounted onto the two adjacent ledges

with the long side of the scaffold spanning the gap between the ledges. The gap length was

adjusted to ~3cm. Scaffolds were then clamped to the ledges, and the bioreactor was

flooded with seeding media. A proprietary software was used to interact with a Phidget

138
controller for operating the stepper motor of the bioreactor (Phidgets Inc., Calgary, CAN).

Using the software-phidget-motor interface, the scaffolds were stretched and relaxed 10%

of the initial gap length 3 times each minute for the first 5 minutes of each hour for 5 days.

Rat LD VML Model and Implantation of TEMR

12 week old Lewis rats were purchased from Charles River Laboratories. Both male

and female animals were used equally across all groups and time points (Table I). The

VML defect was created by making an incision along the spine, separating the fascia, and

removing a 1.7cm wide and 2.5cm long rectangular section of tissue, ~2mm from the lateral

edge of the muscle and with the lower lateral corner in line with the last palpable rib. For

the NR group, the defect was left alone. For the BAM and TEMR treatment groups, one

BAM or TEMR construct was secured below the defect window with 6-0 vicryl suture by

making ten equally spaced knots. Another construct was then secured above the window

in a similar fashion (Figure 1). The fascia was then sutured closed with 6-0 vicryl and the

skin was closed with 4-0 prolene. The animals were then allowed to recover.

139
Table I. Treatment Groups

Figure 1. Surgical Flow of Rat LD VML Injury and repair with

TEMR. The LD muscle is exposed and defect is outlined (A), the

defect is created (B), and TEMR constructs are sutured below and

above the created defect (C).

140
Muscle Function Analysis

The entire LD muscle was isolated from the thoracolumbar fascia to the humeral

tendon from rats while under anesthesia and the tendon and facial ends were tied with 0-0

silk suture (Ethicon) and transferred to ice cold Krebs-Ringer buffer solution (Sigma;

composition: pH 7.4; concentration in mM: 121.0 NaCl, 5.0 KCl, 0.5 MgCl2, 1.8 CaCl2,

24.0 NaHCO3, 0.4 NaH2PO4, and 5.5 glucose). Muscles were transferred to individual

chambers of a DMT 750 tissue organ bath system (DMT, Ann Arbor, MI) filled with

Krebs-Ringer buffer at 25 °C bubbled with 95% O2 and 5% CO2. The muscles were

positioned between custom-made platinum electrodes with the proximal tendon attached

to a force transducer and the distal tendon to a fixed support. Direct muscle electrical

stimulation (0.2ms pulse at 30 V) was applied across the LD muscle using a Grass S88

stimulator (Grass, Warwick, RI). Real time display and recording of all force

measurements were performed on a PC with Power Lab/8sp (AD Instruments, Colorado

Springs, CO). Once the LD muscles were mounted in the organ bath, the muscles were

allowed to equilibrate for 5 min prior to determining optimal physiological muscle length

(Lo) via a series of twitch contractions. Force as a function of stimulation frequency (1–

150Hz) was measured at 25 °C during isometric contractions (750ms trains of 0.2ms

pulses), with 2 min between contractions. The maximal force of isometric contraction, Po,

was normalized to an approximate physiological cross-sectional area (PCSA), which was

calculated using the following equation, where muscle density is 1.06 g/cm3:

𝑃𝑃𝑃𝑃𝑃𝑃𝑃𝑃 = (𝑤𝑤𝑤𝑤𝑤𝑤 𝑤𝑤𝑤𝑤𝑤𝑤𝑤𝑤ℎ𝑡𝑡 (𝑔𝑔)/ [𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚 𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑𝑑 (𝑔𝑔/𝑐𝑐𝑐𝑐3) 𝑥𝑥 𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚𝑚 𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙𝑙ℎ, 𝐿𝐿𝑜𝑜 (𝑐𝑐𝑐𝑐)]

Equation 1: Physiological Cross Sectional Area (PCSA) Calculation.

141
Additionally the force-frequency curves were fit to the following dose response equation:

𝑓𝑓(𝑥𝑥) = 𝑚𝑚𝑚𝑚𝑚𝑚 − (𝑚𝑚𝑚𝑚𝑚𝑚 − min)/[1 + (𝑥𝑥 ⁄ 𝐸𝐸𝐸𝐸 50 )−𝑛𝑛 ]

Equation 2: Dose Response Equation.

where x is the stimulation frequency in Hz, min is the lowest observed force generated,

equivalent to the twitch force or Pt, max is the largest observed force or Po, EC50 is the

stimulation frequency which yields half the amplitude of the maximum observed force (Po-

Pt), and n is the slope of the linear portion of the force-frequency curve.

Immunohistochemistry and Imaging

No explanted LDs were specifically dedicated for the use of immunohistochemical

analysis, all histology and immunohistochemistry were performed on muscles that had

been through the organ bath analysis. When preparing the tissue, the rat LD, the muscle

was cut in half transversely in the middle of the defect site such that the samples were a

more manageable size for frozen and paraffin sectioning. Hematoxylin and eosin and

Masson’s trichrome stains were conducted by standard techniques to determine the basic

morphology of cells in and around the implant and to observe any inflammatory response.

Images were captured and digitized using a DM4000B Leica Upright Microscope.

Statistical Analysis

Data values are shown as mean ± SEM unless otherwise noted. Data were

statistically analyzed using one way ANOVA. When significance was found (α set to 0.05)

a post-hoc multiple comparison test was used to compare group means (α set to 0.05) using

142
a Fisher Least Significant Difference (LSD) significance test. Statistical analysis was

conducted using GraphPad Prism 6.0 for Windows La Jolla, California.

Results

In vitro force measurement was conducted as described at 2, 4, and 6 months after

implant on whole LD operated and contralateral control muscles harvested from rats in the

three treatment groups at each time point Table I. Measured physical and functional

parameters for each group and parameter statistics are listed in Tables II-IV. EC50 and Hill

slope were calculated by fitting the dose-response function in Equation 2 to force-

frequency curves as shown in Figure 2 for 2 months post-injury male groups. Comparison

of Po, Pt, and P30 Hz is shown for all groups at each time point for males and females (Figures

3-8).

143
144
Table II. Summary of Latissimus Dorsi Muscle Physical and Functional

Measurement Data at 2 Months Post-Injury. Values are mean ± standard error

of the mean. Muscle weight was measured after completion of functional testing.

Lo is the optimal muscle length associated with peak twitch response (Pt). PCSA

was determined using the measured Lo and wet weight using Equation 1. Peak

twitch (Pt), contraction force at 30hz (P30), and maximal isometric contraction force

(Po) were measured by soliciting contractions via direct electric stimuli (0.2ms,

30V) in the organ bath setup. Measured Po was normalized to PCSA to determine

specific force (Specific Po). The stimulation frequency which yielded 50% of the

absolute maximum amplitude (EC50) and the slope of the linear portion of the force-

frequency curves as shown in Figure 2 for selected data sets (n coefficient) were

determined by fitting the curves to function described by Equation 2. Significant

differences (p < 0.05) between TEMR, BAM, and NR treated muscles were

determined by ANOVA and further evaluated using a Fisher Least Significant


a
Difference post-hoc analysis. denotes significant difference from both other

treatment groups

145
146
Table III. Summary of Latissimus Dorsi Muscle Physical and Functional

Measurement Data at 4 Months Post-Injury. Values are mean ± standard error

of the mean. Muscle weight was measured after completion of functional testing.

Lo is the optimal muscle length associated with peak twitch response (Pt). PCSA

was determined using the measured Lo and wet weight using Equation 1. Peak

twitch (Pt), contraction force at 30hz (P30), and maximal isometric contraction force

(Po) were measured by soliciting contractions via direct electric stimuli (0.2ms,

30V) in the organ bath setup. Measured Po was normalized to PCSA to determine

specific force (Specific Po). The stimulation frequency which yielded 50% of the

absolute maximum amplitude (EC50) and the slope of the linear portion of the force-

frequency curves as shown in Figure 2 for selected data sets (n coefficient) were

determined by fitting the curves to function described by Equation 2. Significant

differences (p < 0.05) between TEMR, BAM, and NR treated muscles were

determined by ANOVA and further evaluated using a Fisher Least Significant


a
Difference post-hoc analysis. denotes significant difference from both other

treatment groups

147
148
Table IV. Summary of Latissimus Dorsi Muscle Physical and Functional

Measurement Data at 6 Months Post-Injury. Values are mean ± standard error

of the mean. Muscle weight was measured after completion of functional testing.

Lo is the optimal muscle length associated with peak twitch response (Pt). PCSA

was determined using the measured Lo and wet weight using Equation 1. Peak

twitch (Pt), contraction force at 30hz (P30), and maximal isometric contraction force

(Po) were measured by soliciting contractions via direct electric stimuli (0.2ms,

30V) in the organ bath setup. Measured Po was normalized to PCSA to determine

specific force (Specific Po). The stimulation frequency which yielded 50% of the

absolute maximum amplitude (EC50) and the slope of the linear portion of the force-

frequency curves shown in Figure 2 for selected data sets (n coefficient) were

determined by fitting the curves to function described by Equation 2. Significant

differences (p < 0.05) between TEMR, BAM, and NR treated muscles were

determined by ANOVA and further evaluated using a Fisher Least Significant


a
Difference post-hoc analysis. denotes significant difference from both other

treatment groups

149
Figure 2. Force-Frequency Curve with Dose-Response Curves for Males at

2mos. Two months post-surgery contractions elicited via electrical stimuli from 1-

150Hz is shown for male groups. Data has been fit with dose response curves

according to Equation 2. Curve fitting was used to determine EC50 and Hill Slope

values shown in Tables II. Data points shown are group mean ± SEM for each

stimulation frequency.

150
Figure 3. Comparison of Po across Treatment Groups and Timepoints in

Males. Maximal isometric contraction force, Po, is shown for each group at 2, 4,

and 6 months post-injury. Forces were measured by eliciting contractions via direct

electric stimuli (0.2ms, 30V) from 1-150Hz in the organ bath setup.

Figure 4. Comparison of Po across Treatment Groups and Timepoints in

Females. Maximal isometric contraction force, Po, is shown for each group at 2, 4,

and 6 months post-injury. Forces were measured by eliciting contractions via direct

electric stimuli (0.2ms, 30V) from 1-150Hz in the organ bath setup.

151
Figure 5. Comparison of Pt across Treatment Groups and Timepoints in

Males. Peak twitch force, Pt, is shown for each group at 2, 4, and 6 months post-

injury. Twitches were measured by eliciting contractions via direct electric stimuli

(0.2ms, 30V) at 1Hz in the organ bath setup.

Figure 6. Comparison of Pt across Treatment Groups and Timepoints in

Females. Peak twitch force, Pt, is shown for each group at 2, 4, and 6 months post-

injury. Twitches were measured by eliciting contractions via direct electric stimuli

(0.2ms, 30V) at 1Hz in the organ bath setup.

152
Figure 7. Comparison of P30 Hz across Treatment Groups and Timepoints in

Males. Submaximal contraction force at 30Hz, P30 Hz, is shown for each group at

2, 4, and 6 months post-injury. Forces were measured by eliciting contractions via

direct electric stimuli (0.2ms, 30V) at 30 Hz in the organ bath setup.

Figure 8. Comparison of P30 Hz across Treatment Groups and Timepoints in

Females. Submaximal contraction force at 30Hz, P30 Hz, is shown for each group at

2, 4, and 6 months post-injury. Forces were measured by eliciting contractions via

direct electric stimuli (0.2ms, 30V) at 30 Hz in the organ bath setup.

153
Animal and Model Specifications

Male rats weighed 308 ± 2g and female rats weighed 195 ± 1g at the time of

implant. The average created defect weighed 377 ± 4mg and measured 4.34 ± 0.02cm2 in

males and 267 ± 3mg and 4.28 ± 0.02 cm2 in females with no significant difference between

treatment groups. Control LD weight and length at the time of explant was not significantly

different between time points in either male or female rats (2.23 ± 0.02g and 6.93 ± 0.06cm

in males and 1.21 ± 0.01g and 6.49 ± 0.06cm in females) while body weight was

significantly different at each time point for both sexes (Table V). The injury created in

this study represented 17% of measured total weight of the contralateral control LD at the

time of explant for males and 22% for females.

Table V. Body Weight at Implant and Explant, Defect Weight & Size, and LD

Weight and Lo for Males and Females. Body weights at implant and explant,

defect size and weight, and control LD weight and optimal length, lo, at explant are

shown for males and females for each of the study time points. Statistical

differences were determined by ANOVA p < 0.05. *denotes difference from all

other timepoints (for males and females separately) p < 0.0001.

154
In Vitro Functional Assessment

For males and females respectively injury resulted in an 80% and 73% reduction in

maximum isometric contraction force (Po), an 86% and 77% reduction in peak twitch force

(Pt), and an 83% and 75% reduction in measured submaximal contraction at 30Hz (P30 Hz)

in the No Repair (NR) group relative to uninjured control muscles evaluated at two months.

Two Months Post-Injury

In both males and females TEMR treated muscles had the highest Po, Pt, P30 Hz, and

specific Po though these findings were not significantly different from those of BAM and

NR treated muscles. Weight and length measurements were not significantly different

among the treatment groups in males while female BAM treated muscles were significantly

heavier than TEMR or NR treated tissues. Analysis of dose response curves fit to force-

frequency data (Example shown in Figure 2) indicated that EC50 values and hill slope

coefficients were not different among treatment groups.

Four Months Post-Injury

At four month post injury there were no differences in Po, Pt, P30 Hz or specific Po

between treatments in either males or females. Female BAM treated muscles remained the

heaviest, but were not significantly heavier than TEMR or NR treated muscles. Similar to

the two month time point there were no significant differences among weight and length

between treatment groups for either sex. Determined EC50 values were significantly higher

for male NR muscles and significantly lower for female NR muscles compared with the

155
other treatment groups. Hill slope coefficients were not significantly different between

treatment groups for either sex.

Six Months Post-Injury

As observed at the previous Po, Pt, P30 Hz and specific Po were not different among

treatment groups for either sex. Female BAM treated muscles were still the heaviest, but

not different. Male NR muscles weighed the least but were longest (ns for either parameter)

which contributed to a significantly lower PCSA compared with TEMR and BAM treated

muscles. EC50 and hill slope coefficients were not different among male experimental

tissues while female TEMR data had a significant forward shift in EC50 without a

significant difference in hill slope compared with BAM and NR groups.

Morphology

Macroscopically, there were visible similarities in tissue morphology within

treatment groups. To showcase patterns within treatment groups, time points, and for males

and females, images of all samples are shown in Supplemental Figures 1-18 and three

selected representative samples from each group are shown for each time point for males

and females in Figures 9-14. A summary of findings surrounding tissue density within the

implant region, vascular structure presence, adipose or scar tissue presence, and apparent

blood clots or inflammation is shown in Tables VI-VII for BAM and TEMR treated tissues.

156
Figure 9. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Males at 2mos. Representative TEMR treated tissues are

shown in A-C, BAM treated samples in D-F, and NR samples are shown in G-I.

Samples are shown with the experimental muscle on the left and the uninjured

contralateral control muscle on the right.

157
Figure 10. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Females at 2mos. Representative TEMR treated tissues

are shown in A-C, BAM treated samples in D-F, and NR samples are shown in G-

I. Samples are shown with the experimental muscle on the left and the uninjured

contralateral control muscle on the right.

158
Figure 11. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Males at 4mos. Representative TEMR treated tissues are

shown in A-C, BAM treated samples in D-F, and NR samples are shown in G-I.

Samples are shown with the experimental muscle on the left and the uninjured

contralateral control muscle on the right.

159
Figure 12. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Females at 4mos. Representative TEMR treated tissues

are shown in A-C, BAM treated samples in D-F, and NR samples are shown in G-

I. Samples are shown with the experimental muscle on the left and the uninjured

contralateral control muscle on the right.

160
Figure 13. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Males at 6mos. Representative TEMR treated tissues are

shown in A-C, BAM treated samples in D-F, and NR samples are shown in G-I.

Samples are shown with the experimental muscle on the left and the uninjured

contralateral control muscle on the right.

161
Figure 14. Macroscopic Comparison of Selected Representative Tissues across

Treatment Groups for Females at 6mos. Representative TEMR treated tissues

are shown in A-C, BAM treated samples in D-F, and NR samples are shown in G-

I. Samples are shown with the experimental muscle on the left and the uninjured

contralateral control muscle on the right.

162
Table VI. Summary of Macroscopic Findings in Males. BAM and TEMR

treated tissues were evaluated for tissue quality. Parameters of interest which were

evaluated include connective tissue density, and presence of blood vessels, scar

tissue, and apparent blood clots or inflammation within the connective tissue layers.

The assessment used a scale from (–): little to no evidence, to (+++): extensive

presence. Reported findings for males were relative to male BAM and TEMR

treated tissues.

163
Table VII. Summary of Macroscopic Findings in Females. BAM and TEMR

treated tissues were evaluated for tissue quality. Parameters of interest which were

evaluated include connective tissue density, and presence of blood vessels, scar

tissue, and apparent blood clots or inflammation within the connective tissue layers.

The assessment used a scale from (–): little to no evidence, to (+++): extensive

presence. Reported findings for females were relative to female BAM and TEMR

treated tissues. *Note BAM scaffold remnants in one sample.

164
TEMR treated tissues

Tissues which were treated with TEMR differed between males and females but

were similar for given time points for each sex and resulted in the greatest tissue quality

relative to NR and BAM treated tissues. In males TEMR treated tissues had tissue present

within the defect site which was more opaque at each subsequent time point. Vascular

structures were present at 2 months and to a greater extent at 4 and 6 months. For males

the TEMR treated tissue did not appear to be inflamed and there was no evident scar tissue

or adipose tissue deposition near or within the implant region. For females the tissue quality

was visibility decreased in comparison with male TEMR treated tissues with regard to

opacity, vascularity, and inflammation. Tissue within defects for females treated with

TEMR was visibly less dense than in males at all timepoints. In females blood clots could

be found in samples at each time point and scar tissue could be seen at 6 months. Female

tissues treated with TEMR, like males, did have vascular structures present within the

implant region at each time point but to a lesser extent than was seen in males.

BAM treated tissues

Similar to the morphology seen in TEMR treated tissues, BAM treated muscles’

morphology differed between males and females but similarities were visible within time

points for each sex and resulted in tissue quality which overall surpassed that seen in NR

tissues. For males at 2 months, BAM treated defects were filled in with connective tissue

to a lesser extent than what was seen in TEMR treated muscles at each time point. The

tissue layer tore easily leaving holes visible in samples (Figure 11E-F, Figure 13F). Within

the implant region, some dark clots were visible at 2 and 4 months, but these findings were

165
not present at the 6 month time point. At 6 months BAM treated tissues were more opaque

than at previous time points, tissues were free of the previously seen clots. Within the BAM

treated tissues, unlike TEMR treated tissues, a small extent of adipose and scar tissue

deposition was apparent at the 6 month time point. BAM treated tissues had vascular

structures present within the region of the defect at 2 months with increasing presence at 4

and 6 months, however the vascularity was visibly less than that seen in TEMR treated

tissues for each time point. In females treated with BAM, a tissue layer formed that was

thinner and more frail than that seen in female TEMR tissues for each time point, resulting

in gaps and tears not seen in TEMR treated tissues. The tissue layer in females had blood

clots present, similar to those seen in male BAM treated muscles, at each time point

including 6 months. Unique to female BAM treated tissues was the presence of remaining

BAM scaffold at 2 months such as that seen in Figure 10F. A small extent of vascular

structures were present at each time point within the implant region though, like that seen

in male BAM tissues, to a lesser extent than was seen in TEMR treated tissues.

NR tissues

At all three time points and in both sexes, tissues in which there was no repair (NR

tissues) had a distinct macroscopic appearance (Figures 9-14 G-I). In males, NR muscles

had thin translucent connective tissue layers at 2 months which tore and resulted in holes

due to handling in some samples. The connective tissue layer appeared thicker at 4 months

and thickest at 6 months, but the layer remained translucent at both subsequent time points.

Little to no vascular structures were visible within the connective tissue layers at the 2 and

4 month time points, while at 6 months a greater, though still modest, extent of vascular

166
structures could be seen. Female NR tissues were very similar in appearance to male NR

tissues, unlike the differences seen between the sexes in TEMR and BAM treated tissues.

In female NR tissues, a very thin, translucent, and delicate connective tissue layer was

visible at 2 and 4 months and appeared slightly more substantial at 6 months. The tissue

layer which formed was the least dense of the treatment groups and tore as a result of

handling at all timepoints for females. There was no apparent inflammation or scar or

adipose tissue deposition in the female NR tissues. Vascular structures were not visible

within defect regions for any female NR tissue at any time point.

Histology

As described, following functional testing explanted tissues were fixed and

processed for histological analysis. Hematoxylin and eosin staining and Masson’s

trichrome staining enabled tissue level visualization of the selected representative samples

(Figures 9-14) for each treatment group with additional images of Masson’s trichrome

staining within the implant region, when feasible, for TEMR and BAM treated tissues

(note: samples for macroscopic comparison were also used for histological

comparison)(Figures 15-32). Samples were evaluated for overall presence and apparent

density of the connective tissue layer at the border of and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular structures, and

mononuclear cell infiltrate. A summary of findings for TEMR and BAM groups can be

found in Tables VIII-IX.

167
168
Figure 15. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 2mos Male TEMR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

169
170
Figure 16. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 2mos Male BAM. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

171
172
Figure 17. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 2mos Male NR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group. Samples

were evaluated for overall presence and apparent density of the connective tissue

layer at the border of native tissue and the defect and within the created defect and

for presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

173
174
Figure 18. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 2mos Female TEMR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

175
176
Figure 19. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 2mos Female BAM. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

177
178
Figure 20. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 2mos Female NR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group. Samples

were evaluated for overall presence and apparent density of the connective tissue

layer at the border of native tissue and the defect and within the created defect and

for presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

179
180
Figure 21. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 4mos Male TEMR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

181
182
Figure 22. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 4mos Male BAM. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

183
184
Figure 23. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 4mos Male NR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group. Samples

were evaluated for overall presence and apparent density of the connective tissue

layer at the border of native tissue and the defect and within the created defect and

for presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

185
186
Figure 24. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 4mos Female TEMR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

187
188
Figure 25. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 4mos Female BAM. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

189
190
Figure 26. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 4mos Female NR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group. Samples

were evaluated for overall presence and apparent density of the connective tissue

layer at the border of native tissue and the defect and within the created defect and

for presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

191
192
Figure 27. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 6mos Male TEMR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

193
194
Figure 28. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 6mos Male BAM. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

195
196
Figure 29. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 6mos Male NR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group. Samples

were evaluated for overall presence and apparent density of the connective tissue

layer at the border of native tissue and the defect and within the created defect and

for presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

197
198
Figure 30. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 6mos Female TEMR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

199
200
Figure 31. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 6mos Female BAM. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group.

Additional images of Masson’s trichrome staining within the implant region (G-I)

are shown for TEMR and BAM treated tissues for samples wherein the implant

region was both intact and distinct from the native tissue interface. Samples were

evaluated for overall presence and apparent density of the connective tissue layer

at the border of native tissue and the defect and within the created defect and for

presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

201
202
Figure 32. Hematoxylin and Eosin and Masson’s Trichrome Staining of

Tissues from Selected Representative Tissues: 6mos Female NR. Following

functional testing explanted tissues were fixed and processed for histological

analysis. Hematoxylin and eosin staining (A-C) and Masson’s trichrome staining

(D-F) enabled tissue level visualization of the native tissue-defect and implant

border for the selected representative samples from each treatment group. Samples

were evaluated for overall presence and apparent density of the connective tissue

layer at the border of native tissue and the defect and within the created defect and

for presence and location of adipose and scar tissue deposition, neurovascular

structures (denoted with white arrows), and mononuclear cell infiltrates (denoted

with black arrows).

203
Table VIII. Summary of Key Histological Findings for TEMR and BAM

Treated Tissues in Males. BAM and TEMR tissues sections from representative

samples were evaluated for apparent connective tissue density, mononuclear cell

infiltrates, and presence of neurovascular structures at the interface between native

tissue and the defect-implant region and within the implant region. The assessment

used a scale from (–): little to no evidence, to (+++): extensive presence. Findings

for males were relative to male BAM and TEMR treated tissues.

204
Table IX. Summary of Key Histological Findings for TEMR and BAM

Treated Tissues in Females. BAM and TEMR tissues sections from representative

samples were evaluated for apparent connective tissue density, mononuclear cell

infiltrates, and presence of neurovascular structures at the interface between native

tissue and the defect-implant region and within the implant region. The assessment

used a scale from (–): little to no evidence, to (+++): extensive presence. Findings

for females were relative to female BAM and TEMR treated tissues.

205
TEMR treated tissues

Males and females treated with TEMR had histological presentations with notable

similarities which highlighted the characteristics of repair associated with TEMR treatment

(Figures 15, 21, and 27 and 18, 24, and 30). These similarities included vascular structure

presence and little to no inflammation at all timepoints. Comparison also highlighted a

divergence in observed tissue quality between males and females (also observed in

comparing males and females in BAM and NR treatment groups).

At 2 months male TEMR treated muscles showed a dense, collagen rich tissue

bridge which spanned the defect created in the native tissue. Vascular structures were

present to a great extent in all evaluated samples at the borders and well within the

implanted constructs. A moderate mononuclear cell presence was observed in one sample

(Figure 15A) while other samples did not show signs of inflammation. At 4 months, the

collagen rich tissue bridge remained intact with vascular structures present throughout and

with some visible adipose tissue deposition within the tissue bridge. Mononuclear cells

were not observed in any of the samples at this timepoint. At 6 months, tissues appeared

similar to those observed at the 4 month time point: the connective tissue layer was collagen

dense and free of mononuclear cells infiltration in all samples. Two samples at this time

point presented with well-defined vascular structures near the implant-native tissue border

which were larger in diameter than vascular structures observed at previous time points for

TEMR (Figure 27A-B).

Female tissues treated with TEMR had thin connective tissue layers present at 2

months post-surgery which appeared to be more collagenous closer to the border of the

defect with increasing adipose presence further from the border and within the implant. In

206
two samples moderate mononuclear cell infiltrates could be observed with a coagulations

of cells visible in one sample (Figure 18A-B). Vascular structures were visible at the border

of the implant and native tissue with decreasing presence further from the native tissue-

implant border. At 4 months female TEMR treated tissues showed a dense collagenous

tissue ‘cap’ beginning to form at the edge of the native tissue. Within the observed collagen

cap, vascular structures were seen. Beyond the collagenous cap tissue layer, the connective

tissue was observed to have a greater extent of adipose tissue deposition and fewer vascular

structures. There was no evidence of mononuclear cell infiltration or inflammation in any

sample at 4 months. The overall tissue density and quality at 6 months was similar to that

seen at 4 months. There was however, an increased presence of vascular cells within the

adipose rich region of the tissue bridge (within the area occupied by the implant).

BAM treated tissues

Males and females treated with BAM had similarities in tissue morphology with

regard to tissue density and inflammation. Comparison of males and females also

exemplified the differences seen in overall tissue quality between the sexes that had also

been seen in TEMR treated tissues (Figures 16, 22, and 28 and 19, 25, and 31).

Males treated with BAM at 2 months had a dense collagenous layer formed within

the implant region. Mononuclear cells could be seen within the layer in all samples with a

visible mass of cells present in one sample (Figure 16C). New blood vessels could be seen

within the tissue layer, with a higher concentration of structures closer to the implant-

muscle interface. At 4 months, the tissue density appeared to be decreased further away

from the native tissue and further into the implant region. Male BAM samples were

207
observed to have little to no mononuclear cell presence surrounding the edge of the implant

at 4 months compared with 2 months. At 4 months, issues had more frequent vascular

structures present at the border than within the implant. At 6 months the tissue density

appeared unchanged. Mononuclear cell presence had decreased as well and vascularity also

appeared to be less prevalent beyond the tissue-implant border than that observed at 4

months.

Females treated with BAM had thin collagen rich tissue layers within the implant

site at 2 months. There was visible inflammation in all samples with congregations of

mononuclear cells observed in each sample. Vascular structures could be seen within the

connective tissue layer in the vicinity of the inflammatory regions and along the edges of

the native tissue-implant interface. At 4 months, tissue density within the implant region

appeared have decreased and had a higher presence of adipose cells. There was also no

visible inflammation at this time point. At 4 months in BAM treated females, there were

few visible vascular structures along the interface or within the implant site. Histological

findings were similar at 6 months with more collagen present along the border of the

implant region which included a greater extent of vascular structures than was seen at 4

months and little to no visible mononuclear cells present.

NR tissues

Among treatment groups, male and female NR tissues were the most similar in

overall appearance and tissue quality (Figures 17, 23, and 29 and 20, 26, and 32). Males

developed a thin adipose heavy tissue layer within the region of the defect at 2 months.

Within the connective tissue layer there was little to no evidence of new vascular structure

208
formation nor was there evidence of inflammation. At 4 months the connective layer was

more collagen heavy forming a cap along the native tissue edge with no change in

vascularity or inflammation. The tissue cap seen at 4 months could also been seen at 6

months and appeared to be thicker. At 6 months there was also no inflammation present

nor extensive new vascular structure compared with the prior timepoints. In females, at 2

months there was a thin connective tissue layer rich with adipose cells. There was little to

no vascular structures or inflammation present compared with other treatment groups in

females. At 4 months, the thin connective tissue layer remained adipose rich and a cap of

tissue surrounding the native tissue could be seen as was observed in male NR tissues.

Observed vascularity remained minimal at 4 months. The tissue layer surrounding the

native tissue was more well defined and collagen heavy at 6 months. As was seen in

previous timepoints for NR female tissues, there was little to no evidence of vascular

formation within the implant region as well as no visible signs of inflammation.

Discussion

Tissue engineering strategies for CL/CP revision have largely surrounded cartilage

and bone repair for CP applications (30-37). Among CL/CP TE research there have been

few endeavors focused on soft tissue repair (21, 38-41). As it would follow, there also

existed a lack of animal models approximating human craniofacial muscles for

investigation of treatment for CL/CP (21, 42-46). As such, the first major goal of the

present research was to develop an injury model in the rat latissimus dorsi approximating

secondary revision of CL. Secondly, the intent of this study was to test the capacity of a

previously established technology to promote muscle healing and regeneration in the newly

209
developed model. The therapy of interest, TEMR, has shown promising results in models

of VML carried out in the mouse latissimus dorsi and the rat tibialis anterior. Results

indicated improved recovery of muscle cell and tissue morphology and corresponding

significantly improved contraction forces (18, 25, 28). As preservation of the OOM is a

major necessity of CL/CP revision, the TEMR technology was well suited for investigation

as a TE alternative graft material (8, 16). Herein, we hypothesized that TEMR would

promote recovery of morphology and function in a newly developed model of VML with

relevance to CL/CP secondary revision.

To test the hypothesis of this study, a new VML model was developed utilizing the

rat latissimus dorsi which would allow the evaluation of TEMR as well as other future

therapies of interest as potential candidates for use in CL/CP revision. In evaluating the

effect of TEMR on muscle repair and regeneration in this model, functional and

morphological outcomes were assessed in comparison with non-repaired (NR) injuries and

BAM alone treated injuries. By investigating the treatments alongside one another in both

males and females and over the course of 6 months, the study enabled the investigation of

the effectiveness of TEMR, it showcased the value of including a cellular component in

VML treatment, and gave insight into recovery timelines in VML.

Use of in vitro contraction force assessment indicated that the model resulted in an

80% reduction in maximal contraction force for NR males compared with control muscles

at 2 months post-surgery and that the contraction force improved to a ~50% reduction by

4 months and remained unchanged at 6 months. In females the same injury size, although

accounting for a greater percentage of the total muscle weight (22% in females versus 17%

in males), resulted in a ~70% reduction in force at 2 months and did not improve until 6

210
months at which time the deficit improved to a 60% reduction of maximal force compared

with control muscles. While the model did result in an injury in which there was

improvement in function over time, the maximal recovery did plateau for males at 4 months

and remained even lower for females at 6 months with 50% and 60% maximal contraction

deficits respectively. The resulting functional deficits in this model are similar to those

created in previously established rodent models of VML and as such this model can

reasonably be considered to result in true VML (18, 25, 28).

Unexpectedly, while functional recovery was greatest for TEMR compared with

NR and BAM at 2 months for males and females the differences in parameters were not

different and at later time points there were no significant differences between treatment

groups for males or females in the assessed contraction parameters. Interestingly the lack

of differences seen functionally did not correlate with the macroscopic and histologic

findings for which there were clear differences between treatment groups. Namely, NR

tissues showed a distinct void where the defect was created while BAM and TEMR treated

tissues had connective tissue layers which for most males and some females spanned the

region of the defect and persisted through the 6 month time point. Furthermore, TEMR

treated tissues in both males and females appeared to be in a distinctly better condition than

either NR or BAM treated tissues. TEMR treated tissues appeared more homogenous

overall macroscopically and had fewer incidences of holes or tears and apparent

inflammation both macroscopically and in histological results. In addition, within the

TEMR tissue’s implant regions there was a greater incidence of vascular structures

compared with BAM tissues in both males and females at all timepoints.

211
The differences between TEMR and BAM treated tissues seen macroscopically and

through histological analysis in spite of differences in functional recovery prompt further

assessment of the impact of the addition of a cellular component. The addition of MPCs

and preconditioning was previously shown to improve both morphology and functional

recovery (18, 25, 28). However, in this study only the former was demonstrated and while

tissue quality within the implant was more vascular and TEMR was apparently less

inflammatory than BAM, there was no evidence of neo-muscle formation as was seen

alongside improved function in those studies (18, 25, 28). In prior work in which TEMR

was employed in a mouse model of VML in the LD muscle a 3x1cm strip carrying 1x106

MPCs/cm2 per side or 6x106 MPCs total was implanted into a defect weighing ~25mg (25,

28). The size of the defect created in the mouse LD model was approximately 11 fold less

than the defect created in the female rat LD in this study, 256mg, and 15 fold less than in

males, 377mg, but in this model only ~3 fold the number of cells was employed: ~17x106

MPCs versus 6x106 MPCs (number of cells implanted in this study based on seeding 1x106

MPCs per side for two scaffolds cut to fit a 4.25cm2 defect). Therefore it may be postulated

that the concentration of cells used in this model was insufficient in promoting functional

recovery for the size of the injury generated based on previous successes. The literature

suggests that implanted muscle progenitor cells may impact regeneration through direct

generation of new muscle or indirectly through paracrine effects on surrounding cells (47-

51). In this study, the histologic findings indicate that while the implanted MPCs were not

capable of resulting in new muscle tissue formation they had paracrine effects on

surrounding tissue which resulted in improved angiogenesis within the implanted

construct. In addition, the cellular component may be attributed to the improved immune

212
modulation seen in TEMR treated tissues as previous studies have indicated that

phagocytosis of muscle cellular debris by macrophages promotes transformation from the

pro-inflammatory to anti-inflammatory phenotype (51).

When measuring success in a CL/CP secondary revision procedure, aesthetic

restoration is a key concern for patients and has been indicated as the primary reason for

undergoing revision procedures in most cases (11-14). The findings of this study indicated

the potential of TEMR constructs for use as a graft material in secondary CL/CP revision

procedures capable of promoting improved tissue morphology. Herein, TEMR

demonstrated the ability to bridge a large defect with a connective tissue layer wherein the

tissue was demonstrated both macroscopically and histologically to be well vascularized

and non-inflammatory. However, the concentration of MPCs applied in the study did not

improve functional recovery relative to NR or BAM treatments as we had seen in previous

work (18, 25, 28). These outcomes will help shape future studies wherein the goal will be

to enhance neo-tissue formation within the region of the implant and correspondingly

increase functional improvement while maintaining or improving the macroscopic tissue

quality that was demonstrated in this study. Compelling follow up investigations might

include increasing the number of TEMR constructs implanted into the defect or

incorporating the addition of a second cell seeding during preconditioning. Adding more

layers of TEMR to the implant would serve to increase the number of cells added and could

potentially improve the longevity of the implant to encourage an improved balance of tissue

remodeling to scaffold degradation within the implant region. Use of an additional cell

seeding of undifferentiated MPCs during preconditioning could benefit this model through

213
promotion of new muscle formation within the scaffold region with potential for

accompanied functional improvement as was shown by Corona et al (28).

This study has benefited skeletal muscle TE research through the introduction of a

VML model in which the size poses a novel challenge for existing technologies wherein

success demonstrates a high potential for translation. The hypothesis of this study could be

partially accepted: TEMR was capable of promoting improved tissue quality after VML

albeit in the absence of improved functional recovery and new muscle generation.

Nonetheless, this work has indicated the potential of TEMR for use in CL/CP secondary

revision and has elucidated the benefits conferred through the addition of cells in TE which

future studies involving TEMR should strive to build upon.

214
Supplemental Figure 1. Macroscopic Images of all 2mos Male TEMR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

215
Supplemental Figure 2. Macroscopic Images of all 2mos Male BAM Tissues. Excised

whole LD muscles are shown after undergoing functional testing. Experimental LD

muscle is shown on the left and contralateral uninjured control muscle is shown on the

right in each image.

216
Supplemental Figure 3. Macroscopic Images of all 2mos Male NR Tissues. Excised

whole LD muscles are shown after undergoing functional testing. Experimental LD

muscle is shown on the left and contralateral uninjured control muscle is shown on the

right in each image.

217
Supplemental Figure 4. Macroscopic Images of all 4mos Male TEMR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

218
Supplemental Figure 5. Macroscopic Images of all 4mos Male BAM Tissues. Excised

whole LD muscles are shown after undergoing functional testing. Experimental LD

muscle is shown on the left and contralateral uninjured control muscle is shown on the

right in each image.

219
Supplemental Figure 6. Macroscopic Images of all 4mos Male NR Tissues. Excised

whole LD muscles are shown after undergoing functional testing. Experimental LD

muscle is shown on the left and contralateral uninjured control muscle is shown on the

right in each image.

220
Supplemental Figure 7. Macroscopic Images of all 6mos Male TEMR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

221
Supplemental Figure 8. Macroscopic Images of all 6mos Male BAM Tissues. Excised

whole LD muscles are shown after undergoing functional testing. Experimental LD

muscle is shown on the left and contralateral uninjured control muscle is shown on the

right in each image.

222
Supplemental Figure 9. Macroscopic Images of all 6mos Male NR Tissues. Excised

whole LD muscles are shown after undergoing functional testing. Experimental LD

muscle is shown on the left and contralateral uninjured control muscle is shown on the

right in each image.

223
Supplemental Figure 10. Macroscopic Images of all 2mos Female TEMR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

224
Supplemental Figure 11. Macroscopic Images of all 2mos Female BAM Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

225
Supplemental Figure 12. Macroscopic Images of all 2mos Female NR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

226
Supplemental Figure 13. Macroscopic Images of all 4mos Female TEMR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

227
Supplemental Figure 14. Macroscopic Images of all 4mos Female BAM Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

228
Supplemental Figure 15. Macroscopic Images of all 4mos Female NR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

229
Supplemental Figure 16. Macroscopic Images of all 6mos Female TEMR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

230
Supplemental Figure 17. Macroscopic Images of all 6mos Female BAM Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

231
Supplemental Figure 18. Macroscopic Images of all 6mos Female NR Tissues.

Excised whole LD muscles are shown after undergoing functional testing. Experimental

LD muscle is shown on the left and contralateral uninjured control muscle is shown on

the right in each image.

232
Literature Cited

1. Grogan, B.F., Hsu, J.R., and Skeletal Trauma Res, C. Volumetric Muscle Loss.

Journal of the American Academy of Orthopaedic Surgeons 19, S35, 2011.

2. Covey, D.C. Blast and fragment injuries of the musculoskeletal system. The Journal of

bone and joint surgery American volume 84-A, 1221, 2002.

3. Bianchi, B., Copelli, C., Ferrari, S., Ferri, A., and Sesenna, E. Free flaps: Outcomes

and complications in head and neck reconstructions. Journal of Cranio-Maxillofacial

Surgery 37, 438, 2009.

4. Genden, E.M., Rinado, A., Suarez, C., Wei, W.I., Bradley, P.J., and Ferlito, A.

Complications of free flap transfers for head and neck reconstruction following cancer

resection. Oral Oncology 40, 979, 2004.

5. Canfield, M.A., Honein, M.A., Yuskiv, N., Xing, J., Mai, C.T., Collins, J.S., Devine,

O., Petrini, J., Ramadhani, T.A., Hobbs, C.A., Kirby, R.S., and Natl Birth Defects

Prevention, N. National estimates and race/ethnic-specific variation of selected birth

defects in the United States, 1999-2001. Birth Defects Research Part a-Clinical and

Molecular Teratology 76, 747, 2006.

6. Nakamura, N., Okawachi, T., Nishihara, K., Hirahara, N., and Nozoe, E. Surgical

Technique for Secondary Correction of Unilateral Cleft Lip-Nose Deformity: Clinical

and 3-Dimensional Observations of Preoperative and Postoperative Nasal Forms. Journal

of Oral and Maxillofacial Surgery 68, 2248, 2010.

7. Fisher, D.M., and Sommerlad, B.C. Cleft Lip, Cleft Palate, and Velopharyngeal

Insufficiency. Plastic and Reconstructive Surgery 128, 342E, 2011.

233
8. Wu, D., Wang, Y., Song, T., Li, H., Wu, J., Qu, W., Ma, H., and Yin, N. Aesthetic

reconstruction of the upper lip with novel split musculomucosal-pedicle cross-lip flap.

Annals of plastic surgery 73 Suppl 1, S88, 2014.

9. Cohen, M., Smith, B.E., and Daw, J.L. Secondary unilateral cleft lip nasal deformity:

Functional and esthetic reconstruction. Journal of Craniofacial Surgery 14, 584, 2003.

10. Stal, S., and Hollier, L. Correction of secondary cleft lip deformities. Plastic and

Reconstructive Surgery 109, 1672, 2002.

11. Cheema, S.A., and Asim, M. An Analysis of Deformities in Revision Surgeries for

Secondary Unilateral Cleft Lip. Jcpsp-Journal of the College of Physicians and Surgeons

Pakistan 24, 666, 2014.

12. Mulliken, J.B., and Martinez-Perez, D. The principle of rotation advancement for

repair of unilateral complete cleft lip and nasal deformity: Technical variations and

analysis of results. Plastic and Reconstructive Surgery 104, 1247, 1999.

13. Monson, L.A., Khechoyan, D.Y., Buchanan, E.P., and Hollier, L.H. Secondary Lip

and Palate Surgery. Clinics in Plastic Surgery 41, 301, 2014.

14. Lee, S.W., Kim, M.H., and Baek, R.M. Correction of secondary vermilion notching

deformity in unilateral cleft lip patients: Complete revision of two errors. Journal of

Cranio-Maxillofacial Surgery 39, 326, 2011.

15. Ridgway, E.B., Estroff, J.A., and Mulliken, J.B. Thickness of Orbicularis Oris

Muscle in Unilateral Cleft Lip: Before and After Labial Adhesion. Journal of

Craniofacial Surgery 22, 1822, 2011.

16. Seagle, M.B., and Furlow, L.T. Muscle reconstruction in cleft lip repair. Plastic and

Reconstructive Surgery 113, 1537, 2004.

234
17. Brown, B.N., and Badylak, S.F. Extracellular matrix as an inductive scaffold for

functional tissue reconstruction. Translational Research 163, 268, 2014.

18. Corona, B.T., Ward, C.L., Baker, H.B., Walters, T.J., and Christ, G.J. Implantation

of In Vitro Tissue Engineered Muscle Repair Constructs and Bladder Acellular Matrices

Partially Restore In Vivo Skeletal Muscle Function in a Rat Model of Volumetric Muscle

Loss Injury. Tissue Engineering Part A 20, 705, 2014.

19. Sicari, B.M., Rubin, J.P., Dearth, C.L., Wolf, M.T., Ambrosio, F., Boninger, M.,

Turner, N.J., Weber, D.J., Simpson, T.W., Wyse, A., Brown, E.H.P., Dziki, J.L., Fisher,

L.E., Brown, S., and Badylak, S.F. An Acellular Biologic Scaffold Promotes Skeletal

Muscle Formation in Mice and Humans with Volumetric Muscle Loss. Science

Translational Medicine 6, 10, 2014.

20. Teodori, L., Costa, A., Marzio, R., Perniconi, B., Coletti, D., Adamo, S., Gupta, B.,

and Tarnok, A. Native extracellular matrix: a new scaffolding platform for repair of

damaged muscle. Frontiers in physiology 5, 218, 2014.

21. Chen, X.Y.K., and Walters, T.J. Muscle-derived decellularised extracellular matrix

improves functional recovery in a rat latissimus dorsi muscle defect model. Journal of

Plastic Reconstructive and Aesthetic Surgery 66, 1750, 2013.

22. Corona, B.T., Wu, X., Ward, C.L., McDaniel, J.S., Rathbone, C.R., and Walters, T.J.

The promotion of a functional fibrosis in skeletal muscle with volumetric muscle loss

injury following the transplantation of muscle-ECM. Biomaterials 34, 3324, 2013.

23. Turner, N.J., Badylak, J.S., Weber, D.J., and Badylak, S.F. Biologic Scaffold

Remodeling in a Dog Model of Complex Musculoskeletal Injury. Journal of Surgical

Research 176, 490, 2012.

235
24. Wolf, M.T., Daly, K.A., Reing, J.E., and Badylak, S.F. Biologic scaffold composed

of skeletal muscle extracellular matrix. Biomaterials 33, 2916, 2012.

25. Machingal, M.A., Corona, B.T., Walters, T.J., Kesireddy, V., Koval, C.N.,

Dannahower, A., Zhao, W.X., Yoo, J.J., and Christ, G.J. A Tissue-Engineered Muscle

Repair Construct for Functional Restoration of an Irrecoverable Muscle Injury in a

Murine Model. Tissue Engineering Part A 17, 2291, 2011.

26. Merritt, E.K., Cannon, M.V., Hammers, D.W., Le, L.N., Gokhale, R., Sarathy, A.,

Song, T.J., Tierney, M.T., Suggs, L.J., Walters, T.J., and Farrar, R.P. Repair of Traumatic

Skeletal Muscle Injury with Bone-Marrow-Derived Mesenchymal Stem Cells Seeded on

Extracellular Matrix. Tissue Engineering Part A 16, 2871, 2010.

27. Merritt, E.K., Hammers, D.W., Tierney, M., Suggs, L.J., Walters, T.J., and Farrar,

R.P. Functional Assessment of Skeletal Muscle Regeneration Utilizing Homologous

Extracellular Matrix as Scaffolding. Tissue Engineering Part A 16, 1395, 2010.

28. Corona, B.T., Machingal, M.A., Criswell, T., Vadhavkar, M., Dannahower, A.C.,

Bergman, C., Zhao, W.X., and Christ, G.J. Further Development of a Tissue Engineered

Muscle Repair Construct In Vitro for Enhanced Functional Recovery Following

Implantation In Vivo in a Murine Model of Volumetric Muscle Loss Injury. Tissue

Engineering Part A 18, 1213, 2012.

29. Kurita, M., Yamazaki, K., Eto, H., Seike, S., Takushima, A., and Harii, K.

REINNERVATION OF SEGMENTED LATISSIMUS DORSI MUSCLE WITH THE

DISTAL STUMP OF THE THORACODORSAL NERVE: A PRELIMINARY

EXPERIMENTAL STUDY IN RATS. Microsurgery 33, 545, 2013.

236
30. Moreau, J.L., Caccamese, J.F., Coletti, D.P., Sauk, J.J., and Fisher, J.P. Tissue

engineering solutions for cleft palates. Journal of Oral and Maxillofacial Surgery 65,

2503, 2007.

31. Hoshi, K., Fujihara, Y., Asawa, Y., Nishizawa, S., Kanazawa, S., Sakamoto, T.,

Watanabe, M., Saijo, H., Mori, Y., and Takato, T. Research and development of implant-

type tissue-engineered cartilage clinically applied for the nasal correction of the cleft lip

and palate patients. Journal of Tissue Engineering and Regenerative Medicine 6, 1, 2012.

32. Bichara, D.A., Zhao, X., Hwang, N.S., Bodugoz-Senturk, H., Yaremchuk, M.J.,

Randolph, M.A., and Muratoglu, O.K. Porous Poly(vinyl alcohol)-Alginate Gel Hybrid

Construct for Neocartilage Formation Using Human Nasoseptal Cells. Journal of Surgical

Research 163, 331, 2010.

33. Sharma, B., and Elisseeff, J.H. Engineering structurally organized cartilage and bone

tissues. Annals of Biomedical Engineering 32, 148, 2004.

34. Solchaga, L.A., Yoo, J.U., Lundberg, M., Dennis, J.E., Huibregtse, B.A., Goldberg,

V.M., and Caplan, A.I. Hyaluronan-based polymers in the treatment of osteochondral

defects. Journal of Orthopaedic Research 18, 773, 2000.

35. Liu, X.H., and Ma, P.X. Polymeric scaffolds for bone tissue engineering. Annals of

Biomedical Engineering 32, 477, 2004.

36. Sachlos, E., Reis, N., Ainsley, C., Derby, B., and Czernuszka, J.T. Novel collagen

scaffolds with predefined internal morphology made by solid freeform fabrication.

Biomaterials 24, 1487, 2003.

237
37. Bueno, D.F., Tanikawa, D., Rocha, D.L., Pinheiro, C., Andrade, M.A., and Reis, L.L.

Bone Tissue Engineering for Cleft Lip and Palate Patients Using Non-Invasive Sources

of Stem Cells. Tissue Engineering Part A 20, S89, 2014.

38. Xiong, X.P., Jia, J., He, S.G., and Zhao, Y.F. Cryopreserved lip mucosa tissue

derived keratinocytes can fabricate tissue engineered palatal mucosa equivalent. Journal

of Biomedical Materials Research Part B-Applied Biomaterials 94B, 165, 2010.

39. Izumi, K., Terashi, H., Marcelo, C.L., and Feinberg, S.E. Development and

characterization of a tissue-engineered human oral mucosa equivalent produced in a

serum-free culture system. Journal of Dental Research 79, 798, 2000.

40. Izumi, K., Feinberg, S.E., Iida, A., and Yoshizawa, M. Intraoral grafting of an ex

vivo produced oral mucosa equivalent: a preliminary report. International Journal of Oral

and Maxillofacial Surgery 32, 188, 2003.

41. Ophof, R., van Rheden, R.E.M., Von den Hoff, J.W., Schalkwijk, J., and Kuijpers-

Jagtman, A.M. Oral keratinocytes cultured on dermal matrices form a mucosa-like tissue.

Biomaterials 23, 3741, 2002.

42. Ophof, R., Maltha, J.C., Kuijpers-Jagtman, A.M., and Von Den Hoff, J.W.

Evaluation of a collagen-glycosaminoglycan dermal substitute in the dog palate. Tissue

Engineering 13, 2689, 2007.

43. Ophof, R., Maltha, J.C., Kuijpers-Jagtman, A.M., and Von den Hoff, J.W.

Implantation of tissue-engineered mucosal substitutes in the dog palate. European Journal

of Orthodontics 30, 1, 2008.

238
44. Letic-Gavrilovic, A., Piattelli, A., and Abe, K. Nerve growth factor beta(NGF beta)

delivery via a collagen/hydroxyapatite (Col/HAp) composite and its effects on new bone

growth. Journal of Materials Science-Materials in Medicine 14, 95, 2003.

45. Edington, H.D.J., Mooney, M.P., Losken, A., Hurwitz, D.J., and Siegel, M.I. The

effects of soft-tissue expansion on lip repair and midfacial growth in a rabbit cleft lip

model. Annals of Plastic Surgery 41, 171, 1998.

46. Mehrara, B.J., Saadeh, P.B., Steinbrech, D.S., Dudziak, M., Grayson, B.H., Cutting,

C.B., McCarthy, J.G., Gittes, G.K., and Longaker, M.T. A rat model of

gingivoperiosteoplasty. Journal of Craniofacial Surgery 11, 54, 2000.

47. Gharaibeh, B., Lavasani, M., Cummins, J.H., and Huard, J. Terminal differentiation

is not a major determinant for the success of stem cell therapy - cross-talk between

muscle-derived stem cells and host cells. Stem Cell Research & Therapy 2, 12, 2011.

48. Paylor, B., Natarajan, A., Zhang, R.H., and Rossi, F. NONMYOGENIC CELLS IN

SKELETAL MUSCLE REGENERATION. In: Pavlath G.K., ed. Current Topics in

Developmental Biology: Myogenesis. San Diego: Elsevier Academic Press Inc; 2011. pp.

139.

49. Li, Y., and Huard, J. Differentiation of muscle-derived cells into myofibroblasts in

injured skeletal muscle. American Journal of Pathology 161, 895, 2002.

50. Zammit, P.S., Heslop, L., Hudon, V., Rosenblatt, J.D., Tajbakhsh, S., Buckingham,

M.E., Beauchamp, J.R., and Partridge, T.A. Kinetics of myoblast proliferation show that

resident satellite cells are competent to fully regenerate skeletal muscle fibers.

Experimental Cell Research 2812002.

239
51. Arnold, L., Henry, A., Poron, F., Baba-Amer, Y., van Rooijen, N., Plonquet, A.,

Gherardi, R.K., and Chazaud, B. Inflammatory monocytes recruited after skeletal muscle

injury switch into antiinflammatory macrophages to support myogenesis. Journal of

Experimental Medicine 204, 1057, 2007.

240
CHAPTER V

Summary, Conclusions, and Future Directions

Hannah Besser Baker

Department of Biomedical Engineering

Wake Forest University School of Medicine, Winston-Salem, NC 27157

Affiliations: Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences,

Winston-Salem, NC 27157; Wake Forest Institute for Regenerative Medicine,

Winston-Salem, NC 27101

Note: This chapter was composed by Hannah Besser Baker

241
Introduction

The work presented in this dissertation further demonstrated the potential of natural

biomaterials for use in skeletal muscle TE applications and represents an effort to further

develop a platform of technologies aimed to meet the diverse presentations of VML. The

two materials of interest: human hair derived keratin hydrogels and porcine derived BAM

showcase the utility and diversity of natural scaffolds. Both are benefitted by certain

intrinsic properties known to many natural biomaterials including the capacity to bind cells

through protein sequences, degradation into biocompatible components, and accessibility

with the capacity to be manufactured on a large scale (1-9).

However similar the two materials may be found to be, they are also widely

different physically especially when comparing the states of the materials investigated in

these studies, a hydrogel versus sheet like scaffold which both offer unique benefits.

Keratin hydrogels may act as cell and drug delivery vehicles, capable of achieving

sustained release of a drug of interest over the course of degradation (10-12). The hydrogel

can flow to fill tortuous voids and in doing so can deliver carried materials to locations

other scaffold geometries cannot interact with as efficiently. The unique biochemical

properties of keratin allow it to be isolated in two distinct ways yielding different

degradation rates. The different keratins may be combined in variable ratios to achieve a

desired degradation time, perhaps unique to a specific injury anatomy, pathology, or

patient.

The BAM scaffold has the strength to retain sutures and therefore can transmit

force- an especially important consideration in skeletal muscle tissue engineering (13-20).

Further, while strong, the collagen rich ECM scaffold is also flexible which allows it to

242
undergo uniaxial stretch enabling physiological preconditioning of seeded cells. The

resulting benefits are a more aligned and mature cell phenotype found to be directly

correlated with improved functional recovery in VML models (13-16).

In covering the work carried out using these two promising materials, the following

chapter will serve to first summarize and refresh the findings of the literature search in the

first chapter and of the studies presented in the subsequent chapters. Thereafter the findings

from each study will compared to highlight observed patterns and phenomena and to

suggest limitations of these efforts. A section detailing potential future endeavors will

follow and lastly translational considerations will be addressed.

Summary of Chapters

The literature review presented in the first chapter covered skeletal muscle clinical

indications, biology with a focus on regeneration, and current tissue engineering strategies

for treating VML with a focus on natural biomaterials. Review of the research literature

highlighted the importance of animal models and animal model selection in skeletal muscle

research and particularly when studying VML. The research reviewed also shed light on

the complexity of chemical cues involved in repair of muscle and showed examples of

successful use of growth factors in TE applications. Finally the review covered background

research surrounding the two materials used in this dissertation, keratin hydrogels and

TEMR, and emphasized their utility for use in skeletal muscle repair applications.

The following two chapters investigated the novel application of keratin hydrogels

in VML models. In these studies keratin hydrogels consisting of a blend of KOS and KTN

keratins capable of achieving sustained release of IGF-1 and bFGF were examined. The

243
growth factors were chosen due to their known effects on skeletal muscle cells: IGF-1 is

associated with promoting satellite cell differentiation and fusion of myoblasts and bFGF

is associated with resulting in satellite cell activation and fusion (21-24). In both studies,

we hypothesized that keratin hydrogels would result in improved recovery of muscle

function and morphology and that the addition of cells and selected growth factors would

further enhance recovery.

In chapter 2, keratin hydrogels were applied in a previously established VML model

in the mouse LD (13, 16). In this model, functional recovery was measured in whole

excised LD muscles at a terminal time point through measurement of contraction force

elicited through electrical stimulation using an a specialized organ bath set-up. Histological

analysis was then conducted on tissues after they have been functionally assessed and

consisted of examining overall tissue morphology via hematoxylin and eosin and Masson’s

trichrome staining and staining for the functional protein myosin. At 8 weeks post-surgery,

tissues treated with keratin in combination with IGF-1 and bFGF in the absence of cells

resulted in the greatest improvement in functional recovery with maximum tetanic

contraction forces significantly greater than all groups besides keratin alone and

KN+M+IGF-1. In addition, histology results indicated that new muscle tissue could be

seen in KN+I+b treated tissue extending from the edge of the native tissue and into the

layer of deposited connective tissue. The results indicated that the number of cells used did

not improve functional recovery or attenuate the immune response more effectively than

acellular keratin groups. These findings showed keratin hydrogels were capable of

enhancing functional and morphological recovery in a mouse skeletal muscle injury model

and encouraged the further exploration of keratin biomaterials to treat VML.

244
Chapter 3 presented the application of same blend of keratin hydrogel coupled with

growth factors and cells used in chapter 2 in a second previously established model of VML

in the rat TA. In the rat TA VML model, functional recovery was measured in vivo at 4, 8,

and 12 weeks post-surgery. TA contraction forces were measured via electrical stimulation

of the peroneal nerve and measurement of subsequent dorsiflexion through a foot plate

force transducer. In this method, force measurements could be taken at several timepoints

in the same animal. Histological analysis was then conducted on tissues removed at 12

weeks after surgery. Overall tissue morphology was examined via hematoxylin and eosin

and Masson’s trichrome staining.

At 8 weeks post-surgery tissues treated with keratin in combination with IGF-1 in

the absence of cells resulted in the greatest improvement in functional recovery with

maximum tetanic contraction forces significantly greater than NR. At 12 weeks post-

surgery, tissues treated with KN+I+b were significantly greater than BAM and NR treated

tissues, but were not different from KN or KN+b treated tissues.

In addition, histology results at 12 weeks indicated that greater evidence of new

muscle tissue formation could be seen in all keratin treated tissues. The results at the 8

week time point (at which cellular groups were included in the study) illustrated that the

number of cells used did not improve functional recovery to a greater extent than the

acellular keratin groups. These findings in the rat TA model provided further evidence of

keratin hydrogels enhancing functional and morphological recovery in a muscle injury and

solidified the basis for further exploration of keratin biomaterials for VML treatment.

TEMR was further investigated for use in VML in Chapter 4 which sought to

evaluate TEMR for use in CL/CP secondary revision procedures by implementing the

245
technology in a newly developed model. The developed injury model consisted of a

rectangular injury in the rat LD which was roughly 4 times larger in weight than the

previously largest injury model used by our group in the rat TA. The unique surgical model

enabled the TEMR constructs to be applied in two layers: one below and above the created

defect in a fashion applicable to CL/CP revision. The created injury was reported to result

in an 80% deficit relative to uninjured control tissues at 2 months post injury in males and

a 70% deficit in females.

Use of the TEMR construct resulted in the highest contraction parameters at 2

months post implant in both males and females, but these findings were not significant

compared with NR and BAM treated tissues and at 4 and 6 months there were also no

significant differences in function between the groups in either sex. Contrary to the

functional data, the macroscopic images and histological findings indicated that NR tissues

did not regenerate and TEMR treatment resulted in greater tissue quality and vascularity

and better attenuated inflammation compared with BAM treated tissues.

Therefore, it was suggested that the number of cells used in this study was

insufficient in improving functional recovery, but the cells likely played a role in improving

tissue quality through either paracrine effects, interaction with invading cells, or a

combination of both. As such the study merited the further exploration of TEMR for use

in treating VML and indicated its potential as a graft material for use in CL/CP secondary

revision procedures.

246
Comparison of Research Findings

Comparing Keratin Hydrogel Application in Two Distinct Rodent Models of VML

Previous work using TEMR showed the technology was capable of significantly

improving functional recovery in two distinct rodent injury models (13, 15, 16). Moving

forward from these successes, the next step in further developing a platform of TE

technologies for VML was to investigate other potential biomaterial scaffolds. Based on

several beneficial characteristics, keratin hydrogels were a strong candidate for

investigation in the existing VML models. Perhaps not surprisingly, keratin hydrogels were

successful in promoting significant improvement in function in both the mouse LD and the

rat TA models.

Key differences in the models make the improvement in function enabled by keratin

in both studies worthy of further consideration. The mouse LD injury resulted in a ~16mg

defect and the rat TA injury resulted in a ~70mg. In the mouse model between and 100-

200µl of hydrogel was injected and similarly in the rat model ~200µl was injected. It could

be argued that for the volume injected in both studies, wound beds were receiving similar

doses of keratin and depending on the group investigated, a similar concentration of growth

factors and/or cells.

It is worth noting, that in both cases the number of cells implanted, 1.2-2.3x105

MPCs, was arguably too low to contribute to functional recovery as much higher

concentrations were used in prior work with success. In a previous study using TEMR in

the rat TA model ~3x106 MPCs were introduced and ~6x106* MPCs were applied in two

previous investigations using TEMR in the mouse LD (* not including additional cell

seeding done during preconditioning for one group in Corona, 2012) (13, 15, 16).

247
In spite of the difference in injury size (rat TA defect was ~4x larger than the mouse

LD defect) the dose of keratin given was effective in both studies, though the timeline of

recovery appeared to be different in the two models. In the mouse model, at 8 weeks post

injury the greatest improvement in function was enabled by keratin in combination with

both growth factors which was significantly greater than all treatment groups besides

keratin alone and keratin combined with cells and IGF-1. At 8 weeks in the rat TA model,

only KN+IGF-1 was significantly greater than NR. However, at 12 weeks post-implant in

the rat model KN+I was no longer different from NR and KN+I+b, keratin alone, and

KN+b were all significantly greater than NR (with deficits approximating data seen at 8

weeks in K+I treated tissues), suggesting that keratin enabled recovery in the rat TA model

took place over a longer time course. In looking at the improvement conferred in each

model, the highest improvement in function was enabled by KN+I+b in both groups. In the

mouse model at 8 weeks, KN+I+b had a 32% functional deficit relative to control tissues

compared with NR tissues which had a 58% deficit. At 12 weeks post-surgery in the rat

model, KN+I+b treated tissues had contraction forces equating to a 29% deficit relative to

control forces whereas NR tissues had a 43% deficit. Therefore, the dose of hydrogel

enabled a 26% improvement over no treatment in the mouse model and a 14%

improvement in the rat model. While a greater improvement in the mouse model may be

expected considering the differences in the defect sizes and resulting relative doses of

hydrogel, these reasons alone are likely not strictly responsible. Other potential factors

might include muscle specific and species specific differences in muscle regeneration rate.

In addition, differences in how the injury models altered the muscle biomechanics could

also play a role in how the injuries healed. As keratin hydrogels have demonstrated a

248
capacity to improve muscle function in two distinct models, the ongoing investigation of

keratin for treating VML, as well as investigation into the considerations posed here, is

encouraged.

Comparing Keratin Hydrogels and TEMR Applied in Rodent Models of VML

In working towards developing a platform of technologies for treating VML

injuries our group has also developed, and is continuing to develop, several in vivo VML

injury models. The collection of these animal models aims to address a variety of potential

injuries and represents a diversity of wound geometries, muscle anatomies, and relative

sizes. In the set of experiments covered by this dissertation, three different models were

employed in studying two different natural biomaterial scaffolds. Previously, the

application of cell and growth factor loaded keratin hydrogels in two different rodent

models was compared. This discussion will consider the use of cells and how the materials

interfaced with the injured tissues in the studies.

In all three studies, the cell density applied in the various injury models was

discussed and considered to be suboptimal. For example, and as previously stated, in the

mouse LD study of keratin hydrogels the number of cells delivered was 26-50 times lower

than that used in previous studies employing TEMR in the mouse LD. Also previously

mentioned was that in the rat LD model for an injury 11-15 times greater in defect weight

than that in the mouse LD, only 3 times as many cells were applied when comparing

numbers from the previous TEMR studies (using the mouse LD model). While both sets

of studies indicated an insufficient number of cells were used, it should be noted that the

determined discrepancy was much greater between the keratin studies and previous work

249
than between the study of TEMR in the rat LD and previous work: 25-50x versus 3-5x

fewer cells respectively. In addition, and potentially more importantly, the cells presented

in the rat LD model were subjected to chemical and physical differentiation cues whereas

the cells delivered in the keratin studies were early passage non-fused muscle derived cells.

The considerable difference in cell number (and arguably as a result of the differences in

cell phenotype) may have caused the differences observed in histology for cellular and

non-cellular groups between the studies. TEMR appeared to improve tissue quality over

BAM whereas cellularized keratin hydrogels did not in comparison with acellular keratin

groups.

Nonetheless, the concentrations of cells used in all three studies were still unable

to enable significant functional improvement whereas growth factor loaded keratin

hydrogels alone were. Which brings to attention the consideration of how the gels

interacted with the surrounding tissue to result in improved function versus application of

BAM scaffolds. As stated repeatedly, it is hypothesized that the cells delivered on the

TEMR scaffold in the rat LD were too few to result in functional improvement relative to

BAM, but there was also no significant difference between BAM, TEMR, and NR. This

suggests that the presence of the scaffolds alone in this study was not capable of providing

a sufficient tissue bridge between non-injured tissues to conduct force transmission and

improve contraction forces over NR, a finding which has been termed functional fibrosis

(25). Interestingly, in the keratin studies BAM treated tissues actually had lower

contraction forces than NR treated tissues. Potentially this is a result of the volume of

scaffold used. Perhaps there was too little scaffold used in the rat LD defect for any

250
functional fibrosis to occur and in the mouse LD and rat TA models, in the absence of cells,

the volume of BAM used was actually detrimental to recovery and regeneration of tissue.

While suture retaining ECM scaffolds are intrinsically capable of achieving

functional fibrosis, injectable keratin hydrogels are not. The significant functional

improvement conferred by the hydrogels was not a result of the remodeling of a physical

bridge as occurs in ECM scaffolds, but instead must be attested to the interaction of the gel

with tissue. Based on previous studies, several mechanisms could be possible. Potentially,

the hydrogels promoted a more favorable environment for regeneration by influencing the

conversion of invading monocytes toward an anti-inflammatory phenotype (26). Or,

perhaps the keratin hydrogels recruited invading blood borne BMMSCs which then

differentiated toward muscle and aided in regeneration (5-7, 27-30) The mechanism

surrounding this interaction was not explicitly explored in either keratin study presented,

but certainly deserves further investigation as keratin has shown a great potential for use

in treating VML.

Suggested Future Research

In summarizing and thoughtfully comparing the findings presented in this

dissertation, serval avenues of future work have been brought to attention. Naturally, the

following suggested efforts are not exhaustive; instead the outlined studies strive to address

major limitations and to present studies which would clearly and directly further this area

of research. Several future studies are described briefly below: optimization of keratin

hydrogel cellularity, in vivo examination of dose-response to selected growth factors in a

muscle injury, assessment of mechanical properties of scaffolds alone and in series with

251
muscle, and examination of the effect of an additional cell seeding or the use of additional

scaffold layers in the rat LD model.

Optimization of Keratin Hydrogel Cellularity

The major limitation of the keratin studies presented in chapters 2 and 3 was the

cellularity of the constructs applied in the VML models. The blend of keratose and

kerateine employed in the two studies was developed under constraints surrounding

degradation and accompanying growth factor release time. A 70:30 blend of 15% KOS and

7% KTN resulted in sustained release of both IGF-1 and bFGF over the course of 4 weeks

(Appendix 3). However, this blend was not capable of carrying more than 1.7x106 rodent

MPCs in the KOS fraction due to viscosity becoming too high for extrusion of the gel

through a large gauge needle (16-19 gauge surgical needles). Work submitted for

publication showed 12% KOS and 8% KTN gels had elastic moduli measured to be

~100Pa, which was associated with gels that were flowable in nature. Since the gels

described in that study had similar w/v KOS and KTN values to those used in this research

and that all of the formulations were delivered via syringe in an in vivo model it is assumed

here that the blend used in these studies was also roughly ~100Pa and that increasing the

number of cells would cause gels to exceed this target elastic moduli (data not shown from

Tomblyn et al, unpublished results).

In order to overcome the viscosity issue and improve the cellularity, the KOS:KTN

blend would need to be optimized in order to maintain the release profile of IGF-1 and

bFGF and the desired elastic moduli of ~100Pa while increasing the cellularity. Methods

necessary to optimize these parameters include culture and expansion of rodent MPCS,

252
testing the elastic moduli of the gels with a rheometer, and conducting ELISA on solutions

surrounded growth factor loaded gels to determine percent release of each factor. To plan

optimizing the w/v of KOS and KTN and ratio of each used, one might begin by examining

lower concentrations of keratin in the KOS blend, which holds the cells, and examining

subsequent increases and decreased in KTN w/v and within each group determining the

maximum cell capacity without exceeding 100Pa as shown in Table I.

The expected outcome from this proposed optimization study would be

identification of a keratin blend which allows a greater cell concentration while

maintaining desirable release rates of IGF-1 and bFGF and striving to maintain an elastic

moduli near 100Pa.

70% KOS 30% KTN 30% KTN 30% KTN


Fraction Fraction Fraction Fraction
Control
15 5 7 9
(% KN w/v)
Test set 1
12 5 7 9
(% KN w/v)
Test set 2
10 5 7 9
(% KN w/v)
Test set 3
8 5 7 9
(% KN w/v)

Table I. Experimental Design for Optimizing Keratin Hydrogel Cellularity

In vivo ‘Dose Response’ to IGF-1 and bFGF Loaded Keratin Hydrogels in VML

The concentration of growth factors administered in the studies presented in

chapters 2 and 3 were based on work wherein the goal was to showcase the ability of keratin

hydrogels to deliver both cells and growth factors, but concentrations were not selected

based on previous work or data suggesting optimal concentrations (data from manuscript

253
under review, Tomblyn et al). The chosen concentrations (100ug/mL of hydrogel or in the

dissertation research studies ~10ug/injection in the mouse LD and ~25ug/injection in the

rat TA) were efficacious in combination with VEGF in causing increased vascular

structures and were attributed to promoting viable muscle tissue. Vascular structures could

be seen within the cell loaded gel and cells in the region stained positive for intermediate

filament desmin at 4 weeks after implant into a subcutaneous dorsal pouch in a mouse (data

not shown, summarized from manuscript under review by Tomblyn et al). In the present

studies, IGF-1 and bFGF administered alone in keratin hydrogels did not improve muscle

function or result in new muscle tissue formation, but when combined (in the absence of

cells) the mixture supported significantly improved function in both a mouse LD injury

and a rat TA injury (chapters 2 and 3 respectively). While these results are promising, a

future study might aim to better understand the interaction between the two growth factors

and within a muscle injury. Specifically a study of long term functional ‘dose-response’ to

varied concentrations of the growth factors may shed light on the complex interaction

between growth factors, regenerating tissue, and surrounding and invading cell

populations. Such an endeavor is certainly daunting, but ultimately if growth factors were

deemed necessary through further research more (and much more complex) studies would

need to be undertaken prior to clinical translation.

The methods involved with pursuing this study include: rodent VML surgery and

associated functional testing to examine resulting functional recovery and histology and

immunohistochemistry to examine cell and tissue morphology with specific consideration

given to identification of immune cell congregation and inflammation, new muscle tissue

presence, vascularization, and scar tissue deposition. In order to glean the most information

254
from the study, it is suggested that the rat TA model of VML be used in order to measure

functional recovery relatively non-invasively with the option of measuring at multiple time

points prior to termination and so that the tissue tested for function may also be used for

histology (unlike the mouse LD model wherein it is preferable to use different tissues for

function and histology as the organ bath tested tissues tend to be of poorer quality for

histology). To simplify a study examining dose-response to growth factors one might wish

to only investigate 10 fold or 5 fold changes in concentrations perhaps as outlined in Table

II. As there might be effects due to both concentration and ratio of the growth factors used

it will be important to include combinations wherein the growth factors are both the same

concentration and the ratio is the same between test sets (i.e. 10µg/ml IGF-1:10µg/ml bFGF

would be the same ratio as that used in chapters 2 and 3, 1:1; 1µg/ml IGF-1:10µg/ml bFGF

and 10 µg/ml IGF-1:100 µg/ml bFGF would be looking at a 1:10 ratio in both cases).

As the interaction of growth factors and their effects on remodeling tissue is

difficult to predict, it is also difficult to propose expected outcomes from this study. Based

on histologic findings from chapters 2 and 3 is seems likely that increased adipose and scar

tissue deposition will be found in groups with higher IGF-1 and bFGF concentrations

respectively. Further, varied concentrations might result entirely different results as it could

alter the interaction between the growth factors. Previous work has shown that in the

presence of bFGF, IGF-1 will behave like a mitogen instead of promoting differentiation

and fusion in muscle- a relationship which this proposed work may determine to be

concentration dependent (21).

255
IGF-1 bFGF bFGF bFGF
Control Test Set
100 500 100 10
(µg factor/ml gel)
Test set 1
10 500 100 10
(µg factor/ml gel)
Test set 3
500 500 100 10
(µg factor/ml gel)

Table II. Experimental Groups for In Vivo Keratin Hydrogel Coupled

Growth Factor Dose Response Experiment

Investigation of Mechanical Properties of Injured and Treated Tissues

Throughout the discussion of the effectiveness of keratin hydrogels and TEMR

constructs in various models of VML in studies previous to this dissertation and within,

there have been several mentions of mechanical interactions between a scaffold and the

wound environment it is applied to. With ECM based scaffolds such as BAM and TEMR,

the material is flexible, but strong enough to hold sutures which anchor the constructs in

place within a wound bed. However, for keratin hydrogels and other materials which

cannot retain sutures, the physical anchoring of the test article cannot serve to bridge one

end of a muscle injury to the other and potentially enabling transmission of contraction and

forces. Further, bridging the gap or enabling force transmission in muscle injuries has been

suggested to protect the surrounding muscle by shielding it from excess strain which could

result in degeneration of tissue (15, 31, 32). In addition to the immediate physical bridge

offered by certain materials, some may result in improvement in the mechanical integrity

of regenerating tissue or remodeling scaffold by interacting with the surrounding tissue via

256
the addition of a cellular component or deliverable substances such as growth factors or

drugs.

To investigate the impact on mechanical properties as a result of VML and

treatment with various scaffolds, mechanical testing of a variety of different materials

could be conducted in several settings. The test parameters of interest might include elastic

modulus and failure strength. Uniaxial tensile testing could be carried out on scaffolds

before and after cell seeding and preconditioning, on muscle strips in series with constructs

immediately after surgical attachment or after being in in vivo, or on strips of muscle in the

vicinity of the defect. An example of a list of groups for testing in shown in Table III.

Methods used to compare the mechanical properties of the scaffolds alone and after

implantation in vivo include cell culture and cyclic stretch bioreactor assembly and

programming, rodent surgery, and use of a mechanical testing system and interpretation of

the resulting data. It should be noted that as muscle is viscoelastic, a set strain rate should

be employed for all tests. Based on previous studies, the expected outcomes of such a study

would be that the cell seeded constructs have a greater stiffness than in non-cell seeded

constructs more closely resembling muscle strips and that the failure strength in cell seeded

constructs in series with tissue is improved relative to BAM in series with muscle just after

implant and after 4 weeks in vivo (33). Studying the mechanical properties of the scaffolds

alone and in situ would help determine how the materials contribute to functional recovery

and may aid in further development of TE therapies for VML.

257
Table III. Experimental Groups for Mechanical Testing of Test Articles in

Series with Rat LD Muscle.

258
Investigation into Laminate TEMR and Multi-Seeded TEMR in Rat LD Model

Lastly, another potential set of studies which may serve to further the research

presented in this dissertation involves additional exploration of TEMR in the rat LD model.

Specifically, it was discussed that the use of TEMR in the large injury model developed in

the rat LD might benefit from an increased cell number. Two such methods of improving

the cell number delivered to the injury site would be to add additional layers of TEMR

(potentially 4 instead of 2 as used previously) or to increase the cell number on each TEMR

construct. The introduction of additional scaffold layers stands to benefit the current model

as more scaffold material may stand to improve force transduction through functional

fibrosis alone- even without a cellular component. Previous work indicated that the

addition of a cell seeding during preconditioning improves functional recovery (16).

As such, these two strategies could be employed separately and together in the rat

LD model in an effort to improve functional recovery. Potential treatment groups for use

in a 2-6 month study are shown in Table IV. Methods involved with this study include cell

culture, assembly and programming of a cyclic stretch bioreactor, rodent surgery, in vitro

functional testing using the organ bath set-up, and immunohistochemistry.

Expected outcomes would be that the addition of TEMR layers results in a longer

scaffold degradation time, serves to improve functional recovery, and tissue quality

compared with single layered TEMR constructs. Further it would be expected that multi-

seeded TEMR results in improved functional recovery and correlated improved tissue

morphology including new muscle tissue formation.

259
Number of Number of Additional
2 Mos. Groups animals (n) Scaffolds (n) MPCs added
TEMR 6 2 no
Double TEMR 6 4 no
TEMR+MPC 6 2 yes
Double
6 4 yes
TEMR+MPC

Table IV. Experimental Groups for Study of Laminate and Multi-Seeded

TEMR

Clinical Translation Considerations

Keratin hydrogels and TEMR have both been shown to have promise for use as a

VML treatment. Conventions within the FDA regulatory processes and agencies have been

historically centered on drugs, medical devices, and biologics alone, however over the last

two decades those processes have had to evolve, and continue to evolve, to accommodate

TE products in which combinations of cells, dugs, and devices are used (34-37).

On top of being a combination therapy, both products face unique challenges prior

to and after submission of an IND in clinical trials. The scaffold material in the products,

the keratin hydrogel alone and the BAM, will need to be manufactured according to Good

Manufacturing Process (GMP) guidelines under which elimination of batch to batch

variability, which is an innate obstacle for naturally derived scaffolds, will need to be

ensured. For keratin hydrogels, streamlined manufacturing is already in use for other

keratin products via KeraNetics, LLC. Manufacturing with GMP for TEMR will also have

to accommodate cell seeding of scaffolds and the use of the cyclic stretch bioreactor which

poses a unique challenge in itself by adding an enabling technology into the process.

260
As part of the IND application, both products must demonstrate non-toxicity

through animal studies. Though these combinations have not been through the vetting

process yet, many of their components have. The individual constituents to consider are

MPCs, BAM, keratin hydrogel, and the administration of growth factors. Clinical use of

allogeneic multipotent stem cells such as BMMSCs have indicated their safety in several

applications and research studies using further differentiated adult stem cells, like MPCs,

have also demonstrated safety (38-40). Many ECM scaffold based materials have gone

through clinical trials successfully and examples of clinically available products include

patches for wound healing, rotator cuff repair, and hernia repair (9, 41). Similarly, a keratin

hydrogel product has also obtained IND status and was submitted for use in a clinical trial

(42, 43).

The remaining component is the application of growth factors. As previously

mentioned in the discussion of potential future studies, the application of growth factors in

TE research is inherently complex. Natural growth factor production and regulation is

dynamic and intricate and through the work of many investigators there have been great

strides made in understanding these mechanisms and their application in tissue engineering

(21, 23, 44-47). The addition of extrinsic factors can have undesirable effects as was seen

in high dose BMP-2 releasing INFUSE devices applied in the cervical spine which led to

ectopic bone formation in the trachea- which was believed to be a result of too high a

concentration of BMP-2 (48). As such, further studies must be carried out in order to

determine safe and effective doses and release rates of growth factors from keratin

hydrogels prior to an IND submission surrounding their combination.

261
Conclusion

The culmination of the work presented in this dissertation has furthered the overall

goal of developing a platform of technologies capable of treating VML. This dissertation

entailed the first time application of cell and growth factor loaded keratin hydrogels in a

VML model and a novel application of TEMR in a newly developed VML model with

relevance to cleft lip secondary revision procedures. In these studies keratin hydrogels and

TEMR have shown promise for clinical translation for treatment of muscle injuries. As

such, future studies which seek to further this research and bring these therapies closer to

translation are well warranted.

262
Literature Cited

1. Juhas, M., and Bursac, N. Engineering skeletal muscle repair. Current Opinion in

Biotechnology 24, 880, 2013.

2. Mikos, A.G., Herring, S.W., Ochareon, P., Elisseeff, J., Lu, H.H., Kandel, R., Schoen,

F.J., Toner, M., Mooney, D., Atala, A., Van Dyke, M.E., Kaplan, D., and Vunjak-

Novakovic, G. Engineering complex tissues. Tissue Engineering 12, 3307, 2006.

3. Ratner, B. Biomaterials Science: An Introduction to Materials in Medicine. 2nd ed.

San Diego, CA: Elsevier Academic Press; 2004.

4. Montemurro, F., Gattazzo, F., De Maria, C., and Vozzi, G. Keratin based hydrogels

for tissue engineering and regenerative medicine. Journal of Tissue Engineering and

Regenerative Medicine 8, 357, 2014.

5. Richter, J.R., de Guzman, R.C., Greengauz-Roberts, O.K., and Van Dyke, M.

Structure-property relationships of meta-kerateine biomaterials derived from human hair.

Acta Biomaterialia 8, 274, 2012.

6. Hill, P., Brantley, H., and Van Dyke, M. Some properties of keratin biomaterials:

Kerateines. Biomaterials 31, 585, 2010.

7. Rouse, J.G., and Van Dyke, M.E. A Review of Keratin-Based Biomaterials for

Biomedical Applications. Materials 3, 999, 2010.

8. Verma, V., Verma, P., Ray, P., and Ray, A.R. Preparation of scaffolds from human

hair proteins for tissue-engineering applications. Biomedical Materials 3, 12, 2008.

9. Brown, B.N., and Badylak, S.F. Extracellular matrix as an inductive scaffold for

functional tissue reconstruction. Translational Research 163, 268, 2014.

263
10. Peyton, C.C., Keys, T., Tomblyn, S., Burmeister, D., Beumer, J.H., Holleran, J.L.,

Sirintrapun, J., Washburn, S., and Hodges, S.J. Halofuginone infused keratin hydrogel

attenuates adhesions in a rodent cecal abrasion model. Journal of Surgical Research 178,

545, 2012.

11. Saul, J.M., Ellenburg, M.D., de Guzman, R.C., and Van Dyke, M. Keratin hydrogels

support the sustained release of bioactive ciprofloxacin. Journal of Biomedical Materials

Research Part A 98A, 544, 2011.

12. de Guzman, R.C., Saul, J.M., Ellenburg, M.D., Merrill, M.R., Coan, H.B., Smith,

T.L., and Van Dyke, M.E. Bone regeneration with BMP-2 delivered from keratose

scaffolds. Biomaterials 34, 1644, 2013.

13. Machingal, M.A., Corona, B.T., Walters, T.J., Kesireddy, V., Koval, C.N.,

Dannahower, A., Zhao, W.X., Yoo, J.J., and Christ, G.J. A Tissue-Engineered Muscle

Repair Construct for Functional Restoration of an Irrecoverable Muscle Injury in a

Murine Model. Tissue Engineering Part A 17, 2291, 2011.

14. Moon, D.G., Christ, G., Stitzel, J.D., Atala, A., and Yoo, J.J. Cyclic mechanical

preconditioning improves engineered muscle contraction. Tissue Engineering Part A 14,

473, 2008.

15. Corona, B.T., Ward, C.L., Baker, H.B., Walters, T.J., and Christ, G.J. Implantation

of In Vitro Tissue Engineered Muscle Repair Constructs and Bladder Acellular Matrices

Partially Restore In Vivo Skeletal Muscle Function in a Rat Model of Volumetric Muscle

Loss Injury. Tissue Engineering Part A 20, 705, 2014.

16. Corona, B.T., Machingal, M.A., Criswell, T., Vadhavkar, M., Dannahower, A.C.,

Bergman, C., Zhao, W.X., and Christ, G.J. Further Development of a Tissue Engineered

264
Muscle Repair Construct In Vitro for Enhanced Functional Recovery Following

Implantation In Vivo in a Murine Model of Volumetric Muscle Loss Injury. Tissue

Engineering Part A 18, 1213, 2012.

17. Farhat, W.A., Chen, J., Haig, J., Antoon, R., Litman, J., Sherman, C., Derwin, K.,

and Yeger, H. Porcine bladder acellular matrix (ACM): protein expression, mechanical

properties. Biomedical Materials 3, 8, 2008.

18. Rosario, D.J., Reilly, G.C., Salah, E.A., Glover, M., Bullock, A.J., and MacNeil, S.

Decellularization and sterilization of porcine urinary bladder matrix for tissue

engineering in the lower urinary tract. Regenerative Medicine 3, 145, 2008.

19. Farhat, W., Chen, J., Erdejan, P., Shemtov, O., Courtman, D., Khoury, A., and

Yeger, H. Porosity of porcine bladder acellular matrix: Impact of ACM thickness. Journal

of Biomedical Materials Research Part A 67A, 970, 2003.

20. Dahms, S.E., Piechota, H.J., Dahiya, R., Lue, T.F., and Tanagho, E.A. Composition

and biomechanical properties of the bladder acellular matrix graft: comparative analysis

in rat, pig and human. British Journal of Urology 82, 411, 1998.

21. Allen, R.E., and Boxhorn, L.K. REGULATION OF SKELETAL-MUSCLE

SATELLITE CELL-PROLIFERATION AND DIFFERENTIATION BY

TRANSFORMING GROWTH FACTOR-BETA, INSULIN-LIKE GROWTH FACTOR-

I, AND FIBROBLAST GROWTH-FACTOR. Journal of Cellular Physiology 138, 311,

1989.

22. Florini, J.R., and Magri, K.A. EFFECTS OF GROWTH-FACTORS ON

MYOGENIC DIFFERENTIATION. American Journal of Physiology 256, C701, 1989.

265
23. Florini, J.R., Ewton, D.Z., and Coolican, S.A. Growth hormone and the insulin-like

growth factor system in myogenesis. Endocrine Reviews 17, 481, 1996.

24. Hayashi, S., Aso, H., Watanabe, K., Nara, H., Rose, M.T., Ohwada, S., and

Yamaguchi, T. Sequence of IGF-I, IGF-II, and HGF expression in regenerating skeletal

muscle. Histochemistry and Cell Biology 122, 427, 2004.

25. Corona, B.T., Wu, X., Ward, C.L., McDaniel, J.S., Rathbone, C.R., and Walters, T.J.

The promotion of a functional fibrosis in skeletal muscle with volumetric muscle loss

injury following the transplantation of muscle-ECM. Biomaterials 34, 3324, 2013.

26. Fearing, B.V., and Van Dyke, M.E. In vitro response of macrophage polarization to a

keratin biomaterial. Acta Biomaterialia 10, 3136, 2014.

27. Sierpinski, P., Apel, P.J., Tawfik, B., Smith, T., Koman, L.A., Atala, A., and Van

Dyke, M. Keratin Biomaterials Activate Schwann Cells via Integrin-Mediated Signaling

and Promote Regeneration of Large Peripheral Nerve Defects in a Rabbit Model. Faseb

Journal 23, 1, 2009.

28. de Guzman, R.C., Merrill, M.R., Richter, J.R., Hamzi, R.I., Greengauz-Roberts,

O.K., and Van Dyke, M.E. Mechanical and biological properties of keratose biomaterials.

Biomaterials 32, 8205, 2011.

29. Fong, E.L.S., Chan, C.K., and Goodman, S.B. Stem cell homing in musculoskeletal

injury. Biomaterials 322011.

30. Ten Broek, R.W., Grefte, S., and Von den Hoff, J.W. Regulatory Factors and Cell

Populations Involved in Skeletal Muscle Regeneration. Journal of Cellular Physiology

224, 7, 2010.

266
31. Stauber, W.T., Knack, K.K., Miller, G.R., and Grimmett, J.G. Fibrosis and

intercellular collagen connections from four weeks of muscle strains. Muscle & Nerve

19, 423, 1996.

32. Stauber, W.T., Smith, C.A., Miller, G.R., and Stauber, F.D. Recovery from 6 weeks

of repeated strain injury to rat soleus muscles. Muscle & Nerve 23, 1819, 2000.

33. Androjna, C., Spragg, R.K., and Derwin, K.A. Mechanical conditioning of cell-

seeded small intestine submucosa: A potential tissue-engineering strategy for tendon

repair. Tissue Engineering 13, 233, 2007.

34. Levine, D.W., Mondano, L., and Halpin, M. FDA Regulatory Pathways for Knee

Cartilage Repair Products. Sports Medicine and Arthroscopy Review 16, 202, 2008.

35. Halme, D.G., andKessler, D.A. FDA regulation of stem-cell-based therapies. New

England Journal of Medicine 355, 1730, 2006.

36. Hamrell, M.R. An overview of the US regulatory environment for drug-device and

biologic-device combination products. Drug Information Journal 40, 23, 2006.

37. Chapekar, M.S. Regulatory concerns in the development of biologic-biomaterial

combinations. Journal of Biomedical Materials Research 33, 199, 1996.

38. Bianco, P., Riminucci, M., Gronthos, S., and Robey, P.G. Bone marrow stromal stem

cells: Nature, biology, and potential applications. Stem Cells 19, 180, 2001.

39. Robey, P.G., and Bianco, P. The use of adult stem cells in rebuilding the human face.

Journal of the American Dental Association 137, 961, 2006.

40. Bianco, P., Robey, P.G., and Simmons, P.J. Mesenchymal stem cells: Revisiting

history, concepts, and assays. Cell Stem Cell 2, 313, 2008.

267
41. Health, U.S.N.I.o. Clinical Trials Involving Small Intestine Submucosa. In: Services

H.a.H., ed. Washington D.C.: U.S. National Institutes of Health; 2011.

42. Burnett, L.R., Richter, J.G., Rahmany, M.B., Soler, R., Steen, J.A., Orlando, G.,

Abouswareb, T., and Van Dyke, M.E. Novel keratin (KeraStat (TM)) and polyurethane

(Nanosan (R)-Sorb) biomaterials are hemostatic in a porcine lethal extremity hemorrhage

model. Journal of Biomaterials Applications 28, 869, 2014.

43. National Cancer Institute (NCI), C.C.C.o.W.F.U. KeraStat Skin Therapy in Treating

Radiation Dermatitis in Patients With Newly Diagnosed Stage 0-IIIA Breast Cancer.

ClinicalTrialsgov [Internet]. 2014 Nov 28 ed. Bethesda (MD): National Library of

Medicine (US); 2014.

44. Yamada, M., Tatsumi, R., Kikuiri, T., Okamoto, S., Nonoshita, S., Mizunoya, W.,

Ikeuchi, Y., Shimokawa, H., Sunagawa, K., and Allen, R.E. Matrix metalloproteinases

are involved in mechanical stretch-induced activation of skeletal muscle satellite cells.

Muscle & Nerve 34, 313, 2006.

45. Calve, S., and Simon, H.G. Biochemical and mechanical environment cooperatively

regulate skeletal muscle regeneration. Faseb Journal 26, 2538, 2012.

46. Biondi, M., Ungaro, F., Quaglia, F., and Netti, P.A. Controlled drug delivery in

tissue engineering. Advanced Drug Delivery Reviews 60, 229, 2008.

47. Ladewig, K. Drug delivery in soft tissue engineering. Expert Opinion on Drug

Delivery 8, 1175, 2011.

48. Shields, L.B.E., Raque, G.H., Glassman, S.D., Campbell, M., Vitaz, T., Harpring, J.,

and Shields, C.B. Adverse effects associated with high-dose recombinant human bone

morphogenetic protein-2 use in anterior cervical spine fusion. Spine 31, 542, 2006.

268
APPENDIX 1

Implantation of in vitro tissue engineered muscle repair (TEMR) constructs and bladder

acellular matrices (BAM) partially restore in vivo skeletal muscle function in a rat model

of volumetric muscle loss (VML) injury

Corona, B.T.1,2, Ward, C.L.1,2, Baker, H.B.1, Walters, T.J.2, & Christ, G.J.1

Affiliations:

1
Wake Forest Institute for Regenerative Medicine, Winston Salem, NC

2
United States Army Institute of Surgical Research, Department of Extremity Trauma and

Regenerative Medicine, Fort Sam Houston, TX

Note: this manuscript has been published and is being used with permission

269
Acknowledgements: We would like to thank Ms. Manasi Vadhavkar for her technical

assistance during surgery and functional testing and Mr. Christopher Bergman for his

technical assistance with histological procedures. The opinions or assertions contained

herein are the private views of the authors and are not to be construed as official or

reflecting the views of the Department of Defense or the United States Government. The

authors (btc, clw, and tjw) are employees of the U.S. government and this work was

prepared as part of their official duties. This work was supported in part by the Armed

Forces Institute for Regenerative Medicine (W81XWH-08-2-0032) and by the U.S. Army

Medical Research and Medical Command (W81XWH-09-2-0177).

Corresponding Author:

George J. Christ, Ph.D.

Professor of Regenerative Medicine

Wake Forest Institute for Regenerative Medicine

Wake Forest University Baptist Medical Center

Richard H. Dean Biomedical Research Building, Room 257

391 Technology Way

Winston-Salem, NC 27101

Ph: 336-713-7296

Fax: 336-713-7290

Email: gchrist@wakehealth.edu

270
Abstract

The frank loss of a large volume of skeletal muscle (i.e., volumetric muscle loss;

VML) can lead to functional debilitation and presents a significant problem to civilian and

military medicine. Current clinical treatment for VML involves the use of free muscle flaps

and physical rehabilitation; however, neither are effective in promoting regeneration of

skeletal muscle to replace the tissue that was lost. Towards this end, skeletal muscle tissue

engineering therapies have recently shown great promise in offering an unprecedented

treatment option for VML. In the current study, we further extend our recent progress

(Machingal et al., 2011, Tissue Eng; Corona et al., 2012, Tissue Eng) in the development

of tissue engineered muscle repair (TEMR) constructs (i.e., muscle derived cells (MDCs)

seeded on a bladder acellular matrix (BAM) preconditioned with uniaxial mechanical

strain) for the treatment of VML. TEMR constructs were implanted into a VML defect in

a tibialis anterior muscle of Lewis rats and observed out to 12 weeks post-injury. The

salient findings of the study were: 1) TEMR constructs exhibited a highly variable capacity

to restore in vivo function of injured TA muscles, wherein TEMR positive responders (n =

6) promoted a ≈61% improvement but negative responders (n = 7) resulted in no

improvement compared to non-repaired controls, 2) TEMR positive and negative

responders exhibited differential immune responses that may underlie these variant

responses, 3) BAM scaffolds (n = 7) without cells promoted an ≈26% functional

improvement compared to uninjured muscles, 4) TEMR positive responders promoted

muscle fiber regeneration within the initial defect area while BAM scaffolds did so only

sparingly. These findings indicate that TEMR constructs can improve the in vivo functional

capacity of the injured musculature at least in part by promoting generation of functional

271
skeletal muscle fibers. In short, the degree of functional recovery observed following

TEMR implantation (BAM + MDCs) was 2.3X-fold greater than that observed following

implantation of BAM alone. As such, this study confirms and extends prior work to further

underscore the utility of including a cellular component in the tissue engineering strategy

for VML injury.

Introduction

Craniofacial and extremity soft tissue trauma resulting in the permanent loss of

skeletal muscle mass is a clinical challenge for both military and civilian medicine. This

type of injury has been termed volumetric muscle loss (VML) and is defined as “the

traumatic or surgical loss of skeletal muscle with resultant functional impairment.”1 The

current standard of care for VML injury involves the use of free muscle transfer (i.e.,

muscle flaps) followed by extensive physical rehabilitation. However, free muscle flaps do

not significantly restore muscle function to the injured tissue, and are often primarily used

for bony coverage. Functional muscle transfer, in which vasculature and innervation are

surgically restored to the graft have been shown to improve strength to an injured muscle

group2 but these procedures require an extraordinary level of surgical expertise that is not

likely available at most treatment centers. In either case, the use of large muscle flaps

presents an added complication of donor site morbidity to the patient.3 Lastly, while there

is little evidence that physical rehabilitation alone is sufficient to significantly restore

strength to muscle with VML injury, physical rehabilitation performed with an energy-

storing ankle-foot orthosis has recently proven to be successful in restoring function to

patients with extremity trauma.4

272
In this scenario, skeletal muscle tissue engineering and regenerative medicine

therapies offer a promising treatment paradigm for severe soft tissue trauma resulting in

the loss of a large volume of musculature. Therapeutic strategies are under development

for the repair of VML to craniofacial and extremity muscles, and similar injury conditions

(e.g., abdominal wall repair).5-15 Serving as a foundation for many of these strategies

biological extracellular matrices (ECMs) derived from the intestine and bladder (porcine),

and skeletal muscle (variety of species) have been studied in in vivo preclinical skeletal

muscle injury models previously.7, 8, 10, 12, 15, 16


Most studies have shown limited

regeneration of muscle fibers after transplantation (e.g.,8, 9, 15, 17). The only clinical report

currently available describes the use of an acellular matrix for the treatment of quadriceps

muscle with VML injury.18 While these initial findings are encouraging for the overall

tissue engineering paradigm, only somewhat minor strength gains were observed with

continued physical therapy.18

As an extension of this approach, the therapeutic benefit of a combined

transplantation of biological scaffold with stem or progenitor cells has also been

characterized thoroughly in preclinical studies in vivo.5, 6, 8-10, 19-21


The rationale for

inclusion of a cellular component to skeletal muscle tissue engineering therapies is based

on the numerous beneficial effects that have been reported. For example, depending on the

cell type used, transplanted cells can improve revascularization, modulate the

inflammatory response, reduce fibrosis, or contribute directly to muscle fiber

regeneration.22-25 In fact, the combination of bone marrow mesenchymal stem cells with

rat muscle extracellular matrix,9, 10 freshly isolated satellite cells with hyaluronan-based

gel,21 and muscle derived progenitor cells with bladder acellular matrix8 have all

273
demonstrated partial restoration of force production following implantation in surgically-

created VML injuries. In all cases, the observed degree of functional muscle repair and

regeneration was superior to the delivery of the respective matrix alone, providing clear

evidence for the therapeutic benefit of including a cellular component to tissue engineering

strategies designed for the treatment of VML.

In this regard, our recent efforts have exploited this latter strategy and focused on

development of an in vitro method to create tissue engineered muscle repair (TEMR)

constructs for the treatment of skeletal muscle tissue following traumatic injury.5, 8, 26 To

date, we have demonstrated that TEMR constructs, which have been created by seeding

Lewis rat muscle derived cells (MDCs) on a bladder acellular matrix (BAM), mediate

restoration of significant functional capacity (60-70% recovery) to VML injured latissimus

dorsi (LD) muscle in nude mice.5, 8 This recovery appears to involve, at least to some

extent, regeneration of a portion of the muscle fibers that were surgically removed. In

support of this supposition, we have observed presumptive formation (regeneration) of new

muscle fibers in the area of the defect where the construct was transplanted, and

furthermore, noted that these regenerating fibers express key proteins required for force

production and transmission, as well as supporting vascular and neural structures.5, 8

Herein, we report our most recent progress in developing TEMR constructs for

eventual clinical use. In the current study, the therapeutic potential of TEMR constructs

was tested in a Lewis rat tibialis anterior (TA) muscle VML model. The use of the model

introduced three important advances relative to our previous studies of the repair of VML

injury: 1) the muscle defect is approximately four-fold greater by weight in the female

Lewis rat TA muscle than the nude mouse LD muscle, 2) the Lewis rat has a competent

274
immune system, and 3) functional analysis is performed in vivo with neural stimulation

nominally requiring regenerating muscle fibers to be innervated to contribute to functional

recovery. The results of this study indicate that TEMR constructs as well as BAM scaffolds

can improve neural-stimulated in vivo functional recovery following VML injury.

However, there was marked variability in the therapeutic response of TEMR constructs

that appears to be related to a differential inflammatory response within the TEMR

construct population. Discussion is provided to highlight underlying mechanisms for these

findings and future directions for the translation of the TEMR technology.

Methods

Experimental Design: This study was designed to perform repeated in vivo functional

measurements with the same animal (leg) over a three month period using a repeated

measures experimental design. Baseline in vivo functional assessments were performed via

neural stimulation before VML injury was surgically created in the TA muscle and then

every month thereafter for three months. After the last in vivo assessment, TA muscles

were harvested for morphological and histological analysis. Because the extensor hallicus

longus (EHL), extensor digitorum longus (EDL), and TA muscle are innervated by the

common peroneal nerve and the EDL and EHL muscles hypertrophy in response to VML

injury in the TA muscle, in all experimental VML groups the EDL and EHL muscles were

ablated. To account for any potential strength changes of the TA muscle due to synergist

ablation and to provide a theoretical baseline for TA muscle recovery, a group of rats served

as a surgical-sham wherein the synergists were ablated but no VML injury was created in

the TA muscle. In total there were 5 experimental groups in this study: 1) An un-operated

275
group that served as a age-matched cage controls (n = 8), a sham/ablation group in which

the TA muscle did not have a VML defect but underwent sham surgery with ablation of

the EDL muscle (n = 6), a VML injured but non-repaired group (n = 4), a VML injured

with BAM repair group (n = 7), and a VML injured TEMR repaired group (n = 13). All

VML injured groups underwent EDL muscle ablation.

Animals: Male Lewis rats aged 3 – 4 weeks used as donors for muscle derived cells

(MDCs), as previously reported.5, 8 VML injury studies were performed with female Lewis

rats aged 12 – 14 weeks at the beginning of the study. All rats were purchased from Harlan

laboratories. All animal procedures were approved by the Wake Forest University IACUC.

Bladder Acellular Matix (BAM) Preparation: BAM scaffolds were prepared from porcine

urinary bladder as previously described.5, 8 Briefly, the bladder was washed and trimmed

to obtain a crude sample of the lamina propria (i.e., some muscle tissue remaining

attached), which was placed in 0.05% Trypsin (Hyclone, Logan, UT) for one hour at 37°C.

The bladder was then transferred to DMEM solution supplemented with 10% FBS and 1%

Antibiotic/Antimycotic and kept overnight at 4˚C. The preparation was then washed in a

solution containing 1% Triton X (Sigma-Aldrich, St Louis, MO) and 0.1% Ammonium

hydroxide (Fisher Scientific, Fairlown, NJ) in de-ionized water for 4 days at 4°C. Finally,

the bladder was then washed in de-ionized water for 3 days at 4°C. The decellularized

scaffold was then further dissected by hand to remove any remaining muscle tissue layers

to obtain a scaffold with of 0.2-0.4 mm thickness. The prepared acellular matrix was then

cut into strips of 3 cm X 2 cm size and placed onto a custom designed seeding chamber

276
made of silicon (McMaster Carr, Cleveland, OH). Scaffolds and silicon seeding chambers

were then individually placed in culture dishes and sterilized by ethylene oxide.

TEMR Construct Creation: TEMR constructs were generated following previously

described cell culture, seeding, and bioreactor preconditioning protocols.5, 8 Briefly, BAM

scaffolds were seeded with MDCs (≈1 million per cm2) on each side, cultured statically for

10 days allowing for proliferation and differentiation, and then preconditioned in a

bioreactor with uniaxial mechanical strain (10% strain, 3 stretches per minute for the first

five minutes of each hour) as described in detail elsewhere.5, 8

Surgical Procedures: Surgical creation of VML injury was performed in the TA muscle as

previously reported.27 Briefly, a longitudinal skin incision was made along the lateral

aspect of the left lower leg. The skin was separated bluntly from the underlying fascia

covering the anterior compartment. The fascia covering the anterior crural muscles was

then separated using blunt dissection. The proximal and distal tendons of the EDL muscle

were then isolated and severed. Next, the EHL muscle located underneath the TA muscle

was isolated and ablated. Care was taken to avoid disruption of the retinaculum holding

the distal tendon of the TA muscle in position. VML injury was created in the TA muscle

by excising approximately 20% of the TA muscle weight at the middle third of the muscle

(Figure 1C).27 A 20% TA muscle defect was approximated using linear regression

determined experimentally (n = 16) to estimate TA muscle mass from rat body weight: TA

wet weight (mg) = 1.553 x Body Weight (g) + 83.084 (R2 = 0.85).

Repair of the TA muscle was accomplished by folding TEMR or BAM constructs

(≈3 x 0.5 cm) in triplicate creating an ≈1 x 0.5 cm construct with three layers (Figure 1A).

277
The transplant therefore approximated the volume of the defect. The thrice-folded

transplant was placed in the defect and the four corners and four margins were sutured (6-

0 Vicryl) to the borders of the defect (Figure 1D). The fascia was closed with 6-0 vicryl

and the skin was closed with 5-0 prolene using interrupted sutures. Skin glue was applied

between the skin sutures to help prevent the incision from opening. Following surgery,

buprenorphine (0.05 mg/kg; sc) was administered every twelve hours for three days.

In vivo Functional Analysis: Torque production of the left anterior crural muscles

(primarily due to the contraction of the TA and EDL muscles) was measured in vivo using

similar methodology as previously described.27-30 After rats were anesthetized (1.5 – 2.5%

isoflurane), the left hindlimb was aseptically prepared. The rat was then placed on a heated

platform. The left knee was clamped and the left foot was secured to a custom-made foot

plate that is attached to the shaft of an Aurora Scientific 305C-LR-FP servomotor, which

in turn was controlled using a PC. Sterilized percutaneous needle electrodes were inserted

through the skin for stimulation of the left common peroneal nerve. Electrical stimulus was

applied using a Grass S88 stimulator with a constant current SIU (Grass Model PSIU6).

Stimulation voltage and needle electrode placement were optimized first with a series of

twitch contractions at 1 hz and then with 5 to 10 isometric contractions (400 ms train of

0.05 - 0.1 ms pulses at 150 Hz). Contractile function of the anterior crural muscles was

assessed by measuring peak isometric tetanic torque derived from the maximal response to

a range of stimulation frequencies (100 - 200 Hz). For real-time analysis of torque and

length changes, voltage outputs were sampled at 4000 Hz, converted to a digital signal

using an A/D board (National Instruments PCI 6221) and recorded using a PC loaded with

278
a custom-made Labview®-based program (provided by the U.S. Army Institute of Surgical

Research).

Morphological Analysis of VML Injured TA Muscle: TA muscles were isolated from the

leg and laid on a platform with the superior aspect of the muscle facing away from the

platform. At the middle of the proximal and distal thirds of the muscle, the width of the

muscle was measured using digital calipers.

Histology and Immunohistochemistry: TA muscles from all experimental groups were

fixed in 10% neutral buffered formalin and stored in 60% ethanol. All samples were

processed (ASP300S, Leica Microsystems, Bannockburn, IL) and then embedded in

paraffin (EG1160, Leica Microsystems, Bannockburn, IL). Seven µm thick serial sections

were cut from the paraffin embedded blocks and Masson’s Trichrome staining and

immunohistochemical staining was performed using standard procedures as previously

described.5 Immunohistochemical staining was performed using antibodies to detect

myosin (MF-20, 1:10, Developmental Studies Hybridoma Bank, Iowa City, IA) and

macrophages (CD68, 1:50, Serotec, Raleigh, NC). Images were captured and digitized

(DM4000B Leica Upright Microscope, Leica Microsystems, Bannockburn, IL) at varying

magnifications.

Statistics: Morphological and functional data were analyzed using one- and two-way

ANOVAs as indicated in the text. Upon finding a significant ANOVA, post-hoc

comparison testing of parameters of interest was performed using Fisher’s LSD. Statistical

279
significance was achieved at an alpha < 0.05. Values presented are mean ± SEM. Unless

otherwise stated, all mean values are expressed as the arithmetic mean.

Notably during the course of the study, TEMR responses demonstrated a markedly

greater variability than either NR or BAM (Figure 3). Because of the relatively large

variability, the sample size for the TEMR group was increased to 13 observations in an

effort to reduce the variability within this group. Ultimately, the coefficient of variation

(CV; standard deviation/mean) for the recovery of peak torque 12 weeks post-injury was

approximately two times greater for TEMR treated (CV = 0.20) than no repair (CV = 0.07)

or BAM (CV = 0.11) groups. In accordance with this greater variance, the recovery of peak

torque (12 wk Post-Injury: Pre) ranged from 0.36 to 0.71, indicating a range of functional

recoveries from essentially zero (no recovery) to complete TA muscle functional recovery,

respectively, across thirteen observations (Figure 3C). In light of this divergent response,

the TEMR group was separated into positive and negative responders based on the

following criteria: A TA muscle transplanted with a TEMR construct that recovered a peak

torque value greater than 1 standard deviation (determined from a combination of BAM

and TEMR observation; mean = 0.57, sd = 0.09) above the mean for the no repair group

was considered a positive responder.

Results

Synergist Muscle Ablation: Repeated in vivo functional testing was performed on each

animal at a monthly interval over a three month time frame to assess the functional capacity

of the TA muscle following VML injury and/or repair. In order to accurately assess the

impact of the VML injury as well as recovery of the TA muscle over time in the various

280
treatment groups, the anterior crural muscle synergists (EDL and EHL muscles) to the TA

muscle were ablated at the time of VML injury. The EDL and EHL muscles comprised a

combined 20.3 ± 1.3% of the anterior crural muscle wet weight (i.e., Uninjured TA, EDL,

& EHL muscle wet weight: 421.0 ± 6.6, 96.7 ± 1.2, 10.9 ± 0.3 mg, respectively), suggesting

the potential for TA muscle overload. Functionally, removal of the EDL and EHL muscles

resulted in a -29.4 ± 7.4% loss of peak torque from 4 to 12 weeks post-ablation when

normalized to pre-injury values (Figure 2). Twelve weeks post-ablation, wet weight was

similar among TA muscles from the ablated (446 ± 35 mg) and contralateral control (440

± 27 mg) legs (p = 0.701), indicating that synergist ablation did not provide a significant

overload to uninjured muscle similar that reported for the posterior compartment.31

Importantly, because synergist ablation resulted in a ≈30% reduction of normalized peak

tetanic torque, a complete recovery of TA muscle function following VML injury is

reflected by the return of ≈70% of pre-injury tetanic torque on that same animal using this

metric.

Creation of VML Injury: VML injury was created by surgically excising ≈20% of muscle

mass from the middle third of the TA muscle. Following surgical excision, animals were

divided into 3 treatment groups, no repair (NR), BAM implantation, and TEMR

implantation groups. Equivalent injuries were created in all treatment groups, as reflected

by the similar weight of muscle removed from the animals in the NR (70.8 ± 6.7 mg), BAM

(63.1 ± 3.1 mg), and TEMR (69.3 ± 1.6 mg) treatment groups). For non-repaired muscles

(surgical defect and synergist ablation), peak tetanic torque was reduced compared to pre-

injury values by 51.5 ± 2.8, 51.4 ± 3.2, and 49.6 ± 3.0% at 4, 8, and 12 weeks post-injury,

281
respectively, indicating that over this time-frame the VML injury did not recover

spontaneously (Figure 3). Twelve weeks post-injury, TA muscle wet weight was ≈17%

less for the VML injured NR group than in contralateral control muscles. Together these

results indicate that the TA muscle surgical defect produces an irrecoverable torque deficit

of ≈20% of the TA muscle functional capacity out to 12 weeks post-injury (with the

remainder of the ≈50% torque deficit attributed to ablation of the EDL and EHL).

Treatment In vivo of VML Injury

TA Muscle Strength Assessment: VML injury was repaired by either transplanting BAM

scaffolds or TEMR constructs to the defect site immediately after the injury. While body

weight was similar among all experimental groups (i.e., no repair, BAM, and TEMR)

during the study, over the 12 weeks post-injury each group gained significant body weight

(Figure 3A; Experimental Group x Time ANOVA p = 0.625, Time ANOVA p < 0.001).

To control for increases in in vivo torque production due to animal growth, peak torque

was normalized to body weight (Nmm/kg body wt).32, 33 There were no differences in pre-

injury (anterior crural muscles intact) torque values among the experimental groups (Figure

3B). As such, the degree of functional recovery observed for no repair, BAM or TEMR

groups was determined by calculating the ratio of peak torque at each post-injury time

relative to each respective pre-injury group mean. The pre-injury group mean was used as

a conservative approach to minimize any undue bias of a single pre-injury measurement

(Figure 3C).

A two-factor ANOVA was used to compare functional outcomes among the

different treatment groups over time. Statistical analysis revealed a significant effect of

282
treatment group (p<0.001), a significant effect of time (p<0.049), but no interaction

between time and treatment group (p=0.591). More specifically, all treatment groups

displayed improved function from 4-12 weeks post-injury. However, post-hoc analysis

revealed that at each time point, BAM significantly improved TA muscle functional

recovery compared to NR or TEMR negative responders (which were not different from

each other), while TEMR positive responders significantly improved functional recovery

to a greater extent than NR, BAM, and TEMR negative responders (Figure 3).

Consistent with the aforementioned observations, comparison of absolute peak

torque values among control (Un-operated and Sham/Synergist Ablation) and treatment

(i.e., no-repair, BAM, and TEMR positive and negative responders) groups at twelve weeks

post-injury revealed that TEMR positive responders had significantly greater peak

isometric torque values compared to non-repaired and BAM-repaired groups (Table 1).

Further, when isometric torque was normalized to TA muscle wet weight (an estimate of

specific torque), BAM treated muscles exhibited a significant deficit compared to control

groups (e.g., vs. Sham, p = 0.0278), while TEMR positive responders were similar to

control values (Table 1). Collectively, these findings indicate that TEMR positive

responders, and to a lesser extent BAM, improves the functional capacity of VML injured

muscle out to 12 weeks post-injury.

Morphological Characterization of Retrieved TA Muscle: VML injury resulted in

significant deformation and gross morphological remodeling of the TA muscle (Figure

4A). In non-repaired VML injured muscle, a longitudinal fissure was commonly observed

283
where the defect was originally created. Both BAM and TEMR positive responder groups,

defined by their functional response, appeared to partially fill this void, attenuating the

atrophic appearance of the muscle. Consistent with this observation, both BAM and TEMR

positive responders partially improved the distal width of the TA muscle (Figure 4B & C).

Functional recovery observed 12 weeks post-injury was significantly and positively

correlated with this improvement in distal TA muscle width (Figure 4D). Similarly, TA

muscle wet weight following VML injury was partially improved by BAM and TEMR

positive responders 12 weeks post-injury (Table 1).

Qualitative Histological Characterization of Retrieved TA Muscle: Twelve weeks post-

injury VML injured non-repaired TA muscle presented with fibrotic scarring at the defect

site with signs of aberrant muscle fiber generation marked by a few small disorganized

muscle fibers (Figure 5B&G). For treated VML injured muscle, the functional outcomes

were generally matched by the qualitative appearance of generated muscle fibers at the site

of the defect: that is, TEMR positive responders and BAM treated TA muscle presented

with signs of muscle fiber regeneration (Figures 5 & 6), marked by the formation of a band

of muscle fibers that regenerated in close proximity to the remaining muscle mass. TEMR

negative responders exhibited little to no signs of muscle regeneration (Figures 5E, J &

6C). In corroboration with the variable functional recovery with TEMR transplantation,

TEMR positive and negative responders appeared to have a differential immune response

– TEMR negative responders were marked with a robust macrophage (CD68 + cells)

presence taking the form of foreign body giant cells (Figure 6F), while TEMR positive

responders were marked also by macrophage presence but in a manner phenotypically

284
similar, albeit apparently greater in magnitude, than BAM treated TA muscles (Figure E &

D). A general summary of the performance of BAM scaffolds and TEMR constructs are

provided in Table 2.

Discussion

Over the past decade a focused research effort has been placed on developing

therapeutic strategies aimed at generating a clinically relevant volume of skeletal muscle

for the purpose of ultimately restoring function to traumatized musculoskeletal tissue.

While a variety of tissue engineering approaches have been successful in promoting partial

regeneration of lost skeletal muscle fibers at the site of VML, ultimately an improvement

in force production (i.e., net torque production) of the injured musculature is the most

meaningful treatment outcome. The primary finding of this study is that transplantation of

the TEMR construct and BAM is associated with significant increases in the magnitude of

TA muscle contraction (i.e., torque) following electrical stimulation of the peroneal nerve

in vivo, for up to twelve weeks post-VML injury in an immune competent Lewis rat model

(Figure 3; Table 1). Importantly, TEMR transplantation resulted in a highly variable

therapeutic response, wherein no (zero) to full recovery of function was observed. Twelve

weeks post-injury, TEMR positive responders (n = 6 of 13 observations; see Methods for

full details) and BAM transplantation promoted a mean recovery of 26 and 61% of the

functional deficit, respectively (Table 1). On the other hand, TEMR negative responders

(n = 7 of 13 observations) did not improve the functional recovery of VML injured muscle.

Our previous findings with TEMR transplantation in a nude mouse LD muscle VML injury

model5, 8 and other studies13, 16 support these findings: that TEMR positive responders and

285
to a lesser extent, BAM transplantation, can improve the functional capacity of skeletal

muscle with VML.

Sheet-based acellular matrix tissue engineering approaches with and without the

inclusion of a stem or progenitor cell source have previously been shown to promote

skeletal muscle fiber regeneration.5, 6, 8-10, 13, 15-17, 19 The current study confirms and extends

our prior work in the murine LD VML injury model5, 8


demonstrating that TEMR

transplantation can result in a low to moderate level of muscle fiber regeneration inside the

transplanted construct within three months after transplantation in an animal model with a

competent immune system. Additionally, in the current study BAM transplantation also

promoted muscle fiber regeneration within the same time frame, similar to that reported

recently in the mouse quadriceps following transplantation of subintestinal submucosa-

ECM material.17. On the other hand, TEMR negative responders exhibited virtually no

evidence of myogenesis in the defect area. This raises the ongoing debate as to whether

inclusion of a cellular source with a matrix is necessary to maximize the therapeutic

response of ECM-based tissue engineering devices. There is currently ample evidence

within the literature to support each approach e.g.,9, 16, 20, 21 While the findings of this study

do not settle this question, they do indicate that the inclusion of a cellular component with

a biological ECM has the potential (i.e., TEMR positive responders) to promote a 61%

functional recovery, a 2.3-fold increase in recovery compared to that elicited by BAM

alone (26% recovery), in this rodent model of VML injury. Clearly, further work is required

to increase the reproducibility of the desired functional outcome, and thus, the eventual

translational value of the TEMR technology.

286
The existing literature suggests that the mechanism of functional recovery after

VML injury mediated by scaffold-based tissue engineering approaches is multifactorial.

Of the VML studies that have reported functional improvements following therapy (e.g.,5,
8, 9, 13
), most have demonstrated a partial regeneration of the volume of muscle tissue loss

– the magnitude of tissue regeneration in these reports and in the current study, however,

are comparable to others that did not measure function. This suggests that functional gains

mediated by delivery of acellular scaffolds with and without and stem or progenitor cell

source across multiple VML models are due not only to the regeneration of muscle fibers,

but also by potentially; 1) improving the functional capacity of the remaining muscle mass

(e.g., functional hypertrophy34), 2) augmenting the efficiency of force transmission across

the defect35-38, or 3) protecting the remaining muscle mass from continued injury related to

mechanical overload that may lead to persistent inflammation and fibrosis39, 40.

Highlighting these possibilities, recently it has been shown that transplantation of

a decellularized muscle ECM, which promoted strictly fibrosis within the defect area, still

resulted in enhanced functional restoration at intermittent time points (e.g., 2 and 4 months

post-injury).41 As an extension of these results, in the current study we observed a

significant atrophy of VML injured muscles that was most severe in the distal third of the

tissue (distal to the defect site)- reminiscent of the effect of laceration on skeletal muscle.42-
44
Interestingly, the atrophic response of the distal portion of the TA muscle was attenuated

with TEMR and BAM transplantation, in a manner directly related to each group’s

functional recovery (Figure 4D). The benefit of mechanical loading on tissue regeneration

has long been appreciated,45 wherein increased and decreased activity can positively and

negatively impact regeneration, respectively. In this context, it is plausible that the

287
transplantation of an acelluar scaffold may improve function by promoting the formation

of a mechanical bridge that is capable of improving force transmission throughout the

remaining muscle.41 This concept appears to parallel the biological response of skeletal

muscle to disruptions in cytoskeletal architecture (e.g., desmin knockout mice46), with

repeated lengthening contraction training39, 40, and clinically following laceration of the

biceps brachii muscle.47 Future studies should address the mechanistic basis for the

functional effects of various tissue engineering strategies following VML injury, as the

mechanisms that drive these distinct aspects of functional recovery will provide important

insight into the development of efficient regenerative strategies.

To continue the development of TEMR technology towards clinical applications, it

is important to identify why this group as a whole presented such a wide range of functional

outcomes in this VML injury model. One possibility is that the additional mechanical

manipulation involved with insertion of the TEMR scaffold in the TA VML injury model

(relative to the previously reported LD model) results in a decline in the retention of, or

damage to, the cellular component at implantation. These disturbances would effectively

diminish the presumptive myogenic5, 8, angiogenic 48, or immunomodulatory 49 effects of

MDCs. This supposition is consistent with the clear delineation in the regeneration of

muscle fibers and macrophage infiltration characterizing the positive and negative TEMR

responders (Figure 6). Moreover, delivery of the “damaged” TEMR construct may have

increased inflammation in a manner similar to that observed upon transplantation of grafted

whole tissue50, and may have therefore interfered with improvements in function mediated

via transplantation of BAM alone.

288
The primary findings of this paper highlight that the transplantation of

decellularized ECM with (TEMR positive responders) and without (BAM) the inclusion

of MDCs can promote significant functional recovery in skeletal muscle with VML injury.

Further, the above findings also highlight the potentially greater regenerative capacity of

therapies that include stem or progenitor cells in addition to a scaffold. Specifically, this

possibility is evidenced by the fact that TEMR positive responders promoted a 2.3-fold

greater functional recovery than did BAM alone. On the other hand, these findings also

point to the additional complexities of cell inclusion, as neither functional recovery nor

muscle fiber regeneration was observed in the TEMR negative responders. The high degree

of variance among TEMR responders appears to be related to pre-surgical handling of the

constructs (e.g., physical manipulation), and therefore, we are confident that further

optimization of this technology (i.e., reduced surgical handling, improved cellular seeding,

layering of scaffold sheets or utilization of more favorable scaffold geometries) will

mitigate these issues. In short, while there is still significant room for functional

improvement, these initial studies provide important guidance for future development of

tissue engineering therapies, and furthermore, document the importance of studying these

technologies in distinct animal models and VML injuries in order to develop the strategies

that will best accommodate the full spectrum of functional and cosmetic deficits that result

from such injuries.

289
Figure Legends

Figure 1. TA muscle VML injury repair with BAM scaffold and TEMR constructs.

A) A 3 x 0.5 cm strip of BAM or TEMR was folded in triplicate prior to transplantation.

B) TEMR constructs were comprised of BAM with an aligned cellular content (MDCs

were seeded at 1 million cells per cm2). Scale bar = 50 µm. C) VML injury was surgically

created by making a ≈ 1 x 0.5 x 0.3 cm defect in the left TA muscle and was D) immediately

treated with BAM or TEMR constructs that were sutured in the fresh wound bed as

depicted and described in the methods.

Figure 2. In vivo isometric torque of the anterior crural muscles before and after TA

muscle synergist ablation and VML. A) Representative digitized torque tracings are

presented for uninjured intact anterior crural muscles (TA, EDL, & EHL muscles), and

after ablation of the EDL and EHL muscles and subsequent VML. Note: Following

ablation (4 – 12 weeks), the TA muscle produced ≈70% of net anterior crural muscle

torque. Subsequent VML of the TA muscle (see methods) resulted in a further ≈20%

deficit, resulting in a combined ≈50% deficit compared to pre-injury (Intact) torque. B)

TA muscle peak isometric torque (Nmm/kg body wt) normalized to pre-injury values was

stable from four to twelve weeks post-synergist ablation.

Figure 3. TEMR improves functional recovery of rat TA muscle with VML injury.

A) Group mean rat body weight over the course of 12 weeks. No differences were observed

among groups at each time point (see results). All means at each time point demarked with

different letters are significantly different (p < 0.05). B) To control for the effect of normal

growth over the three month study on peak isometric torque production, torque was
290
normalized to body weight (Nmm/kg body weight). Prior to VML, the average peak

isometric torque for each experimental group was similar. C) Individual responses at each

post-injury time point (4, 8, & 12 weeks) were normalized to their respective pre-injury

group mean. Individual responses and group means at each time point post-injury are

presented. Across all time points, BAM significantly restored torque to a greater extent

than NR or TEMR negative responders (*), while TEMR positive responders improved

torque recovery to a greater extent than all other experimental groups (†). Where

applicable, values are listed as mean ± SEM. Group sample sizes are listed in parentheses

in the 12-week response in panel C.

Figure 4. TA muscle gross morphology 12 weeks after VML and treatment. A) The

gross appearance of VML injured muscle without (NR, no repair) and with (BAM or

TEMR) treatment was clearly distinguishable from uninjured muscle. Note: A longitudinal

fissure in the middle third of the muscle was visible in NR muscles. Implantation of either

BAM or TEMR filled this void. B & C) TA muscles appeared atrophied, with distinctly

smaller widths of the proximal and distal thirds of the muscle. D) The extent of atrophy in

the distal TA width was positively correlated with functional recovery 12 weeks post-

injury. Values are means ± SEM. Means at each time point demarked with different letters

are significantly different (p < 0.05).

291
Figure 5. TA muscle tissue morphology following transplantation of BAM and TEMR

constructs. TA muscles were harvested 12 weeks after VML injury. Longitudinal (A – E)

and cross-sections (F – J) from uninjured (A, F), non-repaired (B, G), BAM-repaired (C,

H), and TEMR positive (D, I) and negative (E, I) responders were stained with Mason’s

Trichrome (Red = Tissue; Blue = Collagen; Black = Nuclei). *denotes the area of the defect

where the constructs were transplanted. Scale bars = 50 µm.

Figure 6. TA muscle fiber generation and inflammation following transplantation of

BAM and TEMR constructs. TA muscles were harvested 12 weeks after VML injury.

Longitudinal sections were probed for myosin expression (MF20; A – C) and cross sections

were probed for macrophage presence (CD68; D – F) in BAM (A, D), TEMR positive (B,

E) and TEMR negative (C, F) responder muscles. *denotes the area of the defect where the

constructs were transplanted. Scale bars = 50 µm.

292
293
294
Figure 1. TA muscle VML injury repair with BAM scaffold and TEMR constructs

295
Figure 2. In vivo isometric torque of the anterior crural muscles before and after TA

muscle synergist ablation and VML

296
Figure 3. TEMR improves functional recovery of rat TA muscle with VML injury

297
Figure 4. TA muscle gross morphology 12 weeks after VML and treatment.

298
Figure 5. TA muscle tissue morphology following

transplantation of BAM and TEMR constructs

299
300
Figure 6. TA muscle fiber generation and inflammation

following transplantation of BAM and TEMR constructs


References

1. Grogan, B.F., Hsu, J.R. Volumetric muscle loss. J Am Acad Orthop Surg.19 Suppl

1 2011.

2. Vekris, M.D., Beris, A.E., Lykissas, M.G., Korompilias, A.V., Vekris, A.D.,

Soucacos, P.N. Restoration of elbow function in severe brachial plexus paralysis

via muscle transfers. Injury.39 Suppl 3 2008.

3. Lin, C.H., Lin, Y.T., Yeh, J.T., Chen, C.T. Free functioning muscle transfer for

lower extremity posttraumatic composite structure and functional defect. Plast

Reconstr Surg.119 2007.

4. Owens, J.G., Blair, J.A., Patzkowski, J.C., Blanck, R.V., Hsu, J.R. Return to

running and sports participation after limb salvage. J Trauma.71 2011.

5. Corona, B.T., Machingal, M.A., Criswell, T., Vadhavkar, M., Dannahower, A.C.,

Bergman, C., et al. Further development of a tissue engineered muscle repair

construct in vitro for enhanced functional recovery following implantation in vivo

in a murine model of volumetric muscle loss injury. Tissue Eng Part A.18 2012.

6. De Coppi, P., Bellini, S., Conconi, M.T., Sabatti, M., Simonato, E., Gamba, P.G.,

et al. Myoblast-acellular skeletal muscle matrix constructs guarantee a long-term

repair of experimental full-thickness abdominal wall defects. Tissue Eng.12 2006.

7. Gamba, P.G., Conconi, M.T., Lo Piccolo, R., Zara, G., Spinazzi, R., Parnigotto,

P.P. Experimental abdominal wall defect repaired with acellular matrix. Pediatr

Surg Int.18 2002.

301
8. Machingal, M.A., Corona, B.T., Walters, T.J., Kesireddy, V., Koval, C.N.,

Dannahower, A., et al. A Tissue-Engineered Muscle Repair Construct for

Functional Restoration of an Irrecoverable Muscle Injury in a Murine Model.

Tissue Eng Part A. 2011.

9. Merritt, E.K., Cannon, M.V., Hammers, D.W., Le, L.N., Gokhale, R., Sarathy, A.,

et al. Repair of traumatic skeletal muscle injury with bone-marrow-derived

mesenchymal stem cells seeded on extracellular matrix. Tissue Eng Part A.16 2010.

10. Merritt, E.K., Hammers, D.W., Tierney, M., Suggs, L.J., Walters, T.J., Farrar, R.P.

Functional assessment of skeletal muscle regeneration utilizing homologous

extracellular matrix as scaffolding. Tissue Eng Part A.16 2010.

11. Perniconi, B., Costa, A., Aulino, P., Teodori, L., Adamo, S., Coletti, D. The pro-

myogenic environment provided by whole organ scale acellular scaffolds from

skeletal muscle. Biomaterials.32 2011.

12. Turner, N.J., Badylak, J.S., Weber, D.J., Badylak, S.F. Biologic Scaffold

Remodeling in a Dog Model of Complex Musculoskeletal Injury. J Surg Res. 2011.

13. Valentin, J.E., Turner, N.J., Gilbert, T.W., Badylak, S.F. Functional skeletal muscle

formation with a biologic scaffold. Biomaterials.31 2010.

14. Willett, N.J., Li, M.T., Uhrig, B.A., Boerckel, J.D., Huebsch, N., Lundgren, T.S.,

et al. Attenuated rhBMP-2 Mediated Bone Regeneration in a Rat Model of

Composite Bone and Muscle Injury. Tissue Eng Part C Methods. 2012.

15. Wolf, M.T., Daly, K.A., Reing, J.E., Badylak, S.F. Biologic scaffold composed of

skeletal muscle extracellular matrix. Biomaterials.33 2012.

302
16. Turner, N.J., Yates, A.J., Jr., Weber, D.J., Qureshi, I.R., Stolz, D.B., Gilbert, T.W.,

et al. Xenogeneic extracellular matrix as an inductive scaffold for regeneration of a

functioning musculotendinous junction. Tissue Eng Part A.16 2010.

17. Sicari, B.M., Agrawal, V., Siu, B.F., Medberry, C.J., Dearth, C.L., Turner, N.J., et

al. A murine model of volumetric muscle loss and a regenerative medicine approach

for tissue replacement. Tissue Eng Part A.18 2012.

18. Mase, V.J., Jr., Hsu, J.R., Wolf, S.E., Wenke, J.C., Baer, D.G., Owens, J., et al.

Clinical application of an acellular biologic scaffold for surgical repair of a large,

traumatic quadriceps femoris muscle defect. Orthopedics.33 2010.

19. Conconi, M.T., De Coppi, P., Bellini, S., Zara, G., Sabatti, M., Marzaro, M., et al.

Homologous muscle acellular matrix seeded with autologous myoblasts as a tissue-

engineering approach to abdominal wall-defect repair. Biomaterials.26 2005.

20. Page, R.L., Malcuit, C., Vilner, L., Vojtic, I., Shaw, S., Hedblom, E., et al.

Restoration of skeletal muscle defects with adult human cells delivered on fibrin

microthreads. Tissue Eng Part A.17 2011.

21. Rossi, C.A., Flaibani, M., Blaauw, B., Pozzobon, M., Figallo, E., Reggiani, C., et

al. In vivo tissue engineering of functional skeletal muscle by freshly isolated

satellite cells embedded in a photopolymerizable hydrogel. FASEB J.25 2011.

22. Gharaibeh, B., Lavasani, M., Cummins, J.H., Huard, J. Terminal differentiation is

not a major determinant for the success of stem cell therapy - cross-talk between

muscle-derived stem cells and host cells. Stem Cell Res Ther.2 2011.

23. Singer, N.G., Caplan, A.I. Mesenchymal stem cells: mechanisms of inflammation.

Annu Rev Pathol.6 2011.

303
24. Zammit, P.S., Partridge, T.A., Yablonka-Reuveni, Z. The skeletal muscle satellite

cell: the stem cell that came in from the cold. J Histochem Cytochem.54 2006.

25. Hawke, T.J., Garry, D.J. Myogenic satellite cells: physiology to molecular biology.

J Appl Physiol.91 2001.

26. Moon Du, G., Christ, G., Stitzel, J.D., Atala, A., Yoo, J.J. Cyclic mechanical

preconditioning improves engineered muscle contraction. Tissue Eng Part A.14

2008.

27. Wu, X., Corona, B.T., Chen, X., Walters, T.J. A Standardized Rat Model of

Volumetric Muscle Loss Injury for the Development of Tissue Engineering

Therapies. BioResearch Open Access.In Press 2012.

28. Corona, B.T., Rouviere, C., Hamilton, S.L., Ingalls, C.P. Eccentric contractions do

not induce rhabdomyolysis in malignant hyperthermia susceptible mice. J Appl

Physiol.105 2008.

29. Criswell, T.L., Corona, B.T., Ward, C.L., Miller, M., Patel, M., Wang, Z., et al.

Compression-induced muscle injury in rats that mimics compartment syndrome in

humans. Am J Pathol.180 2012.

30. Ingalls, C.P., Warren, G.L., Armstrong, R.B. Dissociation of force production from

MHC and actin contents in muscles injured by eccentric contractions. J Muscle Res

Cell Motil.19 1998.

31. Kandarian, S.C., Young, J.C., Gomez, E.E. Adaptation in synergistic muscles to

soleus and plantaris muscle removal in the rat hindlimb. Life sciences.51 1992.

304
32. Corona, B.T., Balog, E.M., Doyle, J.A., Rupp, J.C., Luke, R.C., Ingalls, C.P.

Junctophilin damage contributes to early strength deficits and EC coupling failure

after eccentric contractions. Am J Physiol Cell Physiol.298 2010.

33. Ingalls, C.P., Warren, G.L., Lowe, D.A., Boorstein, D.B., Armstrong, R.B.

Differential effects of anesthetics on in vivo skeletal muscle contractile function in

the mouse. J Appl Physiol.80 1996.

34. Gollnick, P.D., Timson, B.F., Moore, R.L., Riedy, M. Muscular enlargement and

number of fibers in skeletal muscles of rats. J Appl Physiol.50 1981.

35. Lehto, M., Duance, V.C., Restall, D. Collagen and fibronectin in a healing skeletal

muscle injury. An immunohistological study of the effects of physical activity on

the repair of injured gastrocnemius muscle in the rat. J Bone Joint Surg Br.67 1985.

36. Purslow, P.P. The structure and functional significance of variations in the

connective tissue within muscle. Comp Biochem Physiol A Mol Integr Physiol.133

2002.

37. Purslow, P.P. Muscle fascia and force transmission. J Bodyw Mov Ther.14 2010.

38. Street, S.F., Ramsey, R.W. Sarcolemma: transmitter of active tension in frog

skeletal muscle. Science.149 1965.

39. Stauber, W.T., Knack, K.K., Miller, G.R., Grimmett, J.G. Fibrosis and intercellular

collagen connections from four weeks of muscle strains. Muscle Nerve.19 1996.

40. Stauber, W.T., Smith, C.A., Miller, G.R., Stauber, F.D. Recovery from 6 weeks of

repeated strain injury to rat soleus muscles. Muscle Nerve.23 2000.

305
41. Corona, B.T., Wu, X., Ward, C.L., Mcdaniel, J.S., Rathbone, C.R., Walters, T.J.

The promotion of a functional fibrosis in skeletal muscle with volumetric muscle

loss injury following the transplantation of muscle-ECM. Biomaterials.34 2013.

42. Kaariainen, M., Kaariainen, J., Jarvinen, T.L., Sievanen, H., Kalimo, H., Jarvinen,

M. Correlation between biomechanical and structural changes during the

regeneration of skeletal muscle after laceration injury. J Orthop Res.16 1998.

43. Lim, A.Y., Lahiri, A., Pereira, B.P., Tan, J.A., Sebastin, S.J., Tan, B.L., et al. The

role of intramuscular nerve repair in the recovery of lacerated skeletal muscles.

Muscle Nerve.33 2006.

44. Pereira, B.P., Tan, J.A., Zheng, L., Tan, B.L., Lahiri, A., Lim, A.Y., et al. The cut

intramuscular nerve affects the recovery in the lacerated skeletal muscle. J Orthop

Res.24 2006.

45. Denny-Brown, D. The influence of tension and innervation on the regeneration of

skeletal muscle. J Neuropathol Exp Neurol.10 1951.

46. Meyer, G.A., Lieber, R.L. Skeletal muscle fibrosis develops in response to desmin

deletion. Am J Physiol Cell Physiol.302 2012.

47. Kragh, J.F., Jr., Basamania, C.J. Surgical repair of acute traumatic closed

transection of the biceps brachii. J Bone Joint Surg Am.84-A 2002.

48. Rhoads, R.P., Johnson, R.M., Rathbone, C.R., Liu, X., Temm-Grove, C., Sheehan,

S.M., et al. Satellite cell-mediated angiogenesis in vitro coincides with a functional

hypoxia-inducible factor pathway. Am J Physiol Cell Physiol.296 2009.

306
49. Lecourt, S., Lepelletier, Y., Vanneaux, V., Jarray, R., Domet, T., Raynaud, F., et

al. Human Muscle Progenitor Cells Displayed Immunosuppressive Effect through

Galectin-1 and Semaphorin-3A. Stem Cells Int.2012 2012.

50. Badylak, S.F., Valentin, J.E., Ravindra, A.K., Mccabe, G.P., Stewart-Akers, A.M.

Macrophage phenotype as a determinant of biologic scaffold remodeling. Tissue

Eng Part A.14 2008.

307
APPENDIX 2

Ethical Considerations in Tissue Engineering Research: Case Studies in Translation

Hannah B. Bakera, John P. McQuillinga, Nancy M. P. Kingb*

Affiliations:
a
Department of Biomedical Engineering, Virginia Tech-Wake Forest School of

Biomedical Engineering and Sciences; Wake Forest Institute for Regenerative Medicine,

391 Technology Way, Winston-Salem, NC 27101

b
Department of Social Sciences and Health Policy and Wake Forest Institute for

Regenerative Medicine, Wake Forest School of Medicine; Center for Bioethics, Health,

and Society and Graduate Program in Bioethics, Wake Forest University

Medical Center Blvd, Winston-Salem, NC 27157


*
corresponding author, nmpking@wakehealth.edu, 336.716.4289

Note: This manuscript has been submitted for publication. Hannah B. Baker, John P.

McQuilling, and Nancy M. P. King co-wrote this manuscript.

308
Abstract

Tissue engineering research is a complex process that requires investigators to focus on the

relationship between their research and anticipated gains in both knowledge and treatment

improvements. The ethical considerations arising from tissue engineering research are

similarly complex when addressing the translational progression from bench to bedside,

and investigators in the field of tissue engineering act as moral agents at each step of their

research along the translational pathway, from early benchwork and preclinical studies to

clinical research. This review highlights the ethical considerations and challenges at each

stage of research, by comparing issues surrounding two translational tissue engineering

technologies: the bioartificial pancreas and a tissue engineered skeletal muscle construct.

We present relevant ethical issues and questions to consider at each step along the

translational pathway, from the basic science bench to preclinical research to first-in-

human clinical trials. Topics at the bench level include maintaining data integrity,

appropriate reporting and dissemination of results, and ensuring that studies are designed

to yield results suitable for advancing research. Topics in preclinical research include the

principle of “modest translational distance” and appropriate animal models. Topics in

clinical research include key issues that arise in early-stage clinical trials, including

selection of patient-subjects, disclosure of uncertainty, and defining success. The

comparison of these two technologies and their ethical issues brings to light many

challenges for translational tissue engineering research and provides guidance for

investigators engaged in development of any tissue engineering technology.

309
Highlights

• Investigators at all stages of translational research are moral agents

• Every trial should produce results that are able to guide researchers in designing the

next study

• Well-designed translational research has value even when ultimately unsuccessful

• Increased discussion of ethics promotes transparency and knowledge-sharing in

research

Keywords:

human research ethics, animal research ethics, translational research ethics, tissue-

engineered skeletal muscle, bioartificial pancreas

Abbreviations (nonstandard):

TESM = tissue engineered skeletal muscle

BAP = bioartificial pancreas

TM = therapeutic misconception

TD = translational distance

FIH = first-in-human

CRO = contract research organization

310
1. Introduction

Tissue engineering research is regenerative medicine research that emphasizes

combining cells, tissues, and various enabling technologies, from scaffolds and capsules to

bioreactors, to develop tissues and other biomaterials capable of replacing or augmenting

physiological and biochemical functions impaired by illness or injury [1, pp. 215-216; 2,

p. 11]. Research to develop so-called “combination products” is complex, both

scientifically and from a regulatory standpoint. The ethical issues that arise in tissue

engineering research are likewise complex and worthy of careful examination [3-5]. It is

often helpful to address research ethics issues in the context of specific research case

studies [6]. This essay enumerates some basic ethical considerations for tissue engineering

research, and examines in detail their application to two representative research case

studies: tissue-engineered skeletal muscle (TESM) constructs and the bioartificial pancreas

(BAP).

It is currently popular, even necessary, to describe research—especially research

involving novel biotechnologies—as “translational.” In addition to being trendy, however,

the term is important, because it helps to illustrate the need for and value of viewing all

health-related research comprehensively relating each line of research to anticipated

knowledge gains and health improvements. Translation should not imply certainty that a

line of research will lead neatly to safe and effective products or treatments. Rather, it

should signal the responsibility of investigators to think ahead along the line of research,

to anticipate the relationship between study design and research ethics, and to consider and

periodically reconsider how to do research that has scientific and societal value, regardless

of the direction taken along the translational pathway from one experiment to the next [7].

311
Investigators at all stages of translational research are moral agents, with

profession-specific moral duties that apply to the design and conduct of their research.

These duties can be described generally, but must also be articulated and applied to the

specific context of a given study. This essay therefore first addresses common ethical

considerations in tissue engineering research at the laboratory, preclinical, and human

research stages. There are important ethical considerations common to all research, and

additional considerations that particular research stages have in common. Next, we

consider these ethical issues as they arise in our two case studies, which were chosen for

their differences, with the goal of comparison and contrast. It is our hope that examining

the application and relative importance of the ethical considerations affecting the design

and conduct of studies of TESM and BAP along the translational pathway will help to

model similar thinking for tissue engineering researchers working with different tissue

constructs, and thus make it easier to engage in thoughtful research at all stages from the

bench to the bedside.

1.1. In the laboratory (in vitro and pre-clinical animal research)

Much tissue engineering research is still in preclinical stages. Although ethical

issues are often underaddressed in research until human trials have begun [8], there are

many issues worthy of consideration at the bench. These include: data integrity,

responsible reporting and dissemination of results, and ensuring that every study is

designed and conducted so that it can yield results suitable to decide on the next research

steps [9]. It is important to recognize that, at any stage, the results of well-designed and

properly conducted research might lead not forward, but back or in a different direction

312
entirely, to refine and expand knowledge at an earlier stage or to explore and develop newly

identified possibilities [7].

The use of animal models at preclinical stages remains vital to the success of tissue

engineering research. Efforts are underway to minimize the use of animals, but potential

alternatives like computer modeling and body-on-a-chip organoid arrays have significant

limitations and require considerable further development [10]. Researchers therefore must

consider the three Rs of animal research -- Reduce, Refine, and Replace [11]. The choice

of animal models, and their humane and appropriate use, helps to ensure that the research

transition from animals to humans adheres to the principle of “modest translational distance”

described by Jonathan Kimmelman. Translational distance (TD) refers to the number and

size of inferential leaps from animals to humans [7], pp. 117-122] – in other words, it is a

measure of uncertainty. In first-in-human (FIH) and other early-stage research, modest TD

may provide an analytical model for considering the relationship of research design and

ethics – a role that cannot be filled by the concept of clinical equipoise, which applies only

to later-stage research such as trials comparing experimental interventions to standard

treatment. Clinical equipoise justifies asking patient-subjects to risk receiving an unproven

intervention in a clinical trial when there is sufficient evidence that reasonable clinicians

consider both the unproven intervention and currently available standard treatment to be a

reasonable treatment choice under the circumstances [12]. Early-stage research cannot

offer the potential for direct benefit to patient-subjects that may be available in later-stage

research. Instead, modest TD justifies asking patient-subjects in early-stage research to risk

receiving an unproven intervention only when the “inference gap” is small enough to

313
predict both that the clinical trial can yield useful results and that it can adequately protect

their safety – not because it is reasonable to anticipate direct benefit.

1.2. Clinical research with patients as subjects

Much has been written about ethical issues in clinical trials [13-15]; this body of

scholarly literature, and the issues it addresses, continues to grow. For tissue engineering,

like other regenerative medicine research, it is most important to address key issues arising

in early-stage clinical trials [16-18]. Those issues include: moving into human subjects;

designing FIH trials; selecting patient-subjects; and informed consent.

In order to move from preclinical to FIH and other early-stage trials, several

questions must be answered in the affirmative:

• Has enough preclinical information been collected so that the only reasonable way

to learn more is to move to humans? This is another way of asking whether modest

TD has been achieved.

• Has enough been done to reduce the risks of harm to humans, and to maximize the

likelihood that the intervention will ultimately show benefit in humans? This

question about the appropriate balancing of the risks of harm and potential benefits

applies at all stages of clinical research, but is particularly important in early-stage

research, because what counts as potential direct benefit to patient-subjects in these

trials is limited and uncertain at best [19].

• Has the point of irreducible uncertainty been reached? In early-stage research, this

question is more applicable than the question “Have the risks of harm been

minimized as far as possible?” because it acknowledges that some risks of harm are

still unknown, and that it is never truly possible to eliminate all uncertainty.

314
• Is the amount of irreducible uncertainty small enough that it is fair to subjects to

ask them to become involved in the research? Addressing this evaluative question

is a key component of the researcher’s ethical inquiry. The answer is less important

than the reasons that can be given to support an affirmative answer. Like the more

familiar requirement that risks of harm and potential benefits “must be balanced

and shown to be in a favorable ratio” [15], this reasoning forms the basis of

productive discussions between researchers, their colleagues, and oversight bodies

like funders, study sections, and institutional review boards (IRBs).

Two key considerations that relate the design of clinical trials to research ethics

considerations are scientific validity and social value. A valid study is methodologically

rigorous, designed and powered to provide useful answers to the questions it asks –

including negative answers. A valuable study asks a socially meaningful and/or

scientifically useful question [14]. Value in research is usually defined as progressive

value—that is, whether a study’s results are able to move the line of research directly

forward to the next phase. However, FIH and other early-stage research is also highly

likely to have translational value that is not progressive, but may be reciprocal, iterative,

or collateral [7], pp. 92-94]. Reciprocal value highlights the necessity of and informs new

preclinical work. Iterative value helps to inform the clinical trial itself by providing new

information that can be productively used for in-trial modifications as the trial goes forward.

Collateral value helps one or more different trials, by making new information, experience,

and techniques more broadly available to researchers in related research or related fields.

Researchers who recognize the broad applicability of study data and design translational

research to take advantage of many different types of value can readily plan both to address

315
what would otherwise simply be regarded as failures of research progress and to enhance

the value of successful research.

Selecting patient-subjects from whom scientifically useful data can be collected and

who are also able to make well-informed choices about research participation is especially

important in early-stage research. Choosing the population from whom the most may be

learned and who also can most readily be helped to make autonomous choices about

participation may be challenging, as sometimes those are two different populations of

potential patient-subjects [16,20]. Researchers must take care to provide good information,

not only to potential subjects in the consent form and process, but also to the media and in

publications discussing study results. Good information reduces the likelihood that the

therapeutic misconception (TM) will cause potential subjects, their families, oversight

bodies, the media and the public, and even fellow researchers to overestimate the potential

benefits of the research or to underestimate its risks of harm [21,22]. The need for long-

term follow-up, both to monitor the health and function of patient-subjects after the

intervention and to determine the intervention’s success or failure, is often overlooked, not

only in study design and budgeting, but also in disclosure to potential subjects.

2. Case study 1: Ethical considerations in translation of TESM technologies

2.1. Introduction to the technology

In this case study, the TESM consists of a cell-seeded biomaterial construct used to

enable repair and/or remodeling in a muscle loss injury or pathology. Such a technology

aims to replace the current standard of care, which is the use of host or cadaveric muscle

flaps for repair and reconstruction. Using host muscle flaps means creating a new, ideally

316
less consequential region of muscle loss in order to repair the initial injury. Using cadaveric

muscle flaps requires an immunosuppressive regimen. In contrast, using TESM, if

perfected, would also require surgical implantation, but would not create an additional

injury or the need for immunosuppression.

In future human studies, the generation of a TESM construct would begin weeks

prior to implant with isolation of muscle cells from a biopsy taken from a patient-subject.

Those cells would then be expanded in culture and eventually seeded onto a biocompatible

scaffold. Next, depending on the particular TESM technology employed, the cellularized

scaffold may be subjected to additional physiological cues to guide further maturation of

the cells on the construct [23,24] Finally, the construct would be sutured into the tissue

void.

Initial proof of concept for a TESM technology must be established in vitro prior

to any animal studies; this alone can take years. This initial effort would be followed by in

vivo studies, which seek to first develop a working animal model of a muscle injury that

the investigator is interested in treating. This in vivo work can be inherently lengthy, as

each study will need to carry on long enough to capture the full potential recovery for the

muscle pathology of interest, which can vary from days for minor injuries, such as sports-

related muscle damage, to months for volumetric muscle loss injuries [23,25]. The

complexity of this research suggests unique ethical considerations at each stage leading up

to clinical translation. This case study explores those issues at both early and pre-clinical

stages and in clinical trials.

2.2. Research ethics in the lab

317
As noted previously, for preclinical research early on in a translational project, the

primary ethical considerations should focus on data integrity, both for the value of the

science and for the sake of future beneficiaries of the research. Every trial should produce

results that are able to guide researchers in designing the next study. In animal studies, in

addition, researchers should apply the three Rs to the maximum extent possible.

2.2.1. In vitro

Ethical considerations at the bench include: keeping good records, practicing good

data collection and management, transparency of data-sharing and realistic representation

of study results, and preparing for transition to animal studies.

In aiming to conduct authentic translational research on TESM, even early stage

investigators must be cognizant of the potential beneficiaries of the technology. Work at

the bench and in the culture hood may someday help treat a wounded warrior, a cancer

patient, or someone born with a congenital birth defect. As such, ethical considerations at

this stage are vital. It means that the quirk identified in the protocol but left undocumented

might be the difference between successful translation of work into the clinic and loss of

progress. Details—in cell culture, making up reagents, and handling of constructs—are all

very important, and good documentation is a key requirement. Staying aware of the

potential benefits of responsible laboratory practices and the ramifications of failure to

attend to detail is important in day-to-day research even at the early stages [9].

In complex research like TESM, it may be necessary to involve an unbiased third

party research group such as a contract research organization (CRO) to repeat some of the

research. Doing so at an early stage may make it easier to reduce TD, show proof of

concept for animal studies, and prepare to move toward clinical trials.

318
In addition to facilitating direct translational progress, in vitro research can also

have knowledge-generating results with broader applications. In TESM research, culture

of muscle cells on scaffolds can help to inform both the use and development of enabling

technologies such as specialized imaging modalities and the use of muscle cells in other

applications [26,27] – important examples of reciprocal and collateral value.

2.2.2. In vivo

Meeting the goals of the three Rs is challenging in TESM research. With respect

to the first R, “Replace”, there exists no potential alternative, such as a computer model or

a cell line, which can completely replace the use of animals to study TESM technology in

a physiologically meaningful manner. Studying muscle cells seeded onto biomaterial

scaffolds can provide useful information about the interaction between the cells and the

construct, but cell-seeded scaffolds alone cannot recapitulate the necessary physiologic

environment. Thus, for in vivo work, the other two Rs, “Reduce” and “Refine”, inform the

planning of humane animal studies. These two Rs help to ensure that the number of animals

used is minimized, and that animals experience the least amount of pain possible in a study

and are housed and cared for according to established guidelines [28].

Reduction entails both making use of only the best animal model and determining

the optimum number of animals needed to achieve significance in results. For example, in

a muscle injury model it would be unwise to test a large number of TESM interventions in

small groups of animals, as this could create additional “noise” in an experiment, impeding

determination of efficacy for any particular intervention. Refinement entails not only

ensuring adequate analgesia for all animals used, but also ensuring that the use of animals

is parsimonious and targeted. This might include conducting in vitro studies and studies

319
with smaller animals (e.g., rodents) to develop techniques for injury production and

application of TESM prior to use in a large animal model (e.g., sheep or pigs).

Because TESM is a complex technology with cellular, biomaterial, and bioreactor

components, all of which must be fully functional for success, the risk profile may be

heightened, as there are several risks of harm associated with each component. Ensuring

modest TD requires demonstrating through pre-clinical evidence that these risks are

manageable. Species to species differences must be ruled out as much as possible when

moving from animals to humans. Ensuring modest TD is particularly difficult in this

context. Although volumetric muscle loss in human patients is not standardizable, creating

a “spontaneous” and unique injury in animal models through surgically induced irregular

trauma or blasts is, difficult to justify in both regulatory and ethical terms. Thus, it is

reasonable to create more uniform and less traumatic defects in animal models, especially

in non-human primates, both for ethical reasons and to establish proof of concept. Even

so, it will be challenging to gather generalizable data because there will always be

irreducible differences in injuries, both across animals and between animals and human

patient-subjects.

Additional considerations when matching animal models of muscle pathologies to

humans include (1) developing a model in which the untreated animals will recover (or not

recover) in the same manner/timeline as the clinical presentation of the particular injury

and (2) matching the muscle fiber type and anatomy in the animal model site to the fiber

type or fiber orientation planned for the clinical application. Studies should also include

both male and female animals of varying ages. Finally, if cells are used, the same cell

source or an equivalent source should be appropriate and planned for human trials.

320
For maximal knowledge generation, animal studies of TESM using an injury model

require proper controls. A negative control for TESM animal studies entails use of a no-

repair group in which an injury is created and allowed to heal without surgical intervention.

This is necessary for muscle injury models in order to study the extent to which the

intervention enables repair and regeneration within the injury site. Positive controls -- the

use of an uninjured control group or treatment with autologous and allogeneic flap transfers

– are also necessary in order to begin determining the effectiveness of the TESM strategy

under study relative to the current standard of care. The ultimate goal of this line of research

is to determine whether TESM can replace flap transfer. In order to show whether TESM

can overcome the problems of graft failure associated with cadaveric transplant and the

cosmetic impairment caused by resection of the patient’s own tissue, clinically relevant

animal studies involving TESM need appropriate controls.

Finally, reducing TD also means ensuring data transparency and both internal and

external validity. Internal validity [7, p.119] is a measure of the strength of causal

inferences arising from trial data, and thus of the predictive power of a trial. Ways to

improve internal validity include randomization and limiting variability within and across

arms (even when blinding is not possible). For TESM studies, one option is ensuring the

ability to repeat the work done previously within the same lab group, by maintaining good

lab notes. Turnover of students, post-doctoral fellows, and lab technicians can hinder this

process. External validity is most fundamentally challenged by the inherent divergence

between animal models and human subjects, and can best be addressed through close

correspondence in study designs and goals, increased use of pilot and feasibility studies in

FIH settings, transparency, and minimization of bias [7, pp. 120-121]. This may be

321
accomplished by making use of a third party research group such as a CRO to repeat some

of the research in order to prove efficacy in animal studies. Finally, pre-clinical data should

be published, so that scientists can learn from one another and further the field of their

research in general. Failures of transparency increase the likelihood that a technology with

low odds of ultimate success will make it to clinical trials.

2.3. Research ethics in early-stage human trials

Every clinical trial should produce results that are able to guide researchers in

designing the next study. Because the ultimate goal of medical research is to improve

clinical practice, all trials should be designed for both validity and value. Trial design and

conduct must also address protections for research subjects: Risks of harm to subjects

should be clearly identified and minimized, and the potential, or lack thereof, for direct

benefit must be addressed; this includes defining success and failure, and examining what

can be done for patient-subjects if/when the experimental intervention fails. Addressing

these considerations in first-in-human and early-stage TESM research may pose special

challenges.

2.3.1 Patient-subject selection and informed consent

Fair and appropriate subject selection is a key component of scientifically and

ethically sound clinical trial design. Should the first subjects in a TESM construct study be

patients with extensive injuries, or with small defects? What control groups are most

appropriate? Early-stage TESM research in humans will necessarily focus only on repair

of injuries and defects of relatively small volumes, primarily because of the need to develop

or provide a vascular supply. Vascularization is an essential aspect of TE research that

requires further development [29]. Consideration might also be given, however, to

322
enrolling patients with more extensive injuries, if proof of concept data could be gathered

prior to standard repair or amputation. Thus, early-stage TESM research might also be

undertaken in patient-subjects with more extensive injuries if there is sufficient time to do

so before standard treatment with cadaveric muscle or flap transfer is attempted. It would

be critical to design and conduct such studies so that (1) standard treatment would remain

available, (2) delaying standard treatment would be minimally disadvantageous to patient-

subjects, and (3) a clear and robust consent process can be undertaken so that patient-

subjects understand the study design and the harm-benefit balance and can make free and

informed decisions about participation. In addition, it might be useful to collect data from

patient-subjects who choose not to have any repair. In early-stage TESM trials where

vascular supply does not pose a barrier, randomization of patient-subjects who would

otherwise choose standard treatment to no-treatment or sham surgery is not ethically

justifiable when the design requires withholding reasonably effective standard treatment

[14,30,31].

A patient who volunteers to take part in a clinical trial should know first and

foremost that he or she is a research partner who, by joining the study, will further scientific

research aimed at advancing the field regardless of the outcome. Potential

patient-subjects must be told that the goal of the study is to find out if TESM technology

can restore function and improve appearance in the area of muscle injury. Outcomes for

patient-subjects are unknown and will vary. Research is not treatment, but rather an

investigation into a possible solution. In TESM research, the need to seek consent to

participation from patients with recent traumatic injuries may make it more difficult to

convey essential information effectively. And even well-informed potential subjects may

323
still believe strongly that in spite of the risk of failure, TESM is sure to work for them.

Such patient-subjects may be expressing acceptable optimism, or may be significantly

underestimating the risks of harm, overestimating potential benefits, and mistakenly

viewing research as treatment – that is, some may be affected by the therapeutic

misconception (TM) [21,32-36]. TM can often be minimized by careful discussion and

expectation management with patients who are potential subjects during screening and

interviews prior to enrollment, as well as continuing discussion during trial participation.

A high degree of attention to informed consent for patient-subjects with recent injuries will

be essential in TESM research [37].

In FIH trials of TESM interventions, there are several important “unknowns.”

Injuries created in animals will not be the same as the injuries in patients: anatomical

differences will be present, and the injuries will be of different geometries and complexities.

In general, every injury is uniquely complex. Thus, researchers in FIH TESM trials must

identify (and then explain to potential subjects) what is unpredictable and unknown. This

is true regardless of study design, as it applies to pilot and feasibility studies as much as it

does to trials with more than one arm.

In translating tissue engineering research like TESM to human patient-subjects, the

risks include not only the risks of harm associated with all surgical procedures, such as

infection at the implant site, but also the risk of failure to meet patient expectations. TESM

is a permanent alteration to the patient-subject’s body, which is only reversible by

additional surgery [5]. Potential subjects must be told that if the experimental intervention

were to fail, an additional surgery would be needed to correct the defect, possibly entailing

replacement of the experimental implant with a standard flap transfer, which poses all the

324
risks of additional surgery and potentially adds the risk of rejection of the flap. Moreover,

a TESM graft might not be physically rejected but might nonetheless fail to meet the

patient-subject’s expectations about appearance or functional recovery. To mitigate

dissatisfaction with a TESM repair outcome, the patient-subject’s expectations should be

addressed and discussed before enrollment – a process similar to standard consent practice

in plastic and reconstructive surgery.

2.3.2. Success and failure in TESM

Several metrics could be used to assess the degree of success achieved in a TESM

study. For repairs involving load-bearing muscles such as those in the extremities, function

could be analyzed through electromyography or more simply through physical strength

tests administered by a physical therapist. Another important metric is the appearance of

the treated muscle after surgery. Improving the aesthetic quality of muscles in the face and

neck or the extremities can greatly improve self-confidence and quality of life, even if the

muscles are not fully functional. Assessment of personal qualitative metrics should be

conducted through careful interviews with patient-subjects before and after surgery. These

measurements are likely to vary in importance from person to person, and for each person,

some bodily characteristics may be more important than others. Before beginning a clinical

trial, these metrics must be clearly defined and perhaps ranked by patient-subjects before,

during, and after the process of surgery and recovery. For some patient-subjects who may

have experienced trauma associated with their muscle loss injuries, mental health and

wellbeing should be especially carefully evaluated to help determine the impact of TESM

on overall quality of life.

325
The above considerations suggest that human clinical trials of a TESM construct

should explicitly interrogate the meaning of success. What counts as success is likely to

depend in large part on the preferences and values of patient-subjects, and thus may vary

considerably between individuals. Success, as defined by the goals and expectations of

patient-subjects, could focus on restoring appearance associated with damaged muscle, or

on recovering muscle function to varying degrees, from simple weight-bearing to returning

to competitive athletics. In this respect, research outcomes in TESM are more “patient-

centered” [60] than is typical in many trials. Researchers should discuss realistic

expectations with potential subjects, learn what patient-subjects seek, and consult with

them both immediately after the intervention and as a matter of long-term follow-up to

evaluate quality of life and assess success. Functional outcomes can be identified and

physiologically relevant outcome measures can be applied, but if, ultimately, patients are

not satisfied with their outcomes, TESM was not successful. Similarly, patients who are

satisfied with a non-functional repair, perhaps due to the restoration of desired appearance,

will deem TESM successful. As success is ultimately crucial in all translational research,

investigators and subjects must talk together about what success means in TESM research

on an individual basis.

Of course, well-designed translational research has value even when ultimately

unsuccessful. Failure in a well-designed FIH TESM study supports investigating potential

reciprocal, iterative, or collateral value. Reciprocal value could be finding out that patient-

subjects with a pre-existing condition react differently from other patient-subjects, leading

to a preclinical TESM study done in animals with that condition. Procedural nuances

necessary for success may present iterative value in FIH trials. Surgical techniques may be

326
refined extensively from one patient-subject to the next in a single protocol [38], and many

other in-trial procedural modifications (e.g. special post-operative treatment, a specific

exercise, bed rest protocol, or specialized physical therapy) may also be developed. Finally,

exposing more surgeons and specialists to TESM is by itself a major foreseeable collateral

value. For instance, muscle scaffolds may be discovered to have new, unanticipated

surgical applications. Thus, research that can identify potential reciprocal, iterative, and

collateral value can respond productively to apparent failure, providing hope that the

research may continue and return to additional clinical trials.

3. Case Study 2: Ethical considerations in the bioartificial pancreas (BAP)

3.1. Introduction to the technology

The BAP is in development as an alternative to standard treatments for Type 1

diabetes. Standard treatments include daily insulin injections, continuous infusion insulin

pumps, pancreas transplantation, and clinical islet transplantation. Research into islet cell

microencapsulation technologies began as early as 1980, in order to overcome the need for

immunosuppression that accompanies transplantation of allogeneic cells, tissues, and

organs. The BAP utilizes either insulin-producing β-cells or aggregates of these cells which

are known as islets. These cells are incorporated into either a macro- or microencapsulating

device, and then implanted into the body for the regulation of blood glucose levels. Several

BAP models have been studied; one key feature of current BAP research addresses optimal

implantation sites, which include the peritoneal cavity and the omentum [39,40]. There

are many similarities between the ethical considerations arising in TESM and the BAP

during pre-clinical research and human trials, but the BAP also poses several ethical

327
considerations that are meaningfully different from those in TESM. Type 1 diabetes is a

chronic condition, with several alternative treatment options. Because of the multiple

treatment options available for Type 1 diabetes, the performance of the BAP comes under

additional scrutiny—the BAP needs to alleviate the long-term comorbidities of Type 1

diabetes and avoid both hypoglycemic and hyperglycemic events. If the BAP is unable to

improve upon the treatment provided through exogenous insulin injection or the use of an

insulin pump, it will not be a viable treatment alternative. The BAP must also ultimately

prove functional over the long-term. Thus, questions arise about the acceptable balance of

harms and benefits and the meaning of success and failure for this tissue engineering

technology, as they do for TESM, but the answers are likely to be different.

3.2. Research ethics in the lab

The same general concerns about data integrity, transparency, and anticipation of

translation to animal and human studies discussed in Case 1 apply to BAP research.

Animal study of the BAP is farther advanced than is TESM research, but different aspects

of BAP research are at very different stages. As always, it is important to follow good

laboratory practices and maintain a clear and transparent record of all data in order to

identify risks of harm that may be associated with a potential future therapy and to avoid

redundant or repetitive studies. Several additional issues unique to the BAP also arise at

the earliest stages of bench research.

3.2.1. In vitro

Most BAPs utilize whole islets; thus, the technology requires researchers to work

with primary cell lines, which are in limited supply and can only be cultured or stored for

a finite amount of time. This feature has led to an important ethical consideration in the

328
laboratory: islets can be maintained in vitro for only a limited time before transplantation,

and there can be significant batch to batch variation between islet isolations. It is thus

essential that the tissue be evaluated as thoroughly as possible in vitro prior to

transplantation, and it is problematic when in vitro evaluation is not sufficient. Some of the

techniques commonly used are insufficient to evaluate both the functionality and the

viability of the islet cells in culture [41]. Utilizing the most appropriate in vitro tests

includes identifying those that are as close to clinical tests as possible, to minimize TD.

For the BAP, this means using in vitro tests that have been shown to strongly correlate with

transplant outcomes, such as tests that measure the metabolic activity or insulin secretion

rates of islets as opposed to more traditional live-dead viability stains [42,41].

The use of more predictive in vitro tests is especially important when considering

the three “R’s” of animal research when moving to animal models: reduction, refinement

and replacement. Ideally, the use of more accurate in vitro tests should reduce the number

of animals or groups needed when research progresses to in vivo studies.

A second issue specific to the BAP also relates to the limited availability of human

cadaveric pancreases. The urgent need for alternative tissue sources affects BAP research

from the earliest stages, because it gives rise to an important ethical issue surrounding

BAPs: the use of non-human tissue in humans. Using xenografts, most often from porcine

tissue, requires a very high level of awareness and vigilance with regard to the risks and

safeguards associated with non-human tissues [43-45]. Of chief concern with porcine cells

is the transfer of zoonotic disease like porcine endogenous retrovirus (PERV) [46]. As a

result of these differences, the risks of harm and potential benefits for the BAP, at every

329
stage of research and ultimately in the clinic, are significantly different from those for

TESM.

3.2.2. In vivo

Identification of the appropriate small animal model and the use of proper controls

for the chosen animal model are important considerations in BAP research, in order both

to conform to the three Rs and to reduce TD. As with TESM, it is especially important to

use a small animal model that best represents the clinical disease. In the case of a BAP, it

is important to consider whether it is preferable to utilize a knockout model (where the

disease develops spontaneously) or an induced model for diabetes (where the animal is

given a chemical such as streptozotocin to cause disease). It is also important to consider

the appropriateness of using an immunocompromised or an immunocompetent animal

model. Using the former concentrates attention on factors other than the immunoprotection

afforded by the BAP, thus, for example, facilitating research on vascularization, which is

as essential for success of the BAP as for TESM. However, using immunocompromised

animals may increase the number of animal studies necessary before moving into humans

to test a BAP with an immunoprotective coating.

The most appropriate animal models in BAP research will always include large

animals. Although large animal models are not necessarily required for all TESM, large

animal models are required for islet cell transplantation because diabetic conditions in

rodents are significantly different from those in humans [47]. When using large diabetic

animal models for BAP studies, the reproducibility of the model is a critical factor. It is

also important to consider the clinical relevance of the insulin therapy given to controls.

330
In the case of the BAP, this means identifying the most appropriate and accurate protocol

for insulin therapy, in order to maintain maximal translational relevance to the clinic.

Like TESM research, animal studies of the BAP are complex, often involving

multiple surgical procedures, to induce diabetes and to implant the BAP. Especially in

research involving creation of an omentum pouch to improve vascularization of the BAP

it is essential to determine how best to maintain study protocol integrity while allowing for

the flexibility needed to improve surgical techniques over the course of the study.

Several considerations unique to the BAP also apply to design of animal studies.

Studies of a BAP that utilizes immunoprotective coatings to prevent immune rejection,

must be able to demonstrate that these coatings remain intact over the course of the study.

It is also important to adhere to clinically relevant islet acceptance criteria [48,49] in

constructing and using the BAP in vivo.

Determining the point at which the research is ready for human trials raises several

provocative considerations in BAP research that differ from TESM research. First,

determining the outer limits of the harm-benefit balance in BAP requires special attention

to ethical and design questions in animal studies before translation to human studies. One

issue is the role of immunosuppressants in BAP research. Ideally, the BAP would not

require the use of immunosuppressants, because long-term immunosuppressant use poses

risks that could outweigh the BAP’s benefits. Thus, the most desired endpoint of BAP

research is an encapsulated device that can restore a sufficient degree of pancreatic function

and that has an immunoprotective coating that is effective over the long-term. However,

studying the use of immunosuppressants with the BAP could ultimately facilitate the earlier

clinical availability of devices that could benefit seriously ill patients, for whom the

331
increased risk might be a reasonable choice under the circumstances. Thus, two very

different clinical applications may follow from research designed for different patient

populations with different options and needs.

Along the same lines, because there are different BAPs, there are different harm-

benefit assessments that can lead to different conclusions. Some BAPs carry higher risks

but are viewed as acceptably risky for broad clinical use. BAPs that make use of xenografts

are risk-bearing due to the concerns associated with the use of porcine or other non-human

tissues, in particular zoonotic disease, but the limited availability of human tissue makes

research focused on the use of porcine tissues reasonable under current circumstances.

Thus, research has proceeded using both allografts and xenografts in non-

immunosuppressed animals [40]. In contrast, however, some types of BAP constructs are

now considered unacceptably risky. BAP constructs that connect directly to the circulatory

system are riskier because there is a higher chance of clot formation and rejection.

Additionally, these devices cannot be retrieved or removed as easily as devices that are

simply implanted. These lines of research have been supplanted as more has been learned

about the inherent risks associated with these approaches [40].

Additional factors to consider when determining the point at which a BAP is ready

for human trials include study duration, size, and reproducibility. As with TESM studies,

one method to externally verify results is the use of a CRO. Challenges here include

establishment of the diabetic animals, technology transfer, and the training of CRO staff

[46,50,51].

Finally, when planning in vivo studies it is necessary to consider how they can be

designed to better identify the causes of failure if they are unsuccessful at achieving insulin

332
independence. The long history of BAP research includes many examples of research that

has provided important iterative value (e.g. in-trial surgical modifications), reciprocal

value (e.g. research focusing on better vascularization to improve the longevity of the BAP

and research utilizing different islet cell sources), or collateral value (e.g. development of

BAPs intended only for limited patient populations, such as those for whom long-term

immunosuppression does not pose a significant risk), even though the ultimate goal of BAP

research has not yet been reached.

3.3. Research ethics in early-stage clinical trials

As compared to TESM, the BAP has one distinct advantage when moving into

clinical trials, and this is the historical precedent for the use of islet cell transplantation.

For BAP, unlike TESM, there is both a network of islet transplant centers and a set of

published donor selection criteria, as well as established guidelines for involvement in an

islet transplant study [48,49]. In this respect, BAP research benefits from the precedent

and expertise of related biotechnologies -- that is, from the collateral value generated by

prior research. BAP researchers can also make use of existing infrastructure and guidance

to help design and conduct trials that minimize risks of harm, maximize the production of

generalizable knowledge, and fully inform potential subjects.

3.3.1 Patient-subject selection and informed consent

Another difference from TESM appears in the selection of patients as research

subjects. Several populations of diabetic patients who are potential subjects for BAP

research have characteristics that put them at greater risk but also may make them better

candidates for some early-phase designs. One group, mentioned previously, is seriously

ill patients, whose disease course and anticipated longevity might in some circumstances

333
make them suitable candidates for BAP designs requiring immunosuppression. Another

such population is patients who have reduced awareness of hypoglycemia. This is a

dangerous condition [52]; consequently, these individuals may be candidates for early

clinical trials involving either islet transplantation or the BAP, since current insulin

therapies are inadequate for them. For populations like these, early-stage research

involving patient-subjects with diabetes most resembles Phase I oncology research, where

patients are chosen as subjects because there are no adequate standard treatments currently

available to them. Unfortunately, TM can be exacerbated in such trials, as it is tempting to

move from “nothing else works well for you” to “this research is your best hope” even

though direct benefit is very unlikely. As always, a clear and complete consent process is

the best way to avoid TM when patients are enrolled as research subjects under such

circumstances.

As trial designs change and scientific thinking about best subject populations also

changes, it may be worth speculating about how BAP research might be regarded if it were

thought that better data could be gathered from young, treatment-naive patient-subjects

than from patient-subjects for whom nothing else has proven effective. A contemporary

parallel might be found in current research using a new smartphone-based application to

regulate insulin pump delivery based on continuous data readouts [53]. In this Type 1

diabetes intervention, a sensor is used to monitor blood sugar in the patient-subject, and

blood sugar levels are managed through a dual pump to supply insulin (to lower blood

sugar levels) and glucagon (to prevent blood sugar level drops) to achieve normoglycemia.

Some patient-subjects in ongoing trials are teenagers, which is a departure from the

traditional adults-first model. This choice of subject population more closely resembles

334
the most likely patient-subjects in FIH trials of TESM constructs: younger patients with

smaller injuries or defects. Determining which patients are the best subjects in FIH trials,

especially when the potential subject populations are as different as very young, treatment-

naive patient subjects and adult patient-subjects for whom no adequate alternatives exist,

represents an extremely important ethical and design challenge. Which patient-subjects

should be first is an ongoing issue in gene transfer research and many other novel

biotechnologies as well [2,28].

3.3.2. Xenografting and risk in the BAP

One of the most discussed issues involving the BAP, which does not arise in TESM

research, is the use of xenografts. As previously mentioned, the use of porcine islets is one

solution to the shortage of available human donor islets. However, the use of xenografts

dramatically increases the risks associated with the BAP. Of particular concern is the risk

of spreading zoonotic diseases. As a result of this increased risk, there has been a

significant amount of research focused on creating disease-free pigs, with a particular focus

on pigs free of PERV. In response to the increased research into xenografts, the

International Xenotransplantation Association has published a consensus statement [54],

which reviews the key ethical requirements of an international regulatory framework for

clinical trials of xenotransplantation, the recommendations on source pigs, release criteria,

and necessary preclinical data required to justify a clinical trial, strategies to prevent the

transmission of PERV, patient selection, and an outline for informed consent [55-58]. Risk

reduction in this area may in the future change what is regarded as an acceptable risk-

benefit balance in BAP human studies and related trials.

3.3.3. Success, failure, and the BAP

335
For the BAP to become a successful treatment, it has to function better than insulin

therapy with its attendant complications. Yet what that means in terms of defining success

in BAP research may not be as clear-cut as it seems. On the one hand, it could be

reasonable to expect that the BAP should ultimately prove a genuine cure. On the other

hand, what if the BAP transforms what can be described as a progressively fatal disease

into a chronic disease? If partial functional correction is an acceptable goal, then a BAP

might be introduced into the treatment armamentarium as another useful “halfway

technology” that is not a cure. BAPs that meet this goal of partial functional correction

would reduce the need for insulin therapy and thus reduce the occurrence of hypoglycemic

unawareness and stabilize or reduce the comorbidities of Type 1 diabetes.

If the BAP is likely to reduce but not eliminate the need for exogenous insulin, then

the preferences and perspectives of patient-subjects may be highly relevant in determining

what counts as success for the BAP, even though there is no aesthetic component as in

TESM. Because Type 1 diabetes is a chronic and progressive condition that begins to

change most patients’ lives at a relatively young age, potential subjects may be more

knowledgeable and under less decision-making pressure than in TESM research, where

most potential subjects will have recently experienced a wide variety of muscle injuries

and loss. Thus, potential subjects for BAP research may consider the impact of different

research outcomes on their decisions to participate and their ultimate satisfaction from a

very different emotional and psychological vantage point. Moreover, the consequences of

partial success—reducing but not eliminating the need for exogenous insulin and the

effects of the many comorbidities of Type 1 diabetes – are quite different from those

potentially faced by TESM subjects.

336
4. Discussion and conclusions

4.1. Strengths and limitations

By examining in some detail two very different examples of tissue engineering

research, we have been able to focus in-depth on some key issues that are not as carefully

reviewed elsewhere. However, a case-based review cannot address all of the many relevant

ethical issues arising in tissue engineering research.

The use of cells and cell lines has been thoroughly examined elsewhere [1,5,16].

So has the problem of access and cost, which is of considerable concern for all novel

biotechnologies [1,2,16]. The overall goals of tissue engineering include not only

developing treatments that are more effective than currently available alternatives, but also

keeping costs low and improving access. The complexity of tissue engineering

interventions represents a challenge to achievement of this goal, but it is nonetheless one

toward which the field strives [59].

4.2. Key lessons learned

Several key ethical issues emerge from these case studies. First, it is essential to

look for iterative value in tissue engineering research. Because tissue engineering research

methods are complex and mixed, including surgery, cells, and a wide range of enabling

technologies in animal and human trials, innovations are almost inevitable. Researchers

should anticipate the likelihood of innovation, and should design and describe protocols

that can not only accommodate innovations but also convert them into study features when

warranted [15,60]. Clear and close communication with research oversight bodies is

therefore essential, both to ensure the proper balance of flexibility and reproducibility in

337
tissue engineering protocols and to amend protocols when amendment is necessary to

capture methodological innovations.

Second, data transparency is critical, even though it is increasingly difficult to

achieve in the current highly competitive funding environment. For tissue engineering

research, it is particularly vital that researchers publish data at all stages, especially data

from early clinical trials, regardless of success or failure. Being transparent about the

research strategy and results of the trial enables scientists at earlier stages of development

to see what is and is not working with the model being used. This will fuel and inspire

future work and advance the field as a whole [61,62]. In addition, making the results of

these trials publicly available and understandable to the lay public will inform those

considering participation in such trials and help to set the stage for understanding the

benefits and limitations not only of trial participation but also of any future treatments and

new research directions that may later emerge from the research.

Third, much work is needed to increase parsimony in animal research while

identifying best animal models and designing effective adjuncts and alternatives to use of

animals. As noted earlier, body-on-a-chip and organ-on-a-chip research shows great

promise in this regard.

Fourth, it may be time to abandon FDA’s traditional phase designations in order to

take better account of novel trial designs and the increasingly nuanced considerations

influencing subject selection. Like Phase I oncology trials and most if not all FIH and other

early-stage clinical trials of novel biotechnologies, early TESM and BAP clinical trials

necessarily enroll patients as subjects, instead of the “healthy volunteers” traditionally

enrolled in Phase I drug trials, and may at times begin with older or younger subjects, and

338
experienced or treatment-naive subjects. Clear justification for a clinical trial’s design and

goals is paramount regardless of phase designation, and thorough consideration of the

implications of enrolling patients as subjects, including but not limited to the consent form

and process, are essential. A related challenge arises when complex tissue engineering

intervention trials cannot effectively or ethically be randomized and blinded. Patients who

are potential subjects may have strong and reasonable preferences for one arm over another,

as their goals and quality of life assessments may differ in ways that significantly affect

their choices about participation. It may be prudent to seek new ways to gather important

information in research that does not culminate in “gold standard” randomized controlled

trials. Much attention is now being given to “patient-centered outcomes” in clinical

research [63,64]. Incorporating patients’ perspectives into research could be well-suited to

some tissue engineering trials, and could increase enrollment.

Methodological challenges will inevitably arise in new trial designs, including not

only statistical design and data credibility but also a potential increase in the likelihood of

TM in investigators, the media, the public, and patient-subjects. These challenges can be

effectively addressed with clear and transparent justification for each step along the

research pathway, strong preclinical data, exemplary communication, and the creativity

that arises from collaborative scholarship along the translational pathway. The result could

yield intervention profiles that are more predictive of outcomes in clinical practice.

Finally, not only is long-term follow-up essential, but in addition, researchers can

play a critical role in facilitating translation to clinical care for these complex tissue

engineering interventions. Clinical use of cell-seeded scaffolds and capsules in TESM and

BAP will continue to pose challenges even after successful research. One often overlooked

339
issue is the infrastructure needed for feasible and efficient tissue engineering treatments.

For instance, hospital personnel would need to be trained in handling cell-scaffold products

and hospitals would need to affiliate with a Good Manufacturing Process facility for

generation of the constructs [65]. Several biomaterial scaffolds have been approved by the

FDA for a variety of applications, including rotator cuff repair, hernia repair, and heart

valve patches [66]. Familiarity with these devices may facilitate acceptance and use of

tissue engineering technologies in the future, as long as researchers and sponsors plan well

in advance for effective clinical translation.

We encourage detailed examination of additional case examples of research using

tissue engineering and other cell-based interventions. Case-based discussion of ethics and

design questions throughout the translational trajectory is the best way we know of to

promote transparency, encourage discussion, and maximize the knowledge gained from

this important research.

340
Acknowledgments

The authors thank Drs. George Christ and Emmanuel Opara for their review of earlier

versions of this paper. Research for this publication was supported in part by the National

Institute of Biomedical Imaging and Bioengineering of the National Institutes of Health

under Award Number T32EB014836. The content is solely the responsibility of the authors

and does not necessarily represent the official views of the National Institutes of Health

(HBB, JPMcQ).

341
References

[1] N.M.P. King , C. Coughlin, M. Furth, Ethical issues in regenerative medicine.

Wake Forest Intellectual Property Law J. 9 (2010) 215-237.

[2] T. Keys, N.M.P. King, A. Atala, “Faith in Science: Professional and Public

Discourse on Regenerative Medicine.” In: M.J. Hyde, J. Herrick (Eds.), The

Language of Our Biotechnological Future. Baylor University Press 2013, pp.

11-39.

[3] R.B.M. deVries, A. Oerlemans, L. Trommelmans, K. Dierickx, B. Gordijn,

Ethical aspects of tissue engineering: a review, Tiss. Eng. Part B. 14 (2008)

367-375.

[4] I. Hyun, O. Lindvall, L. Ährlund-Richter, E. Cattaneo, M. Cavazzana-Calvo,

G. Cossu, M. De Luca, I.J. Fox, C. Gerstle, R.A. Goldstein, New ISSCR

guidelines underscore major principles for responsible translational stem cell

research, Cell Stem Cell 3(6) (2008) 607-609.

[5] D.A. Taylor, A.L. Caplan, P. Macchiarini, Ethics of bioengineering organs

and tissues, Expert Opin Biol Ther. 14(7) (2014) 879-82.

[6] B. Lo, L. Parham, Ethical issues in stem cell research, Endocrine Revs. 30(3)

(2009) 204-213.

[7] J. Kimmelman, Gene Transfer and the Ethics of First-in-Human Research:

Lost in Translation, Cambridge University Press, 2009.

[8] A.J. London, J. Kimmelman, and M. E. Emborg, Beyond access vs. protection

in trials of innovative therapies, Science 328 (2010) 829-30.

342
[9] S. Joffe, F. G. Miller, Mapping the moral terrain of clinical research, Hastings

Center Rep. 38(2) (2008) 30-42.

[10] M. B. Esch, T. L. King, M.L. Shuler, The role of body-on-a-chip devices in

drug and toxicity studies. Ann Rev Biomed Eng 2011, 13:55-72]

[11] W.M.S. Russell and R.L. Burch,The Principles of Humane Experimental

Technique, Methuen & Co. Ltd, London 1959. Reprint Charles C. Thomas,

Springfield, Illinois, 1960. Reissue, Universities Federation for Animal

Welfare, 1992. Available online at:

http://altweb.jhsph.edu/pubs/books/humane_exp/het-toc

[12] B. Freedman, Equipoise and the ethics of clinical research. NEJM 317 (1987)

141-145.

[13] R. Levine, Ethics and Regulation of Clinical Research, 2nd ed., Yale

University Press, New York, 1988.

[14] E.J. Emanuel, C. Grady, D. Wendler, What makes clinical research ethical? J

Am Med Assoc 283 (2000) 2701-2711.

[15] National Commission for the Protection of Human Subjects of Biomedical

and Behavioral Research, The Belmont Report: Ethical Principles and

Guidelines for the Protection of Human Subjects of Research (April 18,

1979). http://www.hhs.gov/ohrp/humansubjects/guidance/belmont.html. Last

accessed 30 October 2014.

[16] N.M.P. King, J. Perrin, Ethical issues in stem cell research and therapy. Stem

Cell Res Ther. 5 (2014) 85-90.

343
[17] N.M.P. King, Ethics in regenerative medicine and transplantation, in: G.

Orlando, J.P. Lerut, S. Soker, R.J. Stratta (Eds), Regenerative Medicine

Applications in Organ Transplantation. Academic Press Elsevier, 2013, pp.

977-986.

[18] N.M.P. King, Early-stage research: Issues in design and ethics, in: L. Hogle

(Ed), Regenerative Medicine Ethics: Governing Research and Knowledge

Practices, Springer-Verlag, 2014, pp. 187-204.

[19] N.M.P. King, Defining and describing benefit appropriately in clinical trials,

J. Law Med. Ethics 28 (2000) 332-343.

[20] N.M.P. King, O. Cohen-Haguenauer, En route to ethical recommendations for

gene transfer clinical trials, Mol. Ther. 16 (2008) 432-438.

[21] N.M.P. King, G.E. Henderson, L.R. Churchill, A.M. Davis, S.C. Hull, D.K.

Nelson, P.C. Parham-Vetter, B.B. Rothschild, M.M. Easter, B. S. Wilfond,

Consent forms and the therapeutic misconception: The example of gene

transfer research. IRB: Ethics Hum. Res. 27:1 (2005) 1-8.

[22] T. Caulfield, Biotechnology and the popular press: hype and the selling of

science. Trends Biotechnol. 22 (2004) 337-339.

[23] M.A. Machingal, B.T Corona, T.J. Walters, V. Kesireddy, C.N. Koval, A.

Dannahower, W.X. Zhao, J.J.Yoo, G.J. Christ, A tissue-engineered muscle

repair construct for functional restoration of an irrecoverable muscle injury in

a murine model. Tiss. Eng. Part A, 17, (17-18) (2011), 2291-230.

344
[24] K. Donnelly, A. Khodabukus, A. Philip, L. Deldicque, R. Dennis, K. Baar, A

Novel Bioreactor for Stimulating Skeletal Muscle in Vitro. Tiss. Eng. Part C,

16 (4), (2010) (711-718)].

[25] B.T. Corona, E.M. Balog, J.A. Doyle, J.C. Rupp, R.C. Luke, C.P. Ingalls,

Junctophilin damage contributes to early strength deficits and EC coupling

failure after eccentric contractions. Am. J. Physiol-Cell Physiol. 298 (2)

(2010), C365-C376.

[26] S.K. Yazdani, B. Watts, M. Machingal, Y.P.R. Jarajapu, M.E. Van Dyke, G.J.

Christ, Smooth muscle cell seeding of decellularized scaffolds: The

importance of bioreactor preconditioning to development of a more native

architecture for tissue-engineered blood vessels. Tiss. Eng Part A. 15(4),

(2009) 827-840.

[27] M.R. Abraham, C.A. Henrikson, L. Tung, M.G. Chang, M. Aon, T. Xue, R.A.

Li, B. O'Rourke, E. Marban, E. Antiarrhythmic engineering of skeletal

myoblasts for cardiac transplantation. Circ. Res. 97(2), (2005) 159-167.

[28] National Research Council. Guide for the Care and Use of Laboratory

Animals: Eighth Edition. Washington, DC: The National Academies Press

(2011).

[29] M. Lovett, K. Lee, A. Edwards, D.L. Kaplan, Vascularization Strategies for

Tissue Engineering. Tiss. Eng. Part B, 15(3), (2009) 353-70.].

[30] S. Horng, F.G. Miller, Ethical framework for the use of sham procedures in

clinical trials. Crit Care Med 31, (2003) S126-130.

345
[31] R. Macklin, The ethical problems with sham surgery in clinical research.

NEJM 341 (1999) 992-995.

[32] P.S. Appelbaum, L.H. Roth, C. Lidz. The therapeutic misconception:

Informed consent in psychiatric research, Int’l J Law Psychiatry 5, (1982)

319-329.

[33] S. Horng, C. Grady, Misunderstanding in clinical research: Distinguishing

therapeutic misconception, therapeutic misestimation, and therapeutic

optimism. IRB 25(1), (2003) 11-16.

[34] K.P. Weinfurt, D. Sells, L. Lin, D.P. Sulmasy, A.B. Astrow, H.I. Hurwitz,

R.B. Cohen, N.J. Meropol, Research participants, high expectations of benefit

in early-phase oncology trials: are we asking the right question? J Clin Oncol,

30, (2012) 4396-4400.

[35] G.E. Henderson, L.R. Churchill, A.M. Davis, C. Grady, S. Joffe, N. Kass,

N.M.P. King, C. W. Lidz, F.G. Miller, D.K. Nelson, J. Peppercorn, B.B.

Rothschild, P. Sankar, B.S. Wilfond, C. Zimmer, Clinical trials and medical

care: Defining the therapeutic misconception, PLoS Med. 4 (2007) 1735-

1738.

[36] R. Dresser, The ubiquity and utility of the therapeutic misconception. Soc.

Phil. Pol’y 19 (2002) 271.

[37] J. Illes, C. Reimer, B. K. Kwon, Stem cell clinical trials for spinal cord injury:

readiness, reluctance, redefinition. Stem Cell Rev 2011, 7:997-1005..

346
[38] A. Atala, S.B. Bauer, S. Soker, J. Yoo, A.B. Retik, Tissue-engineered

autologous bladders for patients needing cystoplasty. Lancet 367 (2006) 1241-

1246.

[39] G. Reach, Bioartificial Pancreas, Diabetic medicine 10 (1993) 105-109.

10.1111/j.1464-5491.1993.tb00025.x, 1993

[40] E.C. Opara, S.H. Mirmalek-Sani, O. Khanna, M.L. Moya, E.M. Brey, Design

of a bioartificial pancreas, Journal of investigative medicine: the official

publication of the American Federation for Clinical Research. 58 (2010) 831.

[41] V. Boyd, O.M. Cholewa, K.K. Papas. Limitations in the use of fluorescein

diacetate/propidium iodide (FDA/PI) and cell permeable nucleic acid stains

for viability measurements of isolated islets of langerhans, Curr Trends

Biotechnol Pharm. 2 (2008) 66-84.

[42] M. S. Hanson, E.E. Park, M.L. Sears, K.K. Greenwood, J.S. Danobeitia, D.A.

Hullett, L.A. Fernandez, A simplified approach to human islet quality

assessment, Transplantation. 89 (2010) 1178.

[43] M. Sykes, M. Sandrin, A. D'Apice. Guidelines for xenotransplantation,

NEJM. 349 (2003) 1294-1295.

[44] M. Sykes, M. Sandrin, E. Cozzi, International cooperation on

xenotransplantation, Nature Med. 10 (2004) 119.

[45] M. Sykes, A. D’Apice, M. Sandrin, I.E. Committee, Position paper of the

ethics committee of the International Xenotransplantation Association,

Transplantation. 78 (2004) 1101-1107.

347
[46] R. Valdes-Gonzalez, L.M. Dorantes, E. Bracho‐Blanchet, A. Rodríguez‐

Ventura, W. DJG, No evidence of porcine endogenous retrovirus in patients

with type 1 diabetes after long‐term porcine islet xenotransplantation, J Med.

Virol. 82 (2010) 331-334.

[47] D. Rees, J. Alcolado, Animal models of diabetes mellitus, Diabetic medicine.

22 (2005) 359-370.

[48] U.S. Department of Health and Social Services, Food and Drug

Administration, Center For Biologics Evaluation and Research. Guidance for

Industry: Considerations for Allogeneic Pancreatic Islet Cell Products (2009).

http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatory

Information/Guidances/CellularandGeneTherapy/ucm182440.htm Last

accessed Nov. 4, 2014.

[49] F. B. Barton, M.R. Rickels, R. Alejandro, B.J. Hering, S. Wease, B.

Naziruddin, J. Oberholzer, J.S. Odorico, M.R. Garfinkel, M. Levy,

Improvement in outcomes of clinical islet transplantation: 1999–2010,

Diabetes care. 35 (2012) 1436-1445.

[50] H.J. Schuurman, Regulatory aspects of pig‐to‐human islet transplantation,

Xenotransplantation. 15 (2008) 116-120.

[51] M.A. Clark, This little piggy went to market: the xenotransplantation and

xenozoonose debate, J. Law Med. Ethics. 27 (1999) 137-152.

[52] R. Rood, D. Cooper, Islet xenotransplantation: are we really ready for clinical

trials?, Am. J. Transplantation. 6 (2006) 1269-1274.

348
[53] S.J. Russell, F.H. El-Khatib, M. Sinha, K.L. Magyar, K. McKeon, L.G.

Goergen, C. Balliro, M.A. Hillard, D.M. Nathan, E.R. Damiano, Outpatient

glycemic control with a bionic pancreas in type 1 diabetes, NEJM, 371 (2014)

313-325.

[54] M. Sykes, A. D’Apice, M. Sandrin, The ethics of xenotransplantation:

position paper of the ethics committee of the international xenotransplantation

association. Xenotransplant. 10 (2003) 194-203.

[55] Welsh, R. Evans, Xenotransplantation, risk, regulation and surveillance: social

and technological dimensions of change, New Genetics Soc’y, 18 (1999) 197-

217.

[56] M. Tallacchini, Risks and rights in xenotransplantation, in: Reframing Rights:

Bioconstitutionalism in the Genetic Age. The MIT Press, Cambridge/London,

2011, pp 169-192.

[57] S. Fovargue, Consenting to bio‐risk: xenotransplantation and the law, Legal

Studies, 25 (2005) 404-429.

[58] L. Chapman, Xenotransplantation: public health risks—patient vs. society in

an emerging field, in: Xeno-transplantation, Springer 2003, pp. 23-45.

[59] J.M. Kress, Xenotransplantation: ethics and economics, Food & Drug Law J

53 (1998) 353.

[60] J. Sugarman, Questions concerning the clinical translation of cell-based

interventions under an innovation pathway. J Law Med. Ethics. 40 (2012)

945-950.

349
[61] K. Outterson, Clinical trial transparency – antidote to weaker off-label-

promotion rules? NEJM. 371 (2014) 1-3.

[62] O.M. Dekkers, C.L. Mummery, T.J. Rabelink, A case for crowd sourcing in

stem cell research. Stem Cells Translational Med. 3 (2014) 1259 – 1261.

[63] D.J. Torgerson, B. Sibbald, What is a patient preference trial? BMJ 316

(1998) 360.

[64] Patient-Centered Outcomes Research Institute, “Patient-Centered Outcomes

Research” http://www.pcori.org/content/patient-centered-outcomes-research

Accessed 10/30/14.

[65] R.B. McGoldrick, K. Hui, J. Chang, Bench to Bedside: Integrating Advances

in Basic Science into Daily Clinical Practice. Hand Clinics. 30(3), (2014)

(305-17)]

[66] B. Brown, S. Badylak, Extracellular Matrix as an Inductive Scaffold for

Functional Tissue Reconstruction. Translational Research, 163(4), (2014)

(268-285).

350
APPENDIX 3

Hannah Besser Baker

Department of Biomedical Engineering

Wake Forest University School of Medicine, Winston-Salem, NC 27157

Affiliations: Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences,

Winston-Salem, NC 27157; Wake Forest Institute for Regenerative Medicine,

Winston-Salem, NC 27101

Note: The following figure represents unpublished data used with permission from

KeraNetics, LLC, Winston-Salem, NC 27101

351
Figure 1. Release of IGF-1 and bFGF from Blends of KOS:KTN

352
CURRICULUM VITAE

Hannah Besser Baker, B.S.


Wake Forest Institute for Regenerative Medicine • 391 Technology Way
Winston-Salem, NC 27101
(651) 402-4941 • hbaker@wakehealth.edu • 6/29/2015

Education
WAKE FOREST UNIVERSITY WINSTON-SALEM, NC
Doctor of Philosophy in Biomedical Engineering Anticipated July 13th, 2015
Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences
• Thesis Committee: George Christ, Ph.D. (Chairman), Grace Almeida-Porada, Ph.D., Aaron
Goldstein, Ph.D., Robert Grange, Ph.D., David Kaplan, Ph.D., Aaron Mohs, Ph.D.
• Awards: Wake Forest Institute for Regenerative Medicine NIBIB T32 Fellowship
UNIVERSITY OF ROCHESTER ROCHESTER, NY
Bachelor of Science in Biomedical Engineering May, 2010
Concentration in Cell and Tissue Engineering with Minor in Biology
• Honors: Dean’s List; Bausch and Lomb Scholarship for Outstanding Achievement in
Sciences
• Involvement: Sigma Delta Tau Sorority; Order of Omega Greek Honor Society; UR
Biodiesel Team

Research Experience
WAKE FOREST INSTITUTE FOR REGENERATIVE MEDICINE WINSTON-SALEM, NC
Biomedical Engineering Graduate Student, Christ Lab Fall 2010-Present
• Skeletal muscle research focused on the use of preconditioned naturally derived materials
for repair and regeneration of volumetric muscle loss associated with craniofacial and
extremity injuries of active military personnel and congenital birth defects including cleft
lip and cleft palate

• Projects have included the investigation of cellularized porous aligned silk, cell and growth
factor loaded keratin hydrogels, and cell loaded and bioreactor preconditioned bladder
acellular matrix in rodent injury models of volumetric muscle loss

• Current efforts surround a preclinical study involving the development of a rodent defect
model with clinical relevance to cleft lip and subsequent investigation of tissue engineered
muscle repair therapies in this model for use in cleft lip secondary revision procedures

RADIATION & ONCOLOGY, STRONG MEDICAL CENTER ROCHESTER, NY


Research Assistant to Dr. Walter O’Dell Fall 2009
• Provided programming assistance for research software which aimed to model and
predict brain tumor metastasis from diffusion MRI images in order to optimize radiation
therapy

353
NEW JERSEY INSTITUTE OF TECHNOLOGY NEWARK, NJ
BioMEMs Summer Institute, Research Experience for Undergraduates Summer 2009
• Trained in microfabrication, microfluidics, and soft lithography with emphasis on
biological applications; designed and manufactured a MEMs device which utilized
ultrasound for the examination of osteoporosis
LAKE REGION MANUFACTURING CHASKA, MN
Quality Assurance Intern Summer 2007
• Conducted mechanical tests on medi cal devi ce guidewires and packaging; prepared
validation reports for aging studies; gained experience in product R&D and clean room
protocol

Publications
Benjamin T. Corona, Ph.D., Catherine L. Ward, Ph.D., Hannah B. Baker, Thomas J. Walters,
Ph.D., George J. Christ, Ph.D. Implantation of in vitro tissue engineered muscle repair
(TEMR) constructs and bladder acellular matrices (BAM) partially restore in vivo skeletal
muscle function in a rat model of volumetric muscle loss (VML) injury. Tissue Engineering
Part A 20 (3-4), 705-715. December, 2013

Oral Presentations
• Hannah B. Baker. Growth Factor Loaded Keratin Hydrogels for Treatment of a Sheet-
like VML Injury in Mice. Tissue Engineering and Regenerative Medicine International
Society Americas Annual Conference, Washington, D.C. December 13-16th, 2014.

• Hannah B. Baker. Keratin Hydrogels as a Cell and Growth Factor Delivery Vehicle for
Treatment of Volumetric Muscle Loss. Biomedical Engineering Society Annual Meeting,
San Antonio, TX.
October 22-25th, 2014.

• Hannah B. Baker. Keratin Hydrogels as a Delivery System for the Regenerative


Treatment of Volumetric Muscle Loss. North Carolina Tissue Engineering and
Regenerative Medicine Society Annual Conference, Winston-Salem, NC. October 18th,
2013.

Poster Presentations
• Hannah B. Baker, Catherine L. Ward, Ph.D., Masood Machingal, Ph.D., Benjamin T.
Corona, Ph.D., Jelena Rnjak, Ph.D., David L. Kaplan, Ph.D., George J. Christ, Ph.D.
Porous Aligned Silk Constructs for Treatment of Volumetric Muscle Loss Injury in a Rat
Model. North Carolina Tissue Engineering and Regenerative Medicine Society Annual
Conference. Raleigh, NC. September 10th, 2012.

• Hannah B. Baker, Catherine L. Ward, Ph.D., Benjamin T. Corona, Ph.D., Masood


Machingal, Ph.D., Jelena Rnjak, Ph.D., David L. Kaplan, Ph.D., George J. Christ, Ph.D.
Further development of a tissue engineered muscle repair (TEMR) construct for the
treatment of volumetric muscle loss (VML) injuries. Armed Forces Institute for
Regenerative Medicine All Hands Meeting. St. Pete Beach, FL. February 13-15th, 2012.

354
Skills
• Cell culture: rodent muscle primary cell isolation, aseptic technique, mammalian
cell line maintenance, scaffold seeding, stretch bioreactor sterile assembly and scaffold
loading technique
• Histology: immunohistochemistry, fluorescent and brightfield microscopy, paraffin
and frozen tissue processing and sectioning
• In vivo experimentation: Rodent skeletal muscle surgical defect and implant surgeries
and dissection, rodent post procedural care, in vivo and ex vivo muscle function testing
• Characterization techniques: Scanning electron microscopy, mechanical testing,
calcium imaging
• GMP Training: Standard Operating Procedure and Master Batch Record preparation and
execution
• Applications: Microsoft Office, LabVIEW, GraphPad, Chart, Image J
• Programming Applications: MATLAB, Mathematica

Biomedical Engineering Design Projects


UNIVERSITY OF ROCHESTER ROCHESTER, NY
Biomedical Engineering Senior Design Project: DPN Diagnostics Pedi-Map 2009-2010
• In a team of four students, designed and built diagnostic device prototype to identify and
track peripheral neuropathies in the foot in order to identify regions at risk for ulceration

UR Biodiesel Independent Study 2009-2010


• With a team of students, created a processor to convert waste vegetable oil from campus
dining halls into fuel for campus buses
• Aided in development, day to day processing, training, and guided efforts to ensure
program sustainability

Leadership Experience
WAKE FOREST INSTITUTE FOR REGENERATIVE MEDICINE WINSTON-SALEM, NC
Biomedical Engineering Graduate Student Researcher Fall 2010-Present

• Summer Research Mentor Summer 2012-2014


Served as a mentor to 11 summer scholars and rotating graduate students on guided
projects and facilitated training in histology, cell culture, microscopy, mechanical testing,
and animal care and use procedures

• Invited Speaker at the Shepherd Center of Winston Salem January 16th, 2014
Presented an overview of the history and progress of tissue engineering to a group of
seniors as part of a series offered in collaboration with the Wake Forest Translational
Science Institute Speakers Bureau

• Invited Speaker at Atkins Academic & Technology High School October 25th, 2013
Presented on Biomedical Engineering as an academic pursuit and career path to interested
students as part of a lecture series geared toward young women interested in science and
engineering careers

355
• Healthcare Career Day Tour Presenter October 20th, 2012
• Explained the research of the WFIRM Physiology Lab to 60 touring high school students
and parents interested in learning more about biomedical research and career opportunities

• First Lego League Volunteer, Winston-Salem, NC Fall 2011, 2013


Provided weekly advice and guidance for a team of students as they built and programmed
a robot and challenge course for a competitive robotics exhibition

UNIVERSITY OF ROCHESTER, BIOMEDICAL ENGINEERING ROCHESTER, NY


• Teaching Assistant: Biosystems and Biosignals Spring 2010

UNIVERSITY OF ROCHESTER, CHEMISTRY ROCHESTER, NY


• Teaching Assistant: General Chemistry II 2008-2010

SIGMA DELTA TAU SORORITY ROCHESTER, NY


• President 2009-2010, Treasurer Fall 2008 2007-2010
• Fundraising Chair Spring 2008, Philanthropy Chair, Spring 2008

SIMLEY SENIOR HIGH SCHOOL INVER GROVE HEIGHTS, MN


• Class Valedictorian June, 2006

356

You might also like