144
In vitro and in vivo Growth Factor Delivery to Chondrocytes and Bone- Marrow-Derived Stromal Cells in Cartilage and in Self-Assembling Peptide Scaffolds by Rachel E. Miller B.S. Bioengineering The Pennsylvania State University, 2005 SUBMITTED TO THE DEPARTMENT OF BIOLOGICAL ENGINEERING IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY IN BIOLOGICAL ENGINEERING AT THE MASSACHUSETTS INSTITUTE OF TECHNOLOGY JUNE 2010 © 2010 Massachusetts Institute of Technology. All rights reserved. Signature of Author _______________________________________________________ Department of Biological Engineering May 20, 2010 Certified by______________________________________________________________ Alan J. Grodzinsky Professor of Biological, Electrical, and Mechanical Engineering Thesis Supervisor Accepted by _____________________________________________________________ Peter C. Dedon Professor of Toxicology and Biological Engineering Associate Head, Department of Biological Engineering Chair, Course XX Graduate Program Committee

In vitro and in vivo Growth Factor Delivery to

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: In vitro and in vivo Growth Factor Delivery to

In vitro and in vivo Growth Factor Delivery to Chondrocytes and Bone-Marrow-Derived Stromal Cells in Cartilage and in Self-Assembling Peptide

Scaffolds

by

Rachel E. Miller

B.S. Bioengineering The Pennsylvania State University, 2005

SUBMITTED TO THE DEPARTMENT OF BIOLOGICAL ENGINEERING IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY IN BIOLOGICAL ENGINEERING

AT THE MASSACHUSETTS INSTITUTE OF TECHNOLOGY

JUNE 2010

© 2010 Massachusetts Institute of Technology. All rights reserved.

Signature of Author _______________________________________________________

Department of Biological Engineering May 20, 2010

Certified by______________________________________________________________

Alan J. Grodzinsky Professor of Biological, Electrical, and Mechanical Engineering

Thesis Supervisor Accepted by _____________________________________________________________

Peter C. Dedon Professor of Toxicology and Biological Engineering

Associate Head, Department of Biological Engineering Chair, Course XX Graduate Program Committee

Page 2: In vitro and in vivo Growth Factor Delivery to

2

Thesis Committee Dr. Alan J. Grodzinsky Thesis Advisor Professor of Biological, Electrical, and Mechanical Engineering Massachusetts Institute of Technology Dr. Linda G. Griffith Thesis Committee Chair Professor of Biological and Mechanical Engineering Massachusetts Institute of Technology Dr. Richard T. Lee Professor of Medicine Harvard Medical School Dr. Parth Patwari Associate Biophysicist, Brigham and Women’s Hospital Instructor in Medicine Harvard Medical School Dr. David B. Schauer Professor of Biological Engineering and Comparative Medicine Massachusetts Institute of Technology

Page 3: In vitro and in vivo Growth Factor Delivery to

3

In vitro and in vivo Growth Factor Delivery to Chondrocytes and Bone-Marrow-Derived Stromal Cells in Cartilage and in Self-Assembling

Peptide Scaffolds by

Rachel E. Miller

Submitted to the Department of Biological Engineering on May 20, 2010 in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy in Biological

Engineering at the Massachusetts Institute of Technology

ABSTRACT The inability of articular cartilage to repair itself after acute injury has been

implicated in the development of osteoarthritis. The objective of this work was to develop methods for delivering growth factors to cartilage and to test the ability of a self-assembling peptide scaffold, (KLDL)3, with or without growth factors to augment repair. Delivery methods included growth factor adsorption, scaffold-tethering, and modification of growth factor structure.

(KLDL)3 was modified to deliver IGF-1 and TGF-β1 to chondrocytes and bone-marrow-derived stromal cells (BMSCs), respectively, by adsorption and by biotin-streptavidin tethering. This study showed that while TGF-β1 can be effectively delivered by adsorption, IGF-1 can not. Additionally, while tethering these factors provided long-term sequestration, tethering did not stimulate proteoglycan production in vitro.

A full-thickness, critically sized, rabbit cartilage defect model was used to test the ability of (KLDL)3 with or without chondrogenic factors (TGF-β1, dexamethasone, and IGF-1) and BMSCs to stimulate cartilage regeneration in vivo. (KLDL)3 alone showed the greatest repair after 12 weeks with significantly higher Safranin-O, collagen II immunostaining, and cumulative histology scores compared to untreated contralateral controls. Ongoing studies include the evaluation of (KLDL)3 in a clinically relevant sized equine defect co-treated with microfracture and subjected to strenuous exercise.

A fusion protein was created by adding a heparin-binding domain to IGF-1 (HB-IGF-1), converting IGF-1 from a short-acting growth factor to one that can be retained and locally delivered in articular cartilage in vivo. It was shown that HB-IGF-1 is retained in cartilage through binding to negatively charged glycosaminoglycan chains, with chondroitin sulfate the most prevalent type in cartilage. HB-IGF-1 was shown to bind adult human cartilage and to be preferentially delivered and retained in rat articular cartilage after intra-articular injection. In contrast, unmodified IGF-1 was not detectable after intra-articular injection. These results suggest that modification of growth factors with heparin-binding domains may be a clinically relevant strategy for local delivery to cartilage.

Taken together, these results show that (KLDL)3 self-assembling peptide hydrogels are customizable for growth factor delivery and can promote cartilage repair in vivo. In addition, the fusion protein HB-IGF-1 is preferentially retained in cartilage tissue compared to un-modified IGF-1. Thesis Supervisor: Alan J. Grodzinsky Title: Professor of Biological, Electrical, and Mechanical Engineering

Page 4: In vitro and in vivo Growth Factor Delivery to

4

EDUCATION Ph.D. in Biological Engineering, concentration in Biomaterials, May 2010 Massachusetts Institute of Technology, Cambridge, MA 02139 Thesis Title: In vitro and in vivo Growth Factor Delivery to Chondrocytes and Bone-Marrow-Derived Stromal Cells in Cartilage and in Self-Assembling Peptide Scaffolds Thesis Advisor: Prof. Alan Grodzinsky GPA: 4.9/5.0 B.S. and Honors in Bioengineering (Materials Science concentration) with highest distinction, May 2005 The Pennsylvania State University, University Park, PA 16802 Thesis Title: The Development of Nano-engineered Hemocompatible Biomaterials Thesis Supervisor: Prof. Erwin Vogler GPA: 3.99/4.0 Annville-Cleona High School, Salutatorian, June 2001 Annville, PA 17003

AWARDS

Siebel Scholar, Class of 2010 National Defense Science and Engineering Graduate Fellowship (2005 – 2008) National Science Foundation (NSF) Graduate Research Fellowship (2008 – 2010) Massachusetts Institute of Technology (MIT) Presidential Fellowship (2005 – 2006) Student Marshal – PSU College of Engineering – 2005 graduating class Barry M. Goldwater Scholarship (2004 – 2005) Astronaut Scholarship Foundation Scholarship (2004 – 2005) National Dean’s List (2004) Society of Distinguished Alumni Scholarship (2004 – 2005) Evan Pugh Scholar Award (top 0.5% of my class 2003-2005) Schreyer Honors College Scholarship (2001 – 2005) PSU College of Engineering Scholarship (2001 – 2005) President’s Freshman Award (Spring 2002) Undergraduate Dean’s List (All Semesters)

REFEREED JOURNAL ARTICLES

Miller R, Grodzinsky A, Vanderploeg E, Ferris D, Lee C, Barrett M, Kisiday J, Frisbie D. Repair of Full-Thickness Articular Cartilage Defect Using Self-Assembling Peptide, Growth Factors, and Bone Marrow Derived Stromal Cells. Osteoarthritis and Cartilage, 2010; accepted pending minor revision. Miller R, Kopesky P, Grodzinsky A. Customizable Scaffolds for Cell and Growth Factor Delivery. Clinical Orthopaedics and Related Research, 2010; submitted. Miller R, Grodzinsky A, Cummings K, Lee R, Patwari P. Heparin-Binding IGF-1 Provides Sustained Delivery of IGF-1 to Cartilage In Vivo. Arthritis and Rheumatism, 2010; submitted.

Page 5: In vitro and in vivo Growth Factor Delivery to

5

Miller R, Grodzinsky A, Ferris D, Lee C, Barrett M, Kisiday J, Frisbie D. Effects of Self-Assembling Peptide on Repair of Equine Articular Cartilage Defects in Combination with Microfracture. In preparation. Tokunou T, Miller R, Patwari P, Davis ME, Segers VFM, Grodzinsky AJ, Lee RT. Engineering Insulin-like Growth Factor-1 for Local Delivery. FASEB J. 2008; 22:1886-1893. Guo Z, Bussard K, Chatterjee K, Miller R, Vogler E, Siedlecki C. Mathematical Modeling of Material-Induced Blood Plasma Coagulation. Biomaterials. 2006; 27:796-806. Miller R, Guo Z, Vogler E, and Siedlecki C. Plasma Coagulation Response to Surfaces with Nanoscale Chemical Heterogeneity. Biomaterials. 2006; 27:208-215. Zhuo R, Miller R, Bussard K, Siedlecki C, and Vogler E. Procoagulant Stimulus Processing by the Intrinsic Pathway of Blood Plasma Coagulation. Biomaterials. 2005; 26:2965-2973.

REFEREED CONFERENCE PRESENTATIONS

Poster presentation (#973): Miller RE, Grodzinsky AJ, Vanderploeg EJ, Kopesky PW, Florine E, Kisiday JD, Frisbie DD. Repair of Full-Thickness Articular Cartilage Defect Using Self-Assembling Peptide, Growth Factors, and BMSCs. 56th Annual Meeting of the Orthopaedic Research Society, New Orleans, LA. March 6-9, 2010. Poster presentation (#982): Miller RE, Grodzinsky AJ, Cummings K, Lee RT, Patwari P. Intra-articular Injection of HB-IGF-1 Sustains Delivery of IGF-1 to Cartilage through Binding to Chondroitin Sulfate. 56th Annual Meeting of the Orthopaedic Research Society, New Orleans, LA. March 6-9, 2010. Poster presentation (#1326): Florine EF, Vanderploeg EJ, Kopesky PW, Miller RE, Grodzinsky AJ. Dexamethasone Suppresses Aggrecan Catabolism in BMSC-seeded Peptide Hydrogels. 56th Annual Meeting of the Orthopaedic Research Society, New Orleans, LA. March 6-9, 2010. Poster presentation: Miller RE, Grodzinsky AJ, Vanderploeg EJ, Kopesky PW, Florine E, Kisiday JD, Frisbie DD. Self-Assembling Peptide Heals Rabbit Defects In Vivo. 2009 Osteoarthritis Research Society International (OARSI) World Congress, Montreal, Sept. 10-13, 2009. Poster presentation: Miller RE, Grodzinsky AJ, Cummings K, Lee RT, Patwari P. Heparin-Binding IGF-1 Provides Sustained Delivery of IGF-1 to Cartilage In Vivo. 2009 Osteoarthritis Research Society International (OARSI) World Congress, Montreal, Sept. 10-13, 2009.

Page 6: In vitro and in vivo Growth Factor Delivery to

6

Poster presentation: Swaminathan K, Stevens AL, Indrakanti R, Miller RE, Wishnok JS, Tannenbaum SR, Grodzinsky AJ. Additive Effects of Injurious Compression and Inflammatory Cytokines on Cartilage Damage and Chondrocyte Response: a Quantitative iTRAQ Proteomics Study. 55th Annual Meeting of the Orthopaedic Research Society, Las Vegas, NV. Feb 22-25, 2009. Oral and Poster (#311) presentation: Tokunou T, Miller R, Patwari P, Davis ME, Segers V, Grodzinsky AJ, Lee RT. A Novel IGF-1 Fusion Protein for Cartilage Delivery. 54th Annual Meeting of the Orthopaedic Research Society, San Francisco, CA. March 2-5, 2008. Oral presentation: Kopesky PW, Lee CSD, Miller RE, Kisiday JD, Frisbie DD, Grodzinsky AJ. Comparable Matrix Production by Adult Equine Marrow-derived MSCs and Primary Chondrocytes in a Self-assembling Peptide Hydrogel: Effect of Age and Growth Factors. 53rd Annual Meeting of the Orthopaedic Research Society, San Diego, CA. Feb 11-14, 2007. Oral presentation: Miller R, Guo Z, Vogler E, and Siedlecki C. Plasma Coagulation Response to Surfaces with Nanoscale Chemical Heterogeneity. Society for Biomaterials Annual Meeting. 28 April 2005. Poster presentation, First Place in Engineering Division: Miller R, Zhuo R, Siedlecki C, and Vogler E. (2004). Procoagulant Stimulus Processing by the Intrinsic Pathway of Blood Coagulation. Penn State Undergraduate Exhibition, The Pennsylvania State University, University Park, PA.

TEACHING EXPERIENCE

Teaching Assistant: Fields, Forces, and Flows in Biological Systems (20.430), MIT Fall 2006

UNIVERSITY SERVICE

Biological Engineering Graduate Student Board, 2006-2009 Biological Engineering Retreat Committee, 2006-2009 Mentor: Undergraduate Research Opportunities Program, 2009-2010 Biomedical Engineering Society, 2003-2005 Mentor: Schreyer Honors College, PSU, Fall 2002, 2003, 2004

Page 7: In vitro and in vivo Growth Factor Delivery to

7

Acknowledgements As a kid in elementary school, I imagined being an archaeologist, meteorologist, or marine biologist, but I only discovered what being a bioengineer meant in high school: the ability to blend my loves of math, science, and medicine. To be writing my doctoral thesis at MIT is still unbelievable. I owe many people thanks for helping me to attain this dream, but my family tops the list. My parents always encouraged my curiosity and taught me to make the most of every opportunity; my brother helped me to develop my rebellious adventurous side. I would also like to thank my high school calculus and computer programming teacher, Mr. Kreiser, for pushing me to challenge myself, and my undergraduate research advisor, Dr. Vogler, for introducing me to research and for making me apply to MIT. Working in the Grodzinsky lab has made this experience one in which I enjoyed coming to work everyday. Al encouraged me to think through problems and gave me the freedom to explore my ideas. I am especially grateful for his generosity in time and spirit and for his ability to foster a sense of community in the lab. I would also like to thank the rest of the Grodzinsky lab for being fun colleagues who were always willing to give advice and to offer their expertise. I have been privileged to work with several great collaborators without whom this work would not have been possible. In particular, I would like to thank Dave Frisbie and John Kisiday at Colorado State University, and Rich Lee, Parth Patwari, and Tomo Tokunou at Harvard Medical School. Linda Griffith and David Schauer ensured that I think outside the cartilage box, and for that I am grateful. My colleagues in the rest of the BE department have also had a large positive impact on my time here. I would like to thank Doug Lauffenburger for always taking time out of his schedule to meet with me and for attracting me to MIT in the first place. I am also grateful to the members of the class of 2005 for helping me to survive my first year, and to all of my other classmates for making the department the fun place it is. I could always count on my classmates to help when it came to borrowing reagents, sharing protocols, and finding equipment to use. Finally, I would not have survived without my friends, Diana Chai, Ta Hang, Megan McBee, and Anna Stevens. Sometimes the little things make all the difference in life, and so I would like to thank the coffee ladies Carrie and Thelma in Lobby 7 for brightening my mornings, the baristas at the Central Square and Ames St Starbucks for remembering my Americanos, and Tiffany at Tommy Doyle’s for making our TGIF’s entertaining. And in case anyone wonders where their tax dollars go, some of them have gone to support my education and research, and for that I am grateful: NIH Grants EB003805, AR33236, and AR045779; National Science Foundation (NSF) and National Defense Science and Engineering Graduate (NDSEG) fellowships. I was also supported during my final year by a Siebel Scholar fellowship.

Page 8: In vitro and in vivo Growth Factor Delivery to

8

Table of Contents List of Tables 10 List of Figures 11 Chapter 1 13 Introduction 1.1 Cartilage Biology 13 1.2 Cartilage Pathogenesis and the Current Strategies for Repair 14 1.3 Cartilage Tissue Engineering 15 1.4 Insulin-like Growth Factor – I 18 1.5 In vivo Animal Models 20 1.6 Thesis Outline 21 1.7 References 22 Chapter 2 28 Growth Factor Delivery via Self-assembling Peptide Scaffolds 2.1 Introduction 29 2.2 Materials and Methods 31 2.3 Results 34 2.4 Discussion 37 2.5 Acknowledgements 40 2.6 References 41 2.7 Figures 45 Chapter 3 49 Effect of Self-assembling Peptide, Chondrogenic Factors, and Bone-Marrow-Derived Stromal Cells on Osteochondral Repair 3.1 Introduction 50 3.2 Materials and Methods 53 3.3 Results 58 3.4 Discussion 62 3.5 Acknowledgements 66 3.6 References 67 3.7 Tables 71 3.8 Figures 80 Chapter 4 85 Intra-articular Injection of HB-IGF-1 Sustains Delivery of IGF-1 to Cartilage through Binding to Chondroitin Sulfate 4.1 Introduction 86 4.2 Materials and Methods 88 4.3 Results 93 4.4 Discussion 97 4.5 Acknowledgements 99 4.6 References 100

Page 9: In vitro and in vivo Growth Factor Delivery to

9

4.7 Figures 104 Chapter 5 109 Conclusions and Future Directions Appendix A: Protocols and data related to Chapter 2 113 Appendix B: Protocols and data related to Chapter 3 128 Appendix C: Protocols and data related to Chapter 4 135 Appendix D: Luminex Pilot Study 143

Page 10: In vitro and in vivo Growth Factor Delivery to

10

List of Tables Chapter 3: Effect of Self-assembling Peptide, Chondrogenic Factors, and Bone Marrow Derived Stromal Cells on Osteochondral Repair Table 3.1 Treatment groups with amounts delivered per animal 71 Table 3.2 Gross Observations Scoring System 72 Table 3.3 Modified O’Driscoll Histological Scoring System 75 Table 3.4 Gross Scores 77 Table 3.5 Histological Scores 79 Appendix B Table B1 Treatment groups not included in Chapter 3 131

Page 11: In vitro and in vivo Growth Factor Delivery to

11

List of Figures Chapter 2: Growth factor delivery via self-assembling peptide scaffolds Figure 2.1 High-affinity tethering prolongs retention of IGF-1 45 Figure 2.2 bIGF-1 is bioactive 46 Figure 2.3 Soluble IGF-1, but not adsorbed or tethered IGF-1, stimulates sGAG 47

production Figure 2.4 Soluble and adsorbed TGF-β1, but not tethered, promotes sGAG 48

production Chapter 3: Effect of Self-assembling Peptide, Chondrogenic Factors, and Bone Marrow Derived Stromal Cells on Osteochondral Repair Figure 3.1 Gross necropsy photographs of treated and untreated joints in KLD, 80

KLD+CF, and KLD+CF+BMSCs Figure 3.2 Gross pathologic observation of joints comparing KLD, KLD+CF, 81

and KLD+CF+BMSCs treated and contralateral untreated defects Figure 3.3 Histologic scores comparing KLD, KLD+CF, and KLD+CF+BMSCs 82

treated and untreated defects Figure 3.4 Safranin-O staining 83 Figure 3.5 Immunohistochemistry scores 84 Chapter 4: Intra-articular Injection of HB-IGF-1 Sustains Delivery of IGF-1 to Cartilage through Binding to Chondroitin Sulfate Figure 4.1 Retention of HB-IGF-1 in bovine cartilage explants following 104

enzymatic digestion of glycosaminoglycans Figure 4.2 Retention of HB-IGF-1 on cells lacking heparan sulfate 105 Figure 4.3 Binding analysis of HB-IGF-1 and IGF-1 to isolated 106

glycosaminoglycans Figure 4.4 Retention of HB-IGF-1 in vivo 107 Figure 4.5 Retention of HB-IGF-1 in human cartilage explants 108 Appendix A Figure A1 Effect of soluble IGF-1 and bIGF-1 on GAG and DNA synthesis 115

of chondrocytes encapsulated in KLD Figure A2 Effect of soluble IGF-1 and bIGF-1 on sulfate incorporation 116

of chondrocytes encapsulated in KLD Figure A3 Effect of tethered IGF-1 on BMSCs encapsulated in KLD 117 Figure A4 Example flow cytometry images 119 Figure A5 Effect of casting order on cell death 120 Figure A6 Chondrocytes encapsulated in KLD with or without tethered 121

IGF-1 were evaluated for cell death and apoptosis Figure A7 TUNEL staining on chondrocytes encapsulated in KLD 124

Page 12: In vitro and in vivo Growth Factor Delivery to

12

Figure A8 Phospho AKT and phopho ERK Western blots 127 Appendix B Figure B1 Circular dichroism spectroscopy to monitor assembly 128 Figure B2 In situ peptide assembly and cell viability 129 Figure B3 In situ filling and joint articulation using 4 mg/mL KLD 130 Figure B4 In situ filling and joint articulation using 3.2 mg/mL KLD 130 Figure B5 Gross necropsy photographs of joints in Groups 4 and 5 131 Figure B6 Gross and histologic effects comparing defects treated with 132

KLD+BMSCs+1.4 ng TGF-β1 in group 3 to defects treated with the same combination in group 4

Figure B7 Immunohistochemistry scores (0-4) 133 Appendix C Figure C1 Western blots on conditioned medium using anti-HS stub 136 Appendix D Figure D1 Luminex data on mouse joints 144

Page 13: In vitro and in vivo Growth Factor Delivery to

13

Chapter 1. Introduction

1.1 Cartilage Biology

Adult articular cartilage is composed of one cell type, the chondrocyte, which accounts

for approximately 1-5% of the tissue volume with the remaining filled by extracellular

matrix (ECM) (15-40%) and water (60-85%)20, 47, 51, 60. Cells maintain a spherical

morphology, though somewhat flattened in the superficial zone, and do not directly

contact other cells58. Cartilage lacks a nervous and vascular supply, resulting in

chondrocytes in adult articular cartilage remaining relatively inactive metabolically and

having a poor repair capacity20, 48.

Chondrocytes synthesize all ECM components, which include collagens (10-30% wet

wt), proteoglycans (PGs) (3-10% wet wt), and non-collagenous proteins and

glycoproteins51, 60. The collagen network provides tensile strength and helps to retain

proteoglycans, which offer compressive resistance. Type 2 collagen is the predominant

type found in articular cartilage with types 9 and 11 incorporated within the basic

structure of this heteropolymer13. The major PG in the extracellular matrix is aggrecan,

which consists of a protein backbone to which glycosaminoglycan (GAG) side chains of

chondroitin sulfate (CS) and keratan sulfate (KS) are covalently attached60. The

negatively charged GAGs of the PGs give cartilage its high fixed-charge density,

controlling ionic equilibrium in the tissue50. Aggrecan molecules bind non-covalently to

hyaluronic acid (HA) chains, stabilized by link protein26. Chondrocytes are directly

surrounded by a pericellular matrix about 2 µm wide58 composed of molecules that can

Page 14: In vitro and in vivo Growth Factor Delivery to

14

interact with the cell including type 6 collagen, HA, a heparan sulfate and chondroitin

sulfate containing proteoglycan (perlecan), and decorin57.

1.2 Cartilage Pathogenesis and the Current Strategies for Repair

Cartilage injuries generally occur when the knee is impacted or subjected to sudden

twisting, often concurrent with meniscal or ligament tears6, 27. These types of injuries

occur in both children and adults55, with roughly 3 million knee injuries reported each

year in the United States1. These injuries result in an acute inflammatory environment

(presence of IL-1β, TNF-α, IL-6, among others) in the joint, which dissipates after 1-2

weeks30, but remains elevated above normal levels for months to years4, 44, 45. These

changes in cytokine levels, in combination with changes in loading of the joint69, are

implicated in the progression of a majority of people who sustain these injuries to

develop osteoarthritis (OA) within 10-20 years24, 45.

OA is a musculoskeletal disorder and the most common form of arthritis, affecting 21.5%

of the U.S. population20, 73. The disease is characterized by progressive destruction of

articular cartilage; symptoms include pain and impaired joint movement leading to

disability20. Current treatments include drug therapy for pain relief, surgical approaches

(e.g., microfracture, drilling, and mosaic-plasty), biological approaches (e.g., autologous

chondrocyte implantation (ACI)), and as a final recourse, joint replacement. None of

these treatments has yielded lasting results or regeneration of tissue equivalent to normal

cartilage20, 60. Microfracture, which creates hemorrhage to induce the migration of

potential repair cells, results in fibrocartilage tissue that is bio-mechanically inferior to

Page 15: In vitro and in vivo Growth Factor Delivery to

15

native tissue65. ACI is not indicated for the treatment of cartilage damage associated with

OA and results have not been shown to be significantly different than microfracture

treatment when used to repair focal defects within joint surfaces32, 38. Joint replacement is

currently the only option for severe articular cartilage degeneration; however, it is not a

permanent solution and often, additional surgery is required. Recently it has been shown

in vitro that it is possible for cartilage explants to recover from initial aggrecanase-

mediated GAG loss but not from collagen loss35, 59. This suggests that early intervention

following cartilage injuries may be key to halting progression to OA, likely with a

combination of anabolic, anti-catabolic, and defect-filling treatments45.

1.3 Cartilage Tissue Engineering

In 1997, a panel held by the National Science Foundation defined tissue engineering as

“… the application of the principles and methods of engineering and life sciences toward

the fundamental understanding of structure-function relationships in normal and

pathological mammalian tissue and the development of biological substitutes to restore,

maintain, or improve function39”. From the previous section, this suggests that cartilage

repair may be improved (1) by augmenting native repair attempts through filling defects

in situ or (2) by implanting more mature constructs grown in vitro and developing

methods for integrating this engineered tissue with native tissue27. In the case of (1), this

could be particularly beneficial in combination with marrow stimulation techniques in

that larger defect sizes may be able to be treated when an external scaffold is supplied to

encourage cellular infiltration and differentiation. This strategy will be the focus of this

thesis work. Many groups have begun efforts towards the development of tissue-

Page 16: In vitro and in vivo Growth Factor Delivery to

16

engineered cartilage; however the growth of cartilage that is comparable to native tissue

and is able to successfully integrate with native tissue has not yet been achieved (see 8, 18,

28, 41, 67 for review). The challenge for successful tissue engineering remains the discovery

of an optimal combination of scaffold, biological signaling, and cells for tissue

regeneration that is also practical for clinical use.

The scaffold should ideally serve as a microenvironment that promotes ECM production

by either endogenous or exogenous cells. A variety of natural and synthetic materials

have been proposed for use in cartilage tissue engineering. As some of the few materials

approved by the Food and Drug Administration (FDA) for use in the body, polyglycolic

acid (PGA), polylactic acid (PLA), and polylactic-co-glycolic acid (PLGA) have been

widely investigated, but these polymers degrade quickly in the body and have been

shown to produce toxic monomers70. Natural polymers that undergo enzymatic

hydrolysis, such as hyaluronic acid, collagen, fibrin, and chitin have also been used in a

variety of combinations as scaffolds28, 70. While these natural materials are all able to

support maintenance of the chondrocyte phenotype, none has been shown to be an ideal

substrate for cartilage formation with biochemical and mechanical properties comparable

to native tissue. Hydrogels are an appealing scaffold choice due to their high water

content, similar to the articular cartilage environment. This attribute also allows for the

encapsulation of chondrocytes and proteins within the network, and these gels have been

shown to maintain the spherical morphology and gene expression of chondrocytes74.

Hydrogels have been developed from alginate, polyethyleneglycol (PEG), chondroitin

sulfate, self-assembling peptides, and other materials41.

Page 17: In vitro and in vivo Growth Factor Delivery to

17

Self-assembling peptides offer the opportunity to create customizable scaffolds that can

be induced to assemble upon placement in a physiological environment. In addition,

scaffolds composed of nanofibers have been shown to produce better outcomes than

microfiber-based scaffolds42. The family of self-assembling peptides all satisfies the

principle that there is no net charge, but an equal number of ionizable basic and acidic

amino acids that promote β-sheet formation upon self-assembly, which can be initiated

by a solution at physiological pH and ionic strength36. The peptide KLD is composed of

the sequence AcN-KLDLKLDLKLDL-CNH2 and is based on the work of Zhang et al.75,

76. KLD has been demonstrated to support the synthesis and accumulation of cartilage-

like ECM by embedded chondrocytes36 and bone marrow derived stromal cells

(BMSCs)37, 40.

This peptide also offers flexibility in design that allows for the opportunity of growth

factor delivery for local, specific, prolonged biological stimulation – key to achieving

optimal tissue induction, especially since growth factors typically have a short half-life

once they are introduced into the body. A variety of growth factors have been shown to

be important in chondrogenesis and in stimulation of matrix production by chondrocytes,

including: TGF-β1, IGF-1, and members of the BMP and GDF families (see 66 for

review). Recently, Davis et al. developed a system for tethering IGF-1 to another self-

assembling peptide, RAD16-II, by a “biotin-sandwich” technique9. By adding a biotin

linkage to the end of the peptide sequence and incorporating a minority of this

biotinylated peptide in a solution of un-modified peptide, self-assembly was not

Page 18: In vitro and in vivo Growth Factor Delivery to

18

disrupted. This allowed for the attachment of biotinylated IGF-1 molecules through the

inclusion of streptavidin. This system provided controlled delivery of IGF-1 to

myocardial tissue in vivo. The non-specific “biotin-sandwich” strategy should be able to

be applied to other self-assembling peptides such as KLD and offers the potential for

multiple growth factors to be incorporated into a hydrogel for local tissue delivery.

The final parameter to be considered for successful tissue engineering is cell source.

There are three main types of cells that have been considered for use in cartilage tissue

engineering: primary chondrocytes, adult bone-marrow-derived stromal cells (BMSCs),

and embryonic stem cells. There is a limited availability of primary chondrocytes in adult

cartilage, and expansion in culture often leads to de-differentiation41, 70. Embryonic stem

cells, while offering great potential for the differentiation into multiple cell types, are

associated with many ethical and safety concerns. Therefore, adult BMSCs may present

the best choice for the future of cartilage tissue engineering. Autologous BMSCs are

obtainable from a patient’s own bone marrow, and they have been shown to be able to

undergo expansion and chondrogenesis without risk of tumorigenesis28, 41, 49, 70. Much

current research is being directed towards the development of optimal expansion and

differentiation conditions12, 20, 28, 29, 41, 49, 60.

1.4 Insulin-like Growth Factor – I

IGF-1 plays a primary role in cartilage homeostasis by stimulating PG synthesis,

promoting cell survival, and inhibiting cartilage degradation. IGF-1 is produced primarily

by the liver, but other cell types, including chondrocytes, are also able to synthesize IGF-

Page 19: In vitro and in vivo Growth Factor Delivery to

19

1. In cartilage, IGF-1 can act both in a paracrine and autocrine manner52. It is a basic, 7.6

kDa protein that binds the IGF-1 receptor (IGF-1R) with a KD of 0.2-1 nM34. IGF-1Rs

exist on the cell surface as disulfide-linked dimers and studies have shown that a single

molecule of IGF-1 is able to activate the receptor64 with approximately 10,000 receptors

on the surface of each chondrocyte5. Human OA chondrocytes express and produce

increased levels of IGF-1 but are hypo-responsive to it52. An explanation for this may be

the increased production of IGF binding proteins (IGFBPs), which can associate with the

cell surface or ECM and may be responsible for decreased amounts of IGF-1 available to

bind IGF-1Rs. Therefore, IGF-1 growth-factor or gene therapy offers a potential

treatment for diminished IGF-1 activity, but to be effective, amounts of IGF-1 delivered

within the joint must exceed the binding capacity of local IGFBPs10.

IGF-1 has also been used as treatment in a variety of cartilage injury models. Inclusion of

IGF-I in fibrin-chondrocyte constructs enhanced chondrogenesis in equine cartilage

defects, including incorporation into surrounding cartilage14. Cartilage explants treated

with IL-1 were partially rescued from proteoglycan degradation by co-culture with

synovial cells transduced with AdIGF-154. Madry et al. demonstrated that chondrocytes

transfected with AdIGF-1 included in alginate scaffolds can enhance repair of rabbit

cartilage defects46. In addition, Goodrich et al. transplanted equine chondrocytes

genetically modified with AdIGF-1 into equine femoral trochlea defects and found that

repair of the defect with hyaline-like cartilage was accelerated22. Direct injections of

AdIGF-1 into the metacarpophalangeal (MCP) joints of horses were also shown to result

in significant elevations of IGF-I in synovial fluid for approximately 21 days with

Page 20: In vitro and in vivo Growth Factor Delivery to

20

minimal detrimental effects21. A critical factor that must be considered in any IGF-1

delivery strategy is that systemic delivery of IGF-1 can cause significant side effects31 as

well as the potential to promote diabetic retinopathy72 and cancer7. These studies suggest

that IGF-1 is a critical component of cartilage repair, yet an optimal delivery technique

has yet to be developed.

1.5 In vivo Animal Models

While in vitro cell work is important for the initial screening and development of tissue-

engineered products, the effectiveness of constructs in vivo must also be considered. To

this end, several animal models have been used for the testing of constructs designed to

regenerate cartilage. Typically, the initial screening for the safety and in vivo efficacy of

such constructs are performed in a small animal model. Nude mice have been used for the

testing of cartilage growth potential by subcutaneous implantation2, 33, 56, 62, 63, however,

this model does not allow for immunological screening of potential constructs nor does it

evaluate the ability for healing of a cartilage defect or for the ability to integrate with

native tissue. An alternative immuno-competent small animal model is the rabbit. Rabbits

offer many of the same advantages of mice in that they are available at low cost with

minimal housing requirements. Appropriate defect models in weight-bearing areas of the

medial femoral condyles of rabbit knee joints have been developed for the testing of

tissue-engineered constructs11, 17, 23, 61.

Once basic testing has been completed, true construct functionality must be evaluated in a

large animal model such as a dog, sheep, goat, pig, or horse. While no defect model is

Page 21: In vitro and in vivo Growth Factor Delivery to

21

directly applicable to humans due to physiological and anatomical differences between

the human joint and model animal joints28, the horse seems to most closely approximate

human cartilage based on the thickness of its articular cartilage16, its susceptibility to

osteoarthritis15, and similar cartilage biochemical composition68. A model for inducing

OA in equines has been developed15, and several studies have used manufactured defects

as models for testing cartilage repair strategies3, 43, 53, 71. It must be kept in mind,

however, that positive results obtained from in vivo animal testing must be viewed with

caution as they are not necessarily predictive of success in humans19, 25.

1.6 Thesis Outline

The objective of this work was to develop methods for delivering growth factors to

cartilage and to test the ability of a self-assembling peptide scaffold, (KLDL)3, with or

without growth factors to augment repair. Delivery methods included growth factor

adsorption, scaffold-tethering, and modification of growth factor structure.

In Chapter 2, IGF-1 and TGF-β1 were adsorbed and tethered to the self-assembling

peptide scaffold KLD and the effects on matrix production by chondrocytes and BMSCs,

respectively, were observed.

In Chapter 3, the effects of KLD, chondrogenic factors, and allogeneic BMSCs on the in

vivo repair of a critically sized full-thickness rabbit cartilage defect were examined.

Page 22: In vitro and in vivo Growth Factor Delivery to

22

In Chapter 4, binding of a novel fusion protein, heparin-binding IGF-1 or HB-IGF-1, in

cartilage tissue was investigated. The mechanism by which HB-IGF-1 is retained in

cartilage was examined and the ability of HB-IGF-1 to provide sustained growth factor

delivery to cartilage in vivo and to human cartilage explants was determined.

Finally, in Chapter 5, the main findings and conclusions are discussed and new questions

motivated by this thesis are explored.

1.7 References

1. United States Bone and Joint Decade: The Burden of Musculoskeletal Diseases in the United States. 2008, Rosemont, IL: American Academy of Orthopaedic Surgeons.

2. Alsberg E, Anderson KW, Albeiruti A, Rowley JA, and Mooney DJ. Engineering growing tissues. Proc Natl Acad Sci USA. 2002; 99:12025-12030.

3. Barnewitz D, Endres M, Kruger I, Becker A, Zimmermann J, Wilke I, Ringe J, Sittinger M, and Kaps C. Treatment of articular cartilage defects in horses with polymer-based cartilage tissue engineering grafts. Biomaterials. 2006; 27:2882-2889.

4. Beynnon BD, Uh BS, Johnson RJ, Abate JA, Nichols CE, Fleming BC, Poole AR, and Roos H. Rehabilitation after anterior cruciate ligament reconstruction: a prospective, randomized, double-blind comparison of programs administered over 2 different time intervals. Am J Sports Med. 2005; 33:347-59.

5. Bonassar LJ and Trippel SB. Interaction of epidermal growth factor and insulin-like growth factor-I in the regulation of growth plate chondrocytes. Exp Cell Res. 1997; 234:1-6.

6. Brophy RH, Zeltser D, Wright RW, and Flanigan D. Anterior Cruciate Ligament Reconstruction and Concomitant Articular Cartilage Injury: Incidence and Treatment. Arthroscopy. 2010; 26:112-120.

7. Chan JM, Stampfer MJ, Giovannucci E, Gann PH, Ma J, Wilkinson P, Hennekens CH, and Pollak M. Plasma Insulin-Like Growth Factor-I and Prostate Cancer Risk: A Prospective Study. Science. 1998; 279:563-566.

8. Chung C and Burdick JA. Engineering cartilage tissue. Adv Drug Deliv Rev. 2008; 60:243-62.

9. Davis ME, Hsieh PCH, Takahashi T, Song Q, Zhang S, Kamm RD, Grodzinsky AJ, Anversa P, and Lee RT. Local myocardial insulin-like growth factor 1 (IGF-1) delivery with biotinylated peptide nanofibers improves cell therapy for myocardial infarction. Proc Natl Acad Sci USA. 2006; 103:8155-8160.

Page 23: In vitro and in vivo Growth Factor Delivery to

23

10. De Ceuninck F, Caliez A, Dassencourt L, Anract P, and Renard P. Pharmacological disruption of insulin-like growth factor 1 binding to IGF-binding proteins restores anabolic responses in human osteoarthritic chondrocytes. Arthritis Res Ther. 2004; 6:R393-403.

11. De Franceschi L, Grigolo B, Roseti L, Facchini A, Fini M, Giavaresi G, Tschon M, and Giardino R. Transplantation of chondrocytes seeded on collagen-based scaffold in cartilage defects in rabbits. Journal of Biomedical Materials Research Part A. 2005; 75A:612-622.

12. Djouad F, Mrugala D, Noel D, and Jorgensen C. Engineered mesenchymal stem cells for cartilage repair. Regen Med. 2006; 1:529-537.

13. Eyre D, Weis M, and Wu J. Articular Cartilage Collagen: An Irreplaceable Framework? Eur Cells Mat. 2006; 12:57-63.

14. Fortier LA, Mohammed HO, Lust G, and Nixon AJ. Insulin-like growth factor-I enhances cell-based repair of articular cartilage. J Bone Joint Surg Br. 2002; 84-B:276-288.

15. Frisbie D, Ghivizzani S, Robbins P, Evans C, and McIlwraith C. Treatment of experimental equine osteoarthritis by in vivo delivery of the equine interleukin-1 receptor antagonist gene. Gene Ther. 2002; 9:12-20.

16. Frisbie D, Cross M, and McIlwraith C. A comparative study of articular cartilage thickness in the stifle of animal species used in human pre-clinical studies compared to articular cartilage thickness in the human knee. Vet Comp Orthop Traumatol. 2006; 19:142-146.

17. Gao J, Dennis JE, Solchaga LA, Goldberg VM, and Caplan AI. Repair of Osteochondral Defect with Tissue-Engineered Two-Phase Composite Material of Injectable Calcium Phosphate and Hyaluronan Sponge. Tissue Engineering. 2002; 8:827-837.

18. Getgood A, Brooks R, Fortier L, and Rushton N. Articular cartilage tissue engineering: today's research, tomorrow's practice? J Bone Joint Surg Br. 2009; 91:565-76.

19. Giannoni P, Crovace A, Malpeli M, Maggi E, Arbico R, Cancedda R, and Dozin B. Species Variability in the Differentiation Potential of in Vitro-Expanded Articular Chondrocytes Restricts Predictive Studies on Cartilage Repair Using Animal Models. Tissue Engineering. 2005; 11:237-248.

20. Goldring MB. Update on the biology of the chondrocyte and new approaches to treating cartilage diseases. Best Practice & Research Clinical Rheumatology. 2006; 20:1003-1025.

21. Goodrich LR, Brower-Toland BD, Warnick L, Robbins PD, Evans CH, and Nixon AJ. Direct adenovirus-mediated IGF-I gene transduction of synovium induces persisting synovial fluid IGF-I ligand elevations. 2006; 13:1253-1262.

22. Goodrich LR, Hidaka C, Robbins PD, Evans CH, and Nixon AJ. Genetic modification of chondrocytes with insulin-like growth factor-1 enhances cartilage healing in an equine model. J Bone Joint Surg Br. 2007; 89-B:672-685.

23. Grigolo B, Roseti L, Fiorini M, Fini M, Giavaresi G, Nicoli Aldini N, Giardino R, and Facchini A. Transplantation of chondrocytes seeded on a hyaluronan derivative (Hyaff(R)-11) into cartilage defects in rabbits. Biomaterials. 2001; 22:2417-2424.

Page 24: In vitro and in vivo Growth Factor Delivery to

24

24. Guilak F, Fermor B, Keefe FJ, Kraus VB, Olson SA, Pisetsky DS, Setton LA, and Weinberg JB. The role of biomechanics and inflammation in cartilage injury and repair. Clin Orthop Relat Res. 2004:17-26.

25. Habibovic P, Woodfield T, de Groot K, and van Blitterswijk C. Predictive value of in vitro and in vivo assays in bone and cartilage repair-what do they really tell us about the clinical performance? Adv Exp Med Biol. 2006; 585:327-360.

26. Hardingham TE and Fosang AJ. Proteoglycans: many forms and many functions. Faseb J. 1992; 6:861-70.

27. Hunziker EB. Articular cartilage repair: basic science and clinical progress. A review of the current status and prospects. Osteoarthritis Cartilage. 2002; 10:432-63.

28. Ikada Y. Challenges in tissue engineering. J R Soc Interface. 2006; 3:589-601. 29. Im G, Jung N, and Tae S. Chondrogenic Differentiation of Mesenchymal Stem

Cells Isolated from Patients in Late Adulthood: The Optimal Conditions of Growth Factors. Tissue Eng. 2006; 12:527-536.

30. Irie K, Uchiyama E, and Iwaso H. Intraarticular inflammatory cytokines in acute anterior cruciate ligament injured knee. Knee. 2003; 10:93-6.

31. Jabri N, Schalch D, Schwartz S, Fischer J, Kipnes M, Radnik B, Turman N, Marcsisin V, and Guler H. Adverse effects of recombinant human insulin-like growth factor I in obese insuin-resistant type II diabetic patients. Diabetes. 1994; 43:369-374.

32. Jakobsen RB, Engebretsen L, and Slauterbeck JR. An Analysis of the Quality of Cartilage Repair Studies. J Bone Joint Surg Am. 2005; 87:2232-2239.

33. Jin CZ, Park SR, Choi BH, Park K, and Min B-H. In Vivo Cartilage Tissue Engineering Using a Cell-Derived Extracellular Matrix Scaffold. Artificial Organs. 2007; 31:183-192.

34. Jones J and Clemmons D. Insulin-like growth factors and their binding proteins: biological actions. Endocr Rev. 1995; 16:3-34.

35. Karsdal MA, Madsen SH, Christiansen C, Henriksen K, Fosang AJ, and Sondergaard BC. Cartilage degradation is fully reversible in the presence of aggrecanase but not matrix metalloproteinase activity. Arthritis Res Ther. 2008; 10:R63.

36. Kisiday J, Jin M, Kurz B, Hung H, Semino C, Zhang S, and Grodzinsky AJ. Self-assembling peptide hydrogel fosters chondrocyte extracellular matrix production and cell division: Implications for cartilage tissue repair. Proc Natl Acad Sci USA. 2002; 99:9996-10001.

37. Kisiday JD, Kopesky PW, Evans CH, Grodzinsky AJ, McIlwraith CW, and Frisbie DD. Evaluation of adult equine bone marrow- and adipose-derived progenitor cell chondrogenesis in hydrogel cultures. J Orthop Res. 2008; 26:322-31.

38. Knutsen G, Engebretsen L, Ludvigsen TC, Drogset JO, Grontvedt T, Solheim E, Strand T, Roberts S, Isaksen V, and Johansen O. Autologous Chondrocyte Implantation Compared with Microfracture in the Knee. A Randomized Trial. J Bone Joint Surg Am. 2004; 86:455-464.

39. Koch R and Gorti G. Tissue engineering with chondrocytes. Facial Plast Surg. 2002; 18:59-68.

Page 25: In vitro and in vivo Growth Factor Delivery to

25

40. Kopesky PW, Vanderploeg EJ, Sandy J, Kurz B, and Grodzinsky A. Self-Assembling Peptide Hydrogels Modulate In Vitro Chondrogenesis of Bovine Bone Marrow Stromal Cells. Tissue Eng Part A. 2010; In press.

41. Kuo C, Li W, Mauck R, and Tuan R. Cartilage tissue engineering: its potential and uses. Curr Opin Rheumatol. 2006; 18:64-73.

42. Li W, Jiang Y, and Tuan R. Chondrocyte Phenotype in Engineered Fibrous Matrix Is Regulated by Fiber Size. Tissue Eng. 2006; 12:1775-1785.

43. Litzke L, Wagner E, Baumgaertner W, Hetzel U, Josimovic-Alasevic O, and Libera J. Repair of extensive articular cartilage defects in horses by autologous chondrocyte transplantation. Ann Biomed Eng. 2004; 32:57-69.

44. Lohmander LS, Hoerrner LA, Dahlberg L, Roos H, Bjornsson S, and Lark MW. Stromelysin, tissue inhibitor of metalloproteinases and proteoglycan fragments in human knee joint fluid after injury. J Rheumatol. 1993; 20:1362-8.

45. Lohmander LS, Englund PM, Dahl LL, and Roos EM. The long-term consequence of anterior cruciate ligament and meniscus injuries: osteoarthritis. Am J Sports Med. 2007; 35:1756-69.

46. Madry H, Kaul G, Cucchiarini M, Stein U, Zurakowski D, Remberger K, Menger MD, Kohn D, and Trippel SB. Enhanced repair of articular cartilage defects in vivo by transplanted chondrocytes overexpressing insulin-like growth factor I (IGF-I). Gene Ther. 2005; 12:1171-1179.

47. Mankin HJ and Thrasher AZ. Water content and binding in normal and osteoarthritic human cartilage. J Bone Joint Surg Am. 1975; 57:76-80.

48. Mankin HJ. The response of articular cartilage to mechanical injury. J Bone Joint Surg Am. 1982; 64:460-6.

49. Marion NW and Mao JJ, Mesenchymal Stem Cells and Tissue Engineering, Methods in Enzymology, in Stem Cell Tools and Other Experimental Protocols, I.K.a.R. Lanza, Editor. 2006, Academic Press. p. 339-361.

50. Maroudas A. Transport of solutes through cartilage: permeability to large molecules. J Anat. 1976; 122:335-347.

51. Maroudas A, Bayliss MT, and Venn MF. Further studies on the composition of human femoral head cartilage. Ann Rheum Dis. 1980; 39:514-23.

52. Martel-Pelletier J, Di Battista J, Lajeunesse D, and Pelletier J. IGF/IGFBP axis in cartilage and bone in osteoarthritis pathogenesis. Inflamm Res. 1998; 47:90-100.

53. Nixon A, Fortier L, Williams J, and Mohammed H. Enhanced repair of extensive articular defects by insulin-like growth factor-I-laden fibrin composites. Journal of Orthopaedic Research. 1999; 17:475-487.

54. Nixon AJ, Haupt JL, Frisbie DD, Morisset SS, McIlwraith CW, Robbins PD, Evans CH, and Ghivizzani S. Gene-mediated restoration of cartilage matrix by combination insulin-like growth factor-I/interleukin-1 receptor antagonist therapy. Gene Ther. 2004; 12:177-186.

55. Oeppen RS, Connolly SA, Bencardino JT, and Jaramillo D. Acute injury of the articular cartilage and subchondral bone: a common but unrecognized lesion in the immature knee. AJR Am J Roentgenol. 2004; 182:111-7.

56. Park S-H, Park SR, Chung SI, Pai KS, and Min B-H. Tissue-engineered Cartilage Using Fibrin/Hyaluronan Composite Gel and Its In Vivo Implantation. Artificial Organs. 2005; 29:838-845.

Page 26: In vitro and in vivo Growth Factor Delivery to

26

57. Poole AR, Kojima T, Yasuda T, Mwale F, Kobayashi M, and Laverty S. Composition and structure of articular cartilage: a template for tissue repair. Clin Orthop Relat Res. 2001:S26-33.

58. Poole CA, Flint MH, and Beaumont BW. Chondrons in cartilage: ultrastructural analysis of the pericellular microenvironment in adult human articular cartilages. J Orthop Res. 1987; 5:509-22.

59. Pratta MA, Yao W, Decicco C, Tortorella MD, Liu RQ, Copeland RA, Magolda R, Newton RC, Trzaskos JM, and Arner EC. Aggrecan protects cartilage collagen from proteolytic cleavage. J Biol Chem. 2003; 278:45539-45.

60. Schulz R and Bader A. Cartilage tissue engineering and bioreactor systems for the cultivation and stimulation of chondrocytes. Eur Biophys J. 2007; 36:539-568.

61. Shao X, Goh JCH, Hutmacher DW, Lee EH, and Zigang G. Repair of Large Articular Osteochondral Defects Using Hybrid Scaffolds and Bone Marrow-Derived Mesenchymal Stem Cells in a Rabbit Model. Tissue Engineering. 2006; 12:1539-1551.

62. Sharma B, Williams C, Khan M, Manson P, and Elisseeff J. In vivo chondrogenesis of mesenchymal stem cells in a photopolymerized hydrogel. Plast Reconstr Surg. 2007; 119:112-120.

63. Shieh S-J, Terada S, and Vacanti JP. Tissue engineering auricular reconstruction: in vitro and in vivo studies. Biomaterials. 2004; 25:1545-1557.

64. Siddle K, Ursø B, Niesler CA, Cope DL, Molina L, Surinya KH, and Soos MA. Specificity in ligand binding and intracellular signalling by insulin and insulin-like growth factor receptors Biochem Soc Trans 2001 29:513-525.

65. Simon T and Jackson D. Articular cartilage: injury pathways and treatment options. Sports Med Arthrosc. 2006; 14:146-154.

66. Steinert AF, Noth U, and Tuan RS. Concepts in gene therapy for cartilage repair. Injury. 2008; 39 Suppl 1:S97-113.

67. Stoop R. Smart biomaterials for tissue engineering of cartilage. Injury. 2008; 39 Suppl 1:S77-87.

68. Vachon A, Keeley F, McIlwraith C, and Chapman P. Biochemical analysis of normal articular cartilage in horses. Am J Vet Res. 1990; 51:1905-1911.

69. Van de Velde SK, Gill TJ, and Li G. Evaluation of kinematics of anterior cruciate ligament-deficient knees with use of advanced imaging techniques, three-dimensional modeling techniques, and robotics. J Bone Joint Surg Am. 2009; 91 Suppl 1:108-14.

70. van der Kraan PM, Buma P, van Kuppevelt T, and van Den Berg WB. Interaction of chondrocytes, extracellular matrix and growth factors: relevance for articular cartilage tissue engineering. Osteoarthritis and Cartilage. 2002; 10:631-637.

71. Wilke M, Nydam D, and Nixon A. Enhanced early chondrogenesis in articular defects following arthroscopic mesenchymal stem cell implantation in an equine model. Journal of Orthopaedic Research. 2007; 25:913-925.

72. Wilkinson-Berka J, Wraight C, and Werther G. The role of growth hormone, insulin-like growth factor and somatostatin in diabetic retinopathy. Curr Med Chem. 2006; 13:3307-3317.

73. Yelin E, Murphy L, Cisternas M, Foreman A, Pasta D, and Helmick C. Medical Care Expenditures and Earnings Losses Among Persons With Arthritis and Other

Page 27: In vitro and in vivo Growth Factor Delivery to

27

Rheumatic Conditions in 2003, and Comparisons With 1997. Arthritis & Rheumatism. 2007; 56:1397-1407.

74. Yoon D and Fisher J. Chondrocyte signaling and artificial matrices for articular cartilage engineering. Adv Exp Med Biol. 2006; 585:67-86.

75. Zhang S, Holmes T, Lockshin C, and Rich A. Spontaneous Assembly of a Self-Complementary Oligopeptide to Form a Stable Macroscopic Membrane. Proc Natl Acad Sci USA. 1993; 90:3334-3338.

76. Zhang S, Holmes TC, DiPersio CM, Hynes RO, Su X, and Rich A. Self-complementary oligopeptide matrices support mammalian cell attachment. Biomaterials. 1995; 16:1385-1393.

Page 28: In vitro and in vivo Growth Factor Delivery to

28

Chapter 2. Growth Factor Delivery via Self-assembling Peptide Scaffolds

The optimal strategy for delivering growth factors to cells for the purpose of cartilage

tissue engineering remains an unmet challenge. The purpose of this study was to engineer

self-assembling peptide scaffolds (i.e., (KLDL)3 or KLD) to deliver growth factors to

encapsulated cells. Chondrocytes or bone marrow stromal cells were encapsulated in

KLD with or without IGF-1 or TGF-β1, respectively. The growth factors were either (1)

pre-mixed with peptide solution prior to assembly to adsorb them to KLD, or (2) tethered

to KLD through biotin-streptavidin bonds. Fluorescently tagged streptavidin was used to

determine IGF-1 kinetics; sGAG and DNA content was measured. Tethering IGF-1 to

KLD significantly increased retention of IGF-1 in KLD compared to adsorption, but

neither method increased sGAG or DNA accumulation above control. Adsorbing TGF-β1

significantly increased proteoglycan accumulation above control, but again, tethering did

not affect sGAG levels. Increasing the amount of tethered TGF-β1 negatively impacted

DNA levels. This study provided an initial evaluation of methods for delivering growth

factors to cells within a KLD hydrogel scaffold and showed that while TGF-β1 can be

effectively delivered by adsorption, IGF-1 can not. Additionally, while tethering these

factors provided long-term sequestration, tethering was not effective in stimulating

proteoglycan production. Self-assembling peptides are a clinically relevant material that

can be injected in vivo. While tethering growth factors to KLD results in long-term

delivery, tethering does not result in the same bioactivity as soluble delivery, indicating

that presentation of proteins is vital when considering a delivery strategy.

Page 29: In vitro and in vivo Growth Factor Delivery to

29

2.1 Introduction

Acute cartilage defects remain a challenge to repair. Currently, interventions such as

microfracture or autologous chondrocyte implantation remain the standard of care25, but

these methods still result in mechanically inferior repair tissue15. Cartilage tissue

engineering has emerged as a possible avenue for improving repair. As such, many

combinations of scaffolds, cells, and growth factors have been proposed.

Growth factors including TGF-β1, IGF-1, and members of the GDF and BMP families

are known to be important in inducing repair, attracting migration of repair cells, and

stimulating chondrogenesis, proliferation, and production of matrix5, 43, 44. Due to the

reality that growth factors act on multiple tissues and can have detrimental side effects if

applied systemically, local delivery is necessary. Although intra-articular injections may

present a simple approach for delivery directly to the joint, often much higher than

physiologic concentrations must be used along with multiple injections in order to

overcome the fact that growth factors have short half-lives and may be cleared rapidly

from the synovial fluid. Therefore, delivery methods using scaffolds have been proposed

to protect growth factors from degradation and to deliver them over longer periods of

time45. Despite the advantages hydrogels offer as scaffolds, growth factors quickly

diffuse out of them28. Therefore, most successful delivery strategies to date have

incorporated growth factor-loaded microspheres within the hydrogel, so that release of

the factors can be controlled by the degradation rate of the microspheres43. While this has

shown positive results, high loading concentrations of growth factors within these

microspheres are still required. This creates the possibility of high localized doses

Page 30: In vitro and in vivo Growth Factor Delivery to

30

through initial bolus release in vivo, which is particularly dangerous for TGF-β1 since it

has been shown to cause side effects such as inflammation and osteophyte formation3.

Hydrogel scaffolds made from the self-assembling peptide sequences (RADA)4 and

(KLDL)3, which we refer to as RAD and KLD, respectively, have been shown to support

maintenance of the chondrocyte phenotype24, 34 and chondrogenesis of bone marrow

stromal cells26. These peptides assemble into hydrogels upon contact with solutions of

physiologic pH and ionic strength49, enabling them to be injected into tissues in vivo,

where they have been shown to encourage cell infiltration7. In addition, bioactive

sequences have been appended to the RAD sequence without disrupting assembly14, 17, 47.

RAD has also been shown to support growth factor delivery: PDGF-BB and other

proteins have been adsorbed to RAD18, 27, SDF-1 was inserted directly onto the RAD

peptide sequence39, and IGF-1 was tethered to RAD through biotin-streptavidin-biotin

bonds8.

The purpose of this study was to engineer the self-assembling peptide, KLD, to deliver

the growth factors IGF-1 and TGF-β1 to encapsulated chondrocytes or bone marrow

stromal cells (BMSCs), respectively, in the context of cartilage tissue engineering. The

growth factors were either (1) pre-mixed with the peptide solution prior to assembly to

adsorb them to the hydrogel, or (2) tethered to the hydrogel with high-affinity prior to

assembly8.

Page 31: In vitro and in vivo Growth Factor Delivery to

31

2.2 Materials and Methods

Materials: KLD peptide with the sequence AcN-(KLDL)3-CNH2 and biotinylated-KLD

(bKLD) peptide with the sequence biotin-(aminocaproic acid)3-(KLDL)3 was synthesized

by the MIT Biopolymers Laboratory (Cambridge, MA) using an ABI Model 433A

peptide synthesizer with FMOC protection or received as a gift from 3DM (3DM, Inc.,

Cambridge, MA). Biotin-conjugated IGF-1 (bIGF-1) (immunological and biochemical

testsystems GmbH, Reutlingen Germany) and biotin-conjugated TGF-β1 (bTGF-β1)

(R&D Systems, Minneapolis, MN) were purchased and used as described below.

Chondrocyte Isolation and Encapsulation: Chondrocytes were isolated from 1-2 week

old bovine calves (Research 87, Marlborough, MA) as described previously38.

Chondrocytes were encapsulated in KLD using acellular agarose molds to initiate self-

assembly as outlined below26, resulting in 6mm diameter, 50 µL peptide gel disks. To

verify bioactivity of bIGF-1, chondrocytes were encapsulated at 12x106 cells/mL in KLD

peptide (0.35% w/v) alone and cultured in IGF-1-free basal medium supplemented with

either soluble IGF-1 (PeproTech Inc., Rocky Hill, NJ), soluble bIGF-1, or soluble bIGF-

1/streptavidin at indicated concentrations (ng/mL); n=3-4. Basal medium consisted of

serum-free high-glucose Dulbecco’s modified Eagle’s medium (DMEM; Invitrogen,

Carlsbad, CA) supplemented with 0.003% ITS+1 (Sigma-Aldrich, St. Louis, MO), 1 mM

sodium pyruvate, 10 mM HEPES buffer, 0.1 mM nonessential amino acids, 0.4 mM

proline, 20 µg/mL ascorbic acid, 100 units/mL penicillin, 100 µg/mL streptomycin, and

0.25 µg/mL amphotericin B. This choice of concentration of ITS+1 is equivalent to 5 nM

Page 32: In vitro and in vivo Growth Factor Delivery to

32

insulin, termed mini-ITS2, 30, and chosen in order to avoid cross-talk of insulin with the

IGF-1 receptor.

For high-affinity tethering of IGF-1, control, soluble, and tethered gels were created by

pre-mixing KLD peptide (0.35% w/v) with 0.0035% w/v bKLD and encapsulating

chondrocytes at 3x106 cells/mL; n=4. For tethered gels, bIGF-1 and fluorescent

streptavidin-AlexaFluor 488 (Invitrogen) were pre-mixed at equimolar concentrations

and added to the KLD/bKLD mixture prior to adding cells. For adsorbed gels, bIGF-1

and fluorescent-streptavidin were pre-mixed as for the tethered gels, but no bKLD was

used in this case. Chondrocyte gels were cultured in basal medium and supplemented

with soluble IGF-1 for the soluble condition (soluble IGF-1 replenished at each medium

change).

BMSC Isolation: Bone marrow was harvested from 1-2 week old bovine calves

(Research 87) and stromal cells (BMSCs) were isolated as described previously6, 26.

BMSCs were selected via differential adhesion and expanded two passages in low

glucose-DMEM with 10% ES-FBS (Invitrogen), 10mM HEPES, 100 units/mL penicillin

G, 100 µg/mL streptomycin, 0.25 µg/mL amphotericin B, and 5 ng/mL FGF-2 (R&D

Systems).

BMSC Encapsulation: BMSCs were encapsulated in KLD mixtures at 3x106 cells/mL

using acellular agarose molds to initiate self-assembly26. For no TGF (n=8) and soluble

TGF (n=12) conditions, BMSCs were encapsulated in KLD peptide (0.35% w/v) alone.

Page 33: In vitro and in vivo Growth Factor Delivery to

33

For adsorbed gels (n=16), 0.35% KLD was pre-mixed with 100 ng/mL TGF-β1 (R&D

Systems) and 100 nM dexamethasone (Sigma-Aldrich, St. Louis, MO) prior to adding

cells. For tethered gels (n=4), 0.35% w/v KLD was pre-mixed with 0.0035% w/v bKLD,

2.1 µg/mL streptavidin (Pierce Biotechnology, Rockford, IL) and 100 or 500 ng/mL

bTGF-β1 prior to adding cells. The resulting 6mm diameter, 50 µL peptide gel disks were

cultured in high glucose-DMEM (Invitrogen) supplemented with 1% ITS+1 (Sigma-

Aldrich), 1 mM sodium pyruvate, 37.5 µg/mL ascorbate-2-phosphate (Wako Chemicals,

Richmond, VA), PSA (100 U/mL penicillin, 0.1 mg/mL streptomycin, 0.25 µg/mL

amphoteracin), 10 mM HEPES, 0.4 mM proline, and 0.1 mM non-essential amino acids.

Soluble gels had additional supplementation of 10 ng/mL recombinant human TGF-β1

(R&D Systems) (replenished at each medium change). No TGF, soluble, and tethered

gels had additional supplementation of 100 nM dexamethasone (replenished at each

medium change).

Cell Viability and Cell-Seeded Hydrogel Biochemistry: Viability was determined by

staining with FDA (live) and ethidium bromide (dead). There were no differences in

viability among conditions for chondrocyte or BMSC experiments, and all experiments

had >75% viability. At each timepoint, gels were digested with 250 µg/mL proteinase-K

(Roche Applied Science, Indianapolis, IN) overnight at 60 ºC. sGAG in each hydrogel

sample was assessed via DMMB dye binding12; DNA was quantified by Hoechst dye

assay23. For chondrocyte gels, digests were analyzed for presence of fluorescent

streptavidin via fluorometer reading at 485/535 nm.

Page 34: In vitro and in vivo Growth Factor Delivery to

34

Statistical Analyses: All data are presented as mean ± standard error of the mean (SEM).

Chondrocyte data were analyzed by one-way ANOVA at each timepoint. BMSC data

were analyzed using a mixed model of variance at each timepoint with animal cell-source

as a random factor. Residual plots were constructed for dependent variable data to test for

normality and data were log transformed if necessary to satisfy this assumption. Post hoc

Tukey tests for significance of pairwise comparisons were performed with a threshold for

significance of p<0.05.

2.3 Results

IGF-1 can be tethered to peptide hydrogels: Using a biotin-sandwich approach as

previously described for a related peptide sequence, RAD8, we tethered IGF-1 to KLD

scaffolds by including 1:100 ratio by weight of biotinylated KLD (bKLD):KLD and pre-

mixing streptavidin with a biotinylated version of the growth factor (bIGF-1). Multivalent

streptavidin and bIGF-1 were mixed in equimolar amounts so that the predominant

complex formed would consist of a single biotinylated growth factor bound to a single

streptavidin molecule, allowing streptavidin to bind bKLD with one of its three

remaining binding sites. Excess bKLD (>130:1 molar ratio bKLD:streptavidin) was used

to ensure that all of the delivered growth factor was tethered. To validate this tethering

approach relative to simple adsorption, we used fluorescently tagged streptavidin to track

the streptavidin/bIGF-1 complex in peptide hydrogels with encapsulated chondrocytes.

Although adsorbed and tethered gels were loaded with equivalent amounts of

streptavidin/bIGF-1, tethered gels retained significantly more bIGF-1 immediately after

encapsulation (Fig 2.1A). After one day, adsorbed streptavidin/bIGF-1 had decreased to

Page 35: In vitro and in vivo Growth Factor Delivery to

35

10-17% of the day 0 levels, and by day 8 it was only 2-6% of day 0 (Fig 2.1B,C).

Tethering allowed amounts to remain at 20-27% of the day 0 levels even by day 8 (Fig

2.1B,C). In contrast, when TGF-β1 is adsorbed to KLD peptide hydrogels ~40% is

retained at day 21 (data not shown).

bIGF is biologically active: To ensure that bIGF-1 had the same biological activity as

normal IGF-1, chondrocytes encapsulated in KLD were stimulated with 100 ng/mL of

soluble bIGF-1 or soluble IGF-1 for 4 days. sGAG content of the gels treated with IGF-1

and bIGF-1 was shown to be equivalent, and both conditions produced significantly more

sGAG than gels incubated without IGF-1 (Fig 2.2A). DNA content was not significantly

different among the treatments (Fig 2.2B). In addition, 100 ng/mL of soluble bIGF-1 was

compared to soluble complexes of 100 ng/mL bIGF-1 pre-mixed with an equimolar

amount of streptavidin to rule out any effect of binding to streptavidin on sGAG

production. Again, after 4 days, bIGF-1 with and without streptavidin produced

equivalent amounts of sGAG, significantly more than gels incubated without IGF-1 (Fig

2.2C), and DNA was equivalent among the treatments (Fig 2.2D). The bioactivity of

bTGF-β1 was also confirmed in similar experiments (data not shown).

Adsorbed and Tethered IGF-1 with chondrocytes: Next, bIGF-1/streptavidin

complexes were either adsorbed to KLD prior to chondrocyte encapsulation or tethered to

KLD prior to encapsulation. Concentrations of bIGF-1 in the KLD hydrogel were varied

for the adsorbed and tethered conditions such that cells would be exposed to amounts

known to stimulate proteoglycan synthesis (>50 ng/mL)13. Although concentrations up to

Page 36: In vitro and in vivo Growth Factor Delivery to

36

1000 ng/mL were tested for adsorbed and tethered bIGF-1, neither method of delivering

IGF-1 was able to stimulate sGAG production over no IGF-1 levels. This outcome for

adsorption of IGF-1 is consistent with Fig 1, which showed adsorbed bIGF-1/streptavidin

rapidly diffused out of KLD (Fig 2.3A). But the lack of bioactivity for tethering was

unexpected since sufficient levels were present throughout the culture period. Gels

treated with 50 ng/mL and 300 ng/mL of soluble IGF-1 produced significantly greater

amounts of sGAG than control IGF-1-free gels at all timepoints, as expected (Fig 2.3A).

DNA levels increased over time but were not different than control IGF-1-free gels for

any treatment at any timepoint (Fig 2.3B).

Adsorbed and Tethered TGF-β1 with BMSCs: We next tested the hypothesis that

since TGF-β1 adsorbed more strongly than IGF-1, it would result in greater proteoglycan

stimulus than no TGF-β1; we also investigated whether tethered TGF-β1 would promote

proteoglycan synthesis. BMSCs were stimulated with either soluble TGF-β1 and soluble

dexamethasone, adsorbed TGF-β1 and adsorbed dexamethasone, or tethered TGF-β1

with soluble dexamethasone. In contrast to IGF-1, adsorbed TGF-β1 was able to

stimulate BMSC sGAG production significantly higher than no TGF-β1 at all timepoints.

By day 21, adsorbed TGF-β1 stimulated 31% as much sGAG as soluble TGF-β1 (Fig

2.4A). Tethering TGF-β1 at up to 500 ng/mL did not stimulate sGAG production by

BMSCs, similar to results seen with chondrocytes exposed to tethered IGF-1. In addition,

tethering at 500 ng/mL significantly inhibited DNA accumulation compared to TGF-β1-

free gels at days 14 and 21 (Fig 2.4B). Soluble TGF-β1 stimulated an increase in DNA

Page 37: In vitro and in vivo Growth Factor Delivery to

37

over TGF-β1-free gels by day 21; adsorbed gels were not different from TGF-β1-free

gels at any timepoint (Fig 2.4B).

2.4 Discussion

The optimal strategy for delivering growth factors to cells for the purposes of cartilage

tissue engineering and cartilage repair remains an unmet challenge. Self-assembling

peptides are a clinically relevant material that can be injected in vivo7, 8 and assemble on

contact with solutions of physiologic pH and ionic strength49. We explored the ability of

the self-assembling peptide hydrogel KLD to deliver IGF-1 and TGF-β1 to cells through

two methods, tethering and adsorption of the growth factors to the peptide. While these

growth factors can be tethered to KLD, they do not promote the same stimulation of

matrix accumulation by chondrocytes or BMSCs compared to soluble delivery of these

factors. In addition, both IGF-1 and TGF-β1 are able to be adsorbed to KLD, but only

TGF-β1 is retained at sufficient quantity and duration to promote proteoglycan

production.

Although biotin-streptavidin tethering was shown by Davis et al. to be effective in

delivering IGF-1 in order to reduce apoptosis of implanted cardiomyocytes8, it was not

effective in delivering IGF-1 or TGF-β1 to stimulate proteoglycan production by

chondrocytes or BMSCs, respectively, in the context of cartilage tissue engineering. Due

to the strength of the non-covalent biotin-streptavidin bond (KD = 4x10-14 M)16 it is

unlikely that the growth factors are released by this tether. Instead, the loss of bIGF-1 by

Page 38: In vitro and in vivo Growth Factor Delivery to

38

day 8 shown in Fig 1 is probably due to the degradation of the gel resulting in loss of the

bKLD fibers to which the growth factors are attached. A difference in peptide gel

degradation rates caused by differences in sequence, (RADA)4 vs. (KLDL)3, and by

differences in cell type could therefore partly explain the differences between this study

and that of Davis et al.8. Cartilage tissue also differs from myocardial tissue in that large

amounts of extracellular matrix are produced, as early as one day after encapsulation.

This matrix synthesis and secretion may prevent the stimulation of chondrocytes and

BMSCs by immobilized factors by sterically blocking receptor-ligand binding. In

addition, high-affinity tethering could block internalization of the ligand-receptor

complex, which could limit bioactivity. Internalization of receptors is not required for

TGF-β1 Smad2 signaling31, while IGF-1 signaling is thought to involve both cell surface

and endosomal signaling4. However, inhibition of internalization may lead to altered

signaling, resulting in lack of proteoglycan stimulation. Finally, while the density of

tethered growth factors may also affect bioactivity42, both IGF-1 and TGF-β receptors

exist as functional complexes on cell surfaces, and binding of one ligand can initiate

signaling10, 41. Therefore, the distance between these immobilized growth factors should

not have inhibited their effectiveness. Increasing the seeding density of chondrocytes

(data not shown) or increasing the amount of growth factors immobilized within the

scaffold had no effect.

Growth factors, including TGF-β1, have been covalently immobilized or tethered onto

other polymer scaffolds or hydrogels1, 9, 21, 22, 29, 35, although sometimes with altered

signaling11, 36, 40. There have been a limited number of other attempts at delivering

Page 39: In vitro and in vivo Growth Factor Delivery to

39

tethered growth factors for the purposes of cartilage tissue engineering37. A possible

solution to increase bioactivity of our tethering may be to include cleavable links in our

peptide sequence. Lutolf et al. have proposed using MMP-cleavable links to deliver

growth factors in poly(ethylene glycol)-based hydrogels upon catabolic events32, 33. In

addition, Segers et al. directly inserted the protein SDF-1 onto the RAD peptide sequence

with an MMP-cleavable linkage39. This idea may be adapted to our system by including

such sequences appended to bKLD so that upon MMP cleavage, the growth factor

complexes would become soluble. Other sequences may be more applicable for

chondrogenic purposes, including sequences cleavable by the aggrecanases, ADMATS-

4/5. Finally, using tethers with a lower binding affinity may allow this method to result in

better biological activity.

Other methods for slow release of IGF-1 and TGF-β1 have been developed (see 28, 45 for

review), but IGF-1 remains difficult to effectively deliver in vivo due to its small size and

rapid diffusion out of tissue. A new fusion protein made by adding the heparin-binding

domain of heparin-binding EGF to IGF-1 (i.e. HB-IGF-1) has been found to be amenable

to delivery in mature articular cartilage46. A single dose of HB-IGF-1 resulted in

sustained delivery and stimulation of proteoglycan synthesis for at least 8 days. The use

of HB-IGF-1 with the KLD peptide system could be enabled by mixing in heparin,

heparan sulfate, or the heparan sulfate proteoglycan, perlecan, with KLD. This approach

has been shown to be effective in delivering other heparin-binding growth factors such as

FGF-2 and BMP-2 to cells within fibrin19, collagen type 148, and PLGA20 scaffolds.

Although adsorption of TGF-β1 to KLD effectively increased proteoglycan production in

Page 40: In vitro and in vivo Growth Factor Delivery to

40

our study, changing the degradation rates of KLD by changing the concentration or

adding cross-links may offer a way of improving delivery.

This study provided an initial evaluation of methods for delivering growth factors to cells

within the self-assembling peptide hydrogel KLD, and showed that while TGF-β1 is able

to be delivered by adsorption, IGF-1 is not. Additionally, while tethering these factors

through a biotin-streptavidin bond provided long-term sequestration, tethered growth

factors were not effective in stimulating proteoglycan production. Therefore, while

peptide sequences are readily functionalized, the manner in which growth factors are

delivered affects bioactivity and varies for specific growth factors and biological systems

of interest.

2.5 Acknowledgements

I would like to thank Paul W. Kopesky, Ph.D. for his help in planning and executing this

study and Alan J. Grodzinsky, Sc.D. for his scientific guidance. I would also like to thank

Emily Florine, Dr. David Frisbie, Dr. John Kisiday, Dr. Bodo Kurz, Dr. Richard Lee, and

Dr. Eric Vanderploeg for helpful discussions. The following funding sources were

instrumental for this work: NIH-NIBIB EB003805, NIH-NIAMS AR33236; NSF and

NDSEG graduate fellowships, Siebel Foundation Scholar fellowship.

Page 41: In vitro and in vivo Growth Factor Delivery to

41

2.6 References

1. Bentz H, Schroeder JA, and Estridge TD. Improved local delivery of TGF-beta2 by binding to injectable fibrillar collagen via difunctional polyethylene glycol. Journal of Biomedical Materials Research. 1998; 39:539-548.

2. Benya PD and Padilla SR. Dihydrocytochalasin B Enhances Transforming Growth Factor-[beta]-Induced Reexpression of the Differentiated Chondrocyte Phenotype without Stimulation of Collagen Synthesis. Experimental Cell Research. 1993; 204:268-277.

3. Blaney Davidson EN, van der Kraan PM, and van den Berg WB. TGF-[beta] and osteoarthritis. Osteoarthritis and Cartilage. 2007; 15:597-604.

4. Chow JC, Condorelli G, and Smith RJ. Insulin-like growth factor-I receptor internalization regulates signaling via the Shc/mitogen-activated protein kinase pathway, but not the insulin receptor substrate-1 pathway. J Biol Chem. 1998; 273:4672-80.

5. Chung C and Burdick JA. Engineering cartilage tissue. Adv Drug Deliv Rev. 2008; 60:243-62.

6. Connelly JT, Garcia AJ, and Levenston ME. Inhibition of in vitro chondrogenesis in RGD-modified three-dimensional alginate gels. Biomaterials. 2007; 28:1071-83.

7. Davis ME, Motion JPM, Narmoneva DA, Takahashi T, Hakuno D, Kamm RD, Zhang S, and Lee RT. Injectable Self-Assembling Peptide Nanofibers Create Intramyocardial Microenvironments for Endothelial Cells. Circulation. 2005; 111:442-450.

8. Davis ME, Hsieh PCH, Takahashi T, Song Q, Zhang S, Kamm RD, Grodzinsky AJ, Anversa P, and Lee RT. Local myocardial insulin-like growth factor 1 (IGF-1) delivery with biotinylated peptide nanofibers improves cell therapy for myocardial infarction. Proc Natl Acad Sci USA. 2006; 103:8155-8160.

9. DeLong SA, Moon JJ, and West JL. Covalently immobilized gradients of bFGF on hydrogel scaffolds for directed cell migration. Biomaterials. 2005; 26:3227-34.

10. Derynck R and Zhang YE. Smad-dependent and Smad-independent pathways in TGF-β family signalling. Nature. 2003; 425:577-84.

11. Fan VH, Au A, Tamama K, Littrell R, Richardson LB, Wright JW, Wells A, and Griffith LG. Tethered Epidermal Growth Factor Provides a Survival Advantage to Mesenchymal Stem Cells. Stem Cells. 2007; 25:1241-1251.

12. Farndale R, Sayers C, and Barrett A. A direct spectrophotometric microassay for sulfated glycosaminoglycans in cartilage cultures. Connect Tissue Res. 1982; 9:247-248.

13. Fortier LA, Mohammed HO, Lust G, and Nixon AJ. Insulin-like growth factor-I enhances cell-based repair of articular cartilage. J Bone Joint Surg Br. 2002; 84:276-88.

14. Genové E, Shen C, Zhang S, and Semino CE. The effect of functionalized self-assembling peptide scaffolds on human aortic endothelial cell function. Biomaterials. 2005; 26:3341-3351.

Page 42: In vitro and in vivo Growth Factor Delivery to

42

15. Getgood A, Brooks R, Fortier L, and Rushton N. Articular cartilage tissue engineering: today's research, tomorrow's practice? J Bone Joint Surg Br. 2009; 91:565-76.

16. Holmberg A, Blomstergren A, Nord O, Lukacs M, Lundeberg J, and Uhlen M. The biotin-streptavidin interaction can be reversibly broken using water at elevated temperatures. Electrophoresis. 2005; 26:501-10.

17. Horii A, Wang X, Gelain F, and Zhang S. Biological designer self-assembling peptide nanofiber scaffolds significantly enhance osteoblast proliferation, differentiation and 3-D migration. PLoS One. 2007; 2:e190.

18. Hsieh PCH, Davis ME, Gannon J, MacGillivray C, and Lee RT. Controlled delivery of PDGF-BB for myocardial protection using injectable self-assembling peptide nanofibers. The Journal of Clinical Investigation. 2006; 116:237-248.

19. Jeon O, Ryu SH, Chung JH, and Kim BS. Control of basic fibroblast growth factor release from fibrin gel with heparin and concentrations of fibrinogen and thrombin. J Control Release. 2005; 105:249-59.

20. Jeon O, Song SJ, Kang SW, Putnam AJ, and Kim BS. Enhancement of ectopic bone formation by bone morphogenetic protein-2 released from a heparin-conjugated poly(L-lactic-co-glycolic acid) scaffold. Biomaterials. 2007; 28:2763-71.

21. Kapur TA and Shoichet MS. Immobilized concentration gradients of nerve growth factor guide neurite outgrowth. J Biomed Mater Res A. 2004; 68:235-43.

22. Kim EJ, Kang IK, Jang MK, and Park YB. Preparation of insulin-immobilized polyurethanes and their interaction with human fibroblasts. Biomaterials. 1998; 19:239-49.

23. Kim Y-J, Sah RLY, Doong J-YH, and Grodzinsky AJ. Fluorometric assay of DNA in cartilage explants using Hoechst 33258. Analytical Biochemistry. 1988; 174:168-176.

24. Kisiday J, Jin M, Kurz B, Hung H, Semino C, Zhang S, and Grodzinsky AJ. Self-assembling peptide hydrogel fosters chondrocyte extracellular matrix production and cell division: implications for cartilage tissue repair. Proc Natl Acad Sci U S A. 2002; 99:9996-10001.

25. Knutsen G, Drogset JO, Engebretsen L, Grontvedt T, Isaksen V, Ludvigsen TC, Roberts S, Solheim E, Strand T, and Johansen O. A randomized trial comparing autologous chondrocyte implantation with microfracture. Findings at five years. J Bone Joint Surg Am. 2007; 89:2105-12.

26. Kopesky P, Vanderploeg E, Sandy J, Kurz B, and Grodzinsky AJ. Self-assembling peptide hydrogels modulate in vitro chondrogenesis of bovine bone marrow stromal cells. Tissue Eng Part A. 2010; In press.

27. Koutsopoulos S, Unsworth LD, Nagai Y, and Zhang S. Controlled release of functional proteins through designer self-assembling peptide nanofiber hydrogel scaffold. Proc Natl Acad Sci USA. 2009; 106:4623-4628.

28. Lee S-H and Shin H. Matrices and scaffolds for delivery of bioactive molecules in bone and cartilage tissue engineering. Advanced Drug Delivery Reviews. 2007; 59:339-359.

29. Liu HW, Chen CH, Tsai CL, Lin IH, and Hsiue GH. Heterobifunctional poly(ethylene glycol)-tethered bone morphogenetic protein-2-stimulated bone

Page 43: In vitro and in vivo Growth Factor Delivery to

43

marrow mesenchymal stromal cell differentiation and osteogenesis. Tissue Eng. 2007; 13:1113-24.

30. Loeser RF and Shanker G. Autocrine stimulation by insulin-like growth factor 1 and insulin-like growth factor 2 mediates chondrocyte survival in vitro. Arthritis Rheum. 2000; 43:1552-9.

31. Lu Z, Murray JT, Luo W, Li H, Wu X, Xu H, Backer JM, and Chen Y-G. Transforming Growth Factor b Activates Smad2 in the Absence of Receptor Endocytosis. J Biol Chem. 2002; 277:29363-29368.

32. Lutolf MP, Lauer-Fields JL, Schmoekel HG, Metters AT, Weber FE, Fields GB, and Hubbell JA. Synthetic matrix metalloproteinase-sensitive hydrogels for the conduction of tissue regeneration: engineering cell-invasion characteristics. Proc Natl Acad Sci U S A. 2003; 100:5413-8.

33. Lutolf MP, Weber FE, Schmoekel HG, Schense JC, Kohler T, Muller R, and Hubbell JA. Repair of bone defects using synthetic mimetics of collagenous extracellular matrices. Nat Biotechnol. 2003; 21:513-8.

34. Maher SA, Mauck RL, Rackwitz L, and Tuan RS. A nanofibrous cell-seeded hydrogel promotes integration in a cartilage gap model. J Tissue Eng Regen Med. 2010; 4:25-9.

35. Mann BK, Schmedlen RH, and West JL. Tethered-TGF-β increases extracellular matrix production of vascular smooth muscle cells. Biomaterials. 2001; 22:439-444.

36. Marcantonio NA, Boehm CA, Rozic RJ, Au A, Wells A, Muschler GF, and Griffith LG. The influence of tethered epidermal growth factor on connective tissue progenitor colony formation. Biomaterials. 2009; 30:4629-4638.

37. Motoyama M, Deie M, Kanaya A, Nishimori M, Miyamoto A, Yanada S, Adachi N, and Ochi M. In vitro cartilage formation using TGF-beta-immobilized magnetic beads and mesenchymal stem cell-magnetic bead complexes under magnetic field conditions. J Biomed Mater Res A. 2010; 92:196-204.

38. Ragan PM, Chin VI, Hung HH, Masuda K, Thonar EJ, Arner EC, Grodzinsky AJ, and Sandy JD. Chondrocyte extracellular matrix synthesis and turnover are influenced by static compression in a new alginate disk culture system. Arch Biochem Biophys. 2000; 383:256-64.

39. Segers VFM, Tokunou T, Higgins LJ, MacGillivray C, Gannon J, and Lee RT. Local Delivery of Protease-Resistant Stromal Cell Derived Factor-1 for Stem Cell Recruitment After Myocardial Infarction. Circulation. 2007; 116:1683-1692.

40. Shen YH, Shoichet MS, and Radisic M. Vascular endothelial growth factor immobilized in collagen scaffold promotes penetration and proliferation of endothelial cells. Acta Biomaterialia. 2008; 4:477-489.

41. Siddle K, Urso B, Niesler CA, Cope DL, Molina L, Surinya KH, and Soos MA. Specificity in ligand binding and intracellular signalling by insulin and insulin-like growth factor receptors. Biochem Soc Trans. 2001; 29:513-25.

42. Sofia SJ, Kuhl PR, and Griffith LG, Preparation and Use of Tethered Ligands as Biomaterials and Tools for Cell Biology, in Tissue Engineering Methods and Protocols. 1999. p. 19-33.

Page 44: In vitro and in vivo Growth Factor Delivery to

44

43. Sohier J, Moroni L, van Blitterswijk C, de Groot K, and Bezemer JM. Critical factors in the design of growth factor releasing scaffolds for cartilage tissue engineering. Expert Opin Drug Deliv. 2008; 5:543-66.

44. Steinert AF, Noth U, and Tuan RS. Concepts in gene therapy for cartilage repair. Injury. 2008; 39 Suppl 1:S97-113.

45. Stoop R. Smart biomaterials for tissue engineering of cartilage. Injury. 2008; 39 Suppl 1:S77-87.

46. Tokunou T, Miller R, Patwari P, Davis ME, Segers VF, Grodzinsky AJ, and Lee RT. Engineering insulin-like growth factor-1 for local delivery. FASEB J. 2008; 22:1886-93.

47. Wang X, Horii A, and Zhang S. Designer functionalized self-assembling peptide nanofiber scaffolds for growth, migration, and tubulogenesis of human umbilical vein endothelial cells. Soft Matter. 2008; 4:2388-2395.

48. Yang WD, Gomes RR, Alicknavitch M, Farach-Carson MC, and Carson DD. Perlecan Domain I Promotes Fibroblast Growth Factor 2 Delivery in Collagen I Fibril Scaffolds. Tissue Engineering. 2005; 11:76-89.

49. Zhang S, Holmes T, Lockshin C, and Rich A. Spontaneous assembly of a self-complementary oligopeptide to form a stable macroscopic membrane. Proc Natl Acad Sci U S A. 1993; 90:3334-8.

Page 45: In vitro and in vivo Growth Factor Delivery to

45

2.7 Figures

Figure 2.1. High-affinity tethering prolongs retention of IGF-1: Fluorescently labeled streptavidin and biotinylated-IGF-1 were either adsorbed or tethered to KLD prior to encapsulation of chondrocytes at 3x106 cells/mL. Background from the peptide was measured on gels without fluorescent streptavidin and subtracted from fluorescence counts. (A) Streptavidin retained immediately after gel assembly. (B) Streptavidin retained after 1, 4, or 8 days of culture. * vs. corresponding concentration adsorbed gel, p<0.05. mean ± SEM. (C) Fluorescence microscopy on gels at day 8 on gels without bIGF-1/fluorescent streptavidin, gels with 1000 ng/mL adsorbed bIGF-1/fluorescent streptavidin, or gels with 1000 ng/mL tethered bIGF-1/fluorescent streptavidin. Scale bar = 250 µm.

Page 46: In vitro and in vivo Growth Factor Delivery to

46

Figure 2.2. bIGF-1 is bioactive: Chondrocytes were encapsulated in KLD at 12x106 cells/mL and cultured in medium with different soluble factors for four days. (A) sGAG and (B) DNA retained after culture in medium with soluble IGF-1 or soluble bIGF-1. (C) sGAG and (D) DNA retained after culture in medium with soluble bIGF-1 or soluble bIGF-1 + soluble streptavidin. mean ± SEM, * vs. No IGF, p<0.05.

Page 47: In vitro and in vivo Growth Factor Delivery to

47

Figure 2.3. Soluble IGF-1, but not adsorbed or tethered IGF-1, stimulates sGAG production: Biotinylated IGF-1 and streptavidin were either adsorbed or tethered to KLD at the indicated concentrations prior to encapsulation of chondrocytes at 3x106 cells/mL. No IGF-1 control gels were cultured in IGF-1-free medium. (A) sGAG and (B) DNA retained in gel. mean ± SEM, * vs. No IGF, p<0.05.

Page 48: In vitro and in vivo Growth Factor Delivery to

48

Figure 2.4. Soluble and adsorbed TGF-β1, but not tethered, promotes sGAG production: TGF-β1 and dexamethasone were adsorbed to KLD prior to assembly or biotinylated TGF-β1 and streptavidin were tethered to KLD prior to encapsulation of BMSCs at 3x106 cells/mL. No TGF control gels were cultured in TGF-β1-free medium. Soluble dexamethasone (100 nM) was included in the medium for all conditions but adsorbed. (A) sGAG and (B) DNA retained in gel. mean ± SEM, * vs. No TGF, p<0.05.

Page 49: In vitro and in vivo Growth Factor Delivery to

49

Chapter 3. Effect of Self-assembling Peptide, Chondrogenic Factors, and Bone-Marrow-Derived Stromal Cells on Osteochondral Repair

The goal of this study was to test the ability of an injectable self-assembling peptide

(KLD) hydrogel with or without chondrogenic factors (CF) and allogeneic bone marrow

stromal cells (BMSCs) to stimulate cartilage regeneration in a full-thickness, critically

sized, rabbit cartilage defect model in vivo. We used CF treatments to test the hypotheses

that CF would stimulate chondrogenesis and matrix production by cells migrating into

acellular KLD (KLD+CF) or by BMSCs delivered in KLD (KLD+CF+BMSCs). Three

groups were tested against contralateral untreated controls: KLD, KLD+CF, and

KLD+CF+BMSCs, n=6-7. TGF-β1, dexamethasone, and IGF-1 were used as

chondrogenic factors (CF) pre-mixed with KLD and BMSCs before injection.

Evaluations included gross, histological, immunohistochemical and radiographic analyses.

KLD without CF or BMSCs showed the greatest repair after 12 weeks with significantly

higher Safranin-O, collagen II immunostaining, and cumulative histology scores than

untreated contralateral controls. KLD+CF resulted in significantly higher aggrecan

immunostaining than untreated contralateral controls. Including allogeneic BMSCs+CF

markedly reduced the quality of repair and increased osteophyte formation compared to

KLD alone. These data show that KLD can fill full-thickness osteochondral defects in

situ and improve cartilage repair as shown by Safranin-O, collagen II immunostaining,

and cumulative histology. In this small animal model, the full-thickness critically sized

defect provided access to the marrow, similar in concept to abrasion arthroplasty or

Page 50: In vitro and in vivo Growth Factor Delivery to

50

spongialization in large animal models, and suggests that combining KLD with these

techniques may improve current practice.

3.1 Introduction

Repair of articular cartilage injuries remains a challenge, despite the development of

surgical treatments such as microfracture, abrasion arthroplasty, and spongialization,

which are used with the goal of recruiting marrow-derived cells by penetration of the

subchondral bone. While these techniques promote increased short-term healing, long-

term repair still consists of mechanically inferior fibrocartilage13. Recent research has

focused on tissue engineering strategies using scaffolds to improve cartilage repair and

regeneration. In particular, hydrogels made from materials such as chitosan-glycerol

phosphate16, polyethyleneglycol (PEG)44, fibrin36, polyglycolic-co-lactic acid (PLGA)46,

and collagen11 have been explored with the goal of improving the accumulation of

extracellular matrix produced by cells (e.g., bone marrow stromal cells (BMSCs))

migrating into the scaffold from marrow.

Stimulation of BMSC chondrogenesis in vitro and in vivo has received much attention. In

vitro studies have shown that transforming growth factor-β1 (TGF-β1), dexamethasone,

and insulin-like growth factor-1 (IGF-1) promote chondrogenesis of BMSCs22, 24, 25, 31, 49,

and methods for delivering these chondrogenic factors have been developed, often in

conjunction with scaffolds5, 17, 23, 38. There have been a number of in vivo studies

performed delivering IGF-136, 40, TGF-β12, 13, 32, or the combination of IGF-1 and TGF-

Page 51: In vitro and in vivo Growth Factor Delivery to

51

β117, 19, 40 to cartilage defects in order to stimulate chondrogenesis of endogenous

BMSCs, but to our knowledge, no in vivo studies have incorporated dexamethasone.

The hypothesis that delivery of exogenous BMSCs to the joint can enhance cartilage

regeneration has prompted the exploration of a wide variety of growth factor and scaffold

combinations to stimulate BMSC chondrogenesis13, 20, 37. In vivo studies have attempted

to deliver BMSCs alone12, 35, BMSCs encapsulated in scaffolds30, 43, 48, and BMSCs

encapsulated in scaffolds with the inclusion of TGF-β14, 7, 14, 15. Despite the in vitro

promise shown by 3D-cultured BMSCs, most long-term in vivo treatments with BMSCs

have resulted in sub-optimal cartilage repair tissue12, 20, 43, 48. Improving BMSC

chondrogenesis in vivo is likely dependent on several factors that are not well understood,

including cell delivery, microenvironment, and a combination of pro-chondrogenic

longitudinal signaling. In addition, an ideal clinical approach would minimize or obviate

the in vitro culture duration and be performed with a single arthroscopic procedure.

Recent studies have shown that hydrogels made from the self-assembling peptide

sequences (RADA)4 and (KLDL)3 (hereafter referred to as KLD) can maintain the

chondrocyte phenotype26 and stimulate chondrogenesis of BMSCs in vitro6, 27, 29. These

hydrogels have the ability to rapidly assemble when exposed to physiological pH and

ionic strength50 and have pore sizes in the range of 100-500 nm42. These synthetic

peptides have been used in vivo without immunogenic reaction3. Furthermore, TGF-β1

has been shown to adsorb to KLD when pre-mixed with the peptide solution prior to

assembly, resulting in extended delivery of TGF-β1 to BMSCs and stimulating

Page 52: In vitro and in vivo Growth Factor Delivery to

52

chondrogenesis in vitro, promoting sGAG production and accumulation comparable to

continuous medium supplementation of TGF-β1 over 21 days28. In addition, including

dexamethasone with TGF-β1 in medium supplementation during the in vitro culture of

BMSCs in RADA results in less catabolic cleavage of aggrecan compared to culture with

TGF-β1 alone8. Finally, IGF-1 can be tethered to peptide scaffolds via biotin-streptavidin

bonds; this tethered IGF-1 has been shown to remain biologically active and to promote

cell survival in rat cardiomyocytes over 28 days3.

The goal of this study was to test the ability of an injectable KLD hydrogel with or

without BMSCs and chondrogenic factors (CF) to stimulate cartilage regeneration in vivo

in a critically sized rabbit full-thickness cartilage defect model. This model provides

access to the marrow, analogous to abrasion arthroplasty or spongialization in large

animal models. We used CF treatments (IGF-1, TGF-β1, and dexamethasone) to test the

hypotheses that CF would stimulate chondrogenesis and matrix production (1) by cells

migrating into acellular KLD and (2) by P2 passaged allogeneic BMSCs delivered in

KLD. IGF-1 was tethered to the peptide with a biotin-streptavidin bond3 to stimulate

long-term production of cartilage ECM, while TGF-β1 and dexamethasone were pre-

mixed with KLD prior to BMSC encapsulation to stimulate chondrogenesis and initial

matrix production. A 12-week timepoint enabled evaluation of mid-term benefits of the

treatment compared to contralateral untreated defects.

Page 53: In vitro and in vivo Growth Factor Delivery to

53

3.2 Materials and Methods

Materials: KLD peptide with the sequence AcN-(KLDL)3-CNH2 was synthesized by the

MIT Biopolymers Laboratory (Cambridge, MA) using an ABI Model 433A peptide

synthesizer with FMOC protection. Human recombinant TGF-β1 (R&D Systems, Inc.,

Minneapolis, MN), dexamethasone (Sigma-Aldrich, St. Louis, MO), sucrose (Sigma-

Aldrich), biotinylated-IGF-1 (bIGF-1) (immunological and biochemical testsystems

GmbH, Reutlingen Germany), streptavidin (Pierce Biotechnology, Inc, Rockford, IL),

biotinylated-KLD (biotin-(aminocaproic acid)3-(KLDL)3 or b-KLD) (MIT Biopolymers

Laboratory), FBS (Invitrogen, Carlsbad, CA), and FGF-2 (R&D Systems, Inc.) were

purchased and used as described.

Cell Isolation: Bone marrow was harvested and pooled from four rabbits used for an

initial pilot study, and BMSCs were isolated as previously described29. BMSCs were

selected via differential adhesion to plastic and expanded two passages in alphaMEM

with 10% FBS and 2 ng/mL FGF-2, 10 mM HEPES, and PSA (100 U/mL penicillin, 100

µg/mL streptomycin, and 250 ng/mL amphotericin), resulting in a total of approximately

four population doublings. Each passage was conducted by seeding at a concentration of

12x103 BMSCs/cm2 and incubating for two days to allow BMSCs to grow to ~75%

confluence.

In Vivo Study Design: All procedures were approved by the Animal Care and Use

Committees at Colorado State University and Massachusetts Institute of Technology.

Twenty skeletally mature, retired, female breeder New Zealand White rabbits (average

Page 54: In vitro and in vivo Growth Factor Delivery to

54

age 11 months and body weight 4.7 kg) were used for this study (Myrtle’s Rabbitry,

Thompson Station, TN). One rabbit died during the study due to neurologic problems

post-surgery and was not included in the analysis. The n values shown in Table 1 do not

reflect this animal. Three different groups were tested against contralateral, untreated,

empty controls: (1) KLD, (2) KLD+chondrogenic factors (CF), and (3)

KLD+CF+BMSCs (Table 3.1). For all groups, KLD was resuspended in 10% sterile

sucrose, and the final KLD concentration was kept constant at 3.2 mg/mL. For groups 2

and 3, KLD peptide (48 µg) was pre-mixed with a CF mixture consisting of 1.4 ng TGF-

β1, 0.6 ng dexamethasone, 4.1 ng biotinylated-IGF-1 (bIGF-1), 30.7 ng streptavidin, and

0.48 µg b-KLD. For group 3, 150x103 BMSCs were encapsulated in the KLD/CF

mixture29. Encapsulation of BMSCs in vitro in this manner resulted in 80-90% viability.

In vitro studies indicated that this amount of TGF-β1 would result in a concentration of

TGF-β1 inside the scaffold sufficient to stimulate chondrogenesis28. At the same time, if

all of the TGF-β1 were to be released from the scaffold at once into the joint space in vivo,

it would amount to approximately 1 ng/mL concentration in the joint space (1.4 ng / 1.4

mL joint space volume47), compared to the native concentration of 52.3 pg/mL found in

adult rabbits47. Streptavidin and bIGF-1 were mixed at a 1:1 molar ratio in order to

achieve, on average, binding of one bIGF-1 to each streptavidin allowing streptavidin to

still bind b-KLD (in 100x molar excess of streptavidin and bIGF-1 to ensure

homogeneous distribution of IGF-1 throughout the gel). The ability of this tethering to

occur using these molar ratios has been shown in vitro and in vivo3. The amount of bIGF-

1 tethered was chosen to provide a local concentration of 300 ng/mL inside the scaffold,

Page 55: In vitro and in vivo Growth Factor Delivery to

55

which is above the threshold shown to be sufficient for chondrocyte stimulation of

proteoglycan synthesis in vivo9.

Defect Creation and Gel Injection: All surgical procedures were performed under

inhalation general anesthesia. A medial-parapatellar arthrotomy approach to the

femoropatellar joint was performed, and the patella was luxated laterally. A 3mm-diam x

2mm-deep full-thickness, critically sized defect was created in the central region of the

femoral trochlear groove (Fig 3.1). Direct pressure was applied with a surgical sponge to

ensure all bleeding was stopped prior to application of the peptide, which was delivered

as a liquid. Defects were filled with designated treatments (15 µL volume) or left

untreated, as dictated by group assignment. The liquid peptide suspension could easily be

seen filling the defect, and defects were filled until visually full. At this time, Lactated

Ringer’s Solution was added to the joint periphery to gently fill the joint and cause

polymerization of the peptide, which was visually inspected to ensure retention of the

implant. Dorsal-caudal and lateral-medial 90º radiographs of each stifle joint were

obtained immediately after surgery. At 12 weeks, rabbits were euthanized with

pentobarbital after sedation. Post-surgical radiographic views were repeated following

euthanasia.

Gross Pathologic Observations of Joints: The limbs and joints were examined and

graded by a blinded observer (DDF) unaware of treatment group (Table 3.2). For the

muscle wastage measurement, the limbs were shaved and a flexible tape measure was

Page 56: In vitro and in vivo Growth Factor Delivery to

56

used in a similar anatomic location of the stifle to make this assessment. This scoring

system was chosen in order to compare to other studies performed by Frisbie et al.11

Synovial Membrane Histology: Synovial membrane was harvested and placed in

neutral-buffered 10% formalin, embedded in paraffin, 5-µm sections created and stained

with hematoxylin and eosin (H&E). Sections were evaluated blindly (DDF) for cellular

infiltration, vascularity, intimal hyperplasia, subintimal edema and subintimal fibrosis on

a scale of 0-4 (0=none, 1=slight, 2=mild, 3=moderate, 4=severe)10.

Articular Cartilage Histology: Femoral sections for histology were fixed in neutral-

buffered 10% formalin, decalcified, embedded in paraffin, sectioned at 5 µm, and stained

with either H&E or Safranin-O, fast green (SOFG) (ThermoScientific/Shandon VeriStain

Gemini ES stainer) for proteoglycan visualization. Sections stained with H&E were

evaluated blindly (DDF) using a modified O’Driscoll scoring system11, 39 (Table 3.3) with

a maximum cumulative histology score of 28 and a higher score indicating a repair more

like native cartilage. Sections stained with SOFG were evaluated blindly (DDF) for

intensity of staining on a scale of 0-3 (Table 3.3) and were included in the cumulative

histology score.

Articular Cartilage Immunohistochemistry: For immunohistochemical analyses,

femoral sections were snap frozen in OCT using liquid nitrogen and sectioned at 8 µm.

Each section was incubated in 0.25 U/mL chondroitinase ABC (Sigma-Aldrich) for 15

min before incubation in primary antibody solution (Collagen I (1:10) #M-38-s and II

Page 57: In vitro and in vivo Growth Factor Delivery to

57

(undiluted) # II-II6B3-s, Hybridoma Bank; Aggrecan (1:100, Alexis Biochemicals)

#ALX-803-313; or rabbit IgG as control). Endogenous peroxidase was blocked using

0.3% H2O2 in methanol. Sections were incubated in goat anti-mouse HRP secondary

antibody solution (1:500, Jackson Immunoresearch, Westgrove, PA)), stained with

Vector Nova RED (Vector Laboratories, Burlingame, CA) and counterstained with Fast

Green. Controls gave no signal. Non-calcified tissues were evaluated blindly (DDF) for

the percentage of repair tissue stained positive (0=no stain, 1=1-25%, 2=26-50%, 3=51-

75%, 4=76-100%).

Radiographic Analysis: Radiographs and photographs were taken of bone segments

from femoral sections. Radiographs were graded for presence of lysis, bony proliferation,

osteophyte presence, and patellar luxation, and a total radiographic score was calculated

by summing these scores (maximum score of 16). All grading was done on a 0-4 scale

(0=none, 1=slight, 2=mild, 3=moderate, 4=severe)10, 12. Additionally, radiographs post-

euthanasia were scored for healing of the defect (0=no healing, 1=slight healing, 2=mild

bone filling, 3=lesion is visible but difficult to measure, 4=lesion not visible to measure),

sclerosis around the defect, and other sclerosis (0-4, none-severe). Sclerosis is defined as

the increase in density of bone seen radiographically and was subjectively scored by

MFB in a blinded fashion.

Statistical Analyses: Scores were evaluated for inter- and intra-group differences using

an ANOVA framework with PROC GLIMMIX (fits generalized linear mixed models) of

SAS (Cary, NC)1 with rabbit as a random variable. For all joint pathologic, histological,

Page 58: In vitro and in vivo Growth Factor Delivery to

58

immunohistochemical, and radiographic analyses, intra-group differences were analyzed

with treatment set as the main effect (one-way); differences among groups were analyzed

using treatment and group as main effects (two-way). Untreated controls were therefore

analyzed separately depending on what treatment the contralateral joint received in order

to account for possible systemic effects18. When main or interaction effects had p-values

that were considered significant (p-value < 0.05) or a trend (0.05 < p-value <0.10),

individual comparisons were made using the least square means procedure12. P-values

presented in the text include which main effect or interaction term they are referring to if

quoted for comparisons among groups. All data are presented as mean ± standard error of

the mean (SEM). Non-parametric analyses were also performed when appropriate. The

authors were able to reach similar conclusions based on either analysis. Because the

authors feel many of the biologic outcome parameters represent more of a continuum

rather than defined categories, the parametric analysis of the data is presented.

3.3 Results

Radiographic Analyses: Radiographic analyses pre-treatment did not show any sclerosis

in any of the rabbits. Treated defects post-treatment showed some slight osteophyte

formation (0.333±0.139, treated; 0.056±0.143, untreated) (treatment p=0.087). There was

no difference among groups (group p=0.490).

Gross Observation of Joints: Upon necropsy, joints and incision areas in all

experimental groups appeared normal by gross examination (Fig 3.1), and no

inflammation or infection was noted, indicating no adverse immune reaction to the

Page 59: In vitro and in vivo Growth Factor Delivery to

59

treatments. Repair cartilage appeared to be moderately to normally attached to

surrounding cartilage in all defects with no differences between treatments (treatment

p=0.299) or groups (group p=0.541) (Table 3.4:XIII). Similarly, repair tissue ranged from

normal to slightly soft compared to surrounding tissue when assessed for attachment to

the subchondral bone, with no differences between treatments (treatment p=0.192) or

groups (group p=0.970) (Table 3.4:XIV). Rabbits receiving KLD+CF+BMSCs (group 3)

demonstrated mild osteophyte formation, with an increased score compared to KLD-

alone (group 1) (group p=0.029) (Fig 3.2A). Treated defects in group 3 also had better

attachment to subchondral bone compared to untreated defects in group 3 (p=0.030)

(Table 3.4:XIV). Overall, treated defects had an increased defect volume filled (treatment

p= 0.032) and the level of treated defects was higher than contralateral untreated defects

(treatment p=0.050) (Fig 3.1; Fig 3.2B,C). Untreated defects also had more muscle

wastage at the site of incision (assessed by circumferential measurement at the proximal

aspect of the patella), with group 1 and 3 treated defects different from their respective

controls (treatment p=0.001, interaction p=0.032) (Fig 3.2D). Treated defects were scored

higher in grade of repair compared to contralateral untreated defects, equivalent to a score

of good (3.040±0.219, treated vs. 2.476±0.219, untreated, on a scale of 0-4) (treatment

p=0.061).

Histologic Examinations:

Synovial Membrane H&E

Page 60: In vitro and in vivo Growth Factor Delivery to

60

Evaluation of the synovial membrane by H&E staining showed some mild intimal

hyperplasia and some mild to moderate vascularity and subintimal fibrosis, but there

were no differences among any groups or treatments.

Articular Cartilage H&E

Assessment of the articular cartilage H&E staining showed that KLD (group 1) had a

higher cumulative histology score compared to contralateral untreated controls when

looking at that group alone (p=0.034) (Fig 3.3A). When comparing among groups and

treatments, KLD and KLD+CF (groups 1 and 2) had higher cumulative scores than

KLD+CF+BMSCs (group 3) (group p=0.030) (Fig 3.3A). Defects treated with KLD

received the highest score for nature of predominant tissue in the defect (see Table 3.3:I

and Table 3.5:I), 1.00±0.276, or similar to fibrocartilage. The other groups and treatments

ranged between 0 and 1, indicating presence of some fibrocartilage and some non-

chondrocytic cells; none of the groups or treatments were significantly different, however

(group p=0.749, treatment p=0.264). Group 3 had significantly lower surface regularity

(Table 3.3: II) with scores indicative of some fissuring of the surface (group p=0.005, Fig

3.3B) and more degenerative change (Table 3.3: VIII) in cartilage surrounding the defect

showing mild to moderate hypocellularity (group p=0.015) compared to KLD and

KLD+CF (Fig 3.3C). Group 2 had more reconstitution of subchondral bone than group 3

(group p=0.030), but all groups were still below normal subchondral bone levels (Fig

3.3D; Table 3.3: IX). Group 2 also showed the highest score for bonding to adjacent

cartilage (Table 3.5:V), but no groups or treatments were significantly different

(treatment p=0.161, group p=0.226). Treated defects had increased cellularity (Table

Page 61: In vitro and in vivo Growth Factor Delivery to

61

3.3:VI) compared to contralateral untreated defects (treatment p=0.031) showing normal

to slight hypocellularity in the treated defects repair tissue (1.778±0.110 vs.

1.556±0.110); group 3 trended higher than the other groups (2.00±0.164, group 3;

1.50±0.177, groups 1 and 2) (group p=0.081).

Articular Cartilage SOFG:

Defects treated with KLD-alone (group 1) had increased Safranin-O staining (scores of

slight to moderate staining) than the contralateral untreated defects or treated defects in

groups 2 and 3, which had only none to slight staining (interaction p=0.011) (Fig 3.4A).

There was no overall effect of treatment or group on this measure (treatment p=0.286,

group p=0.604). As shown in Fig 3.4B, treatment with KLD-alone resulted in greater

staining throughout the repair tissue.

Immunohistochemistry Evaluations: Looking at only group 1, defects treated with

KLD showed increased collagen II immunostaining compared to contralateral untreated

defects (p=0.028); although aggrecan immunostaining for defects treated with KLD

received a score of 2.7, it was not different from the contralateral untreated control

(p=0.526) (Fig 3.5A,B). This is in contrast to the difference observed in Safranin-O

staining between defects treated with KLD and the contralateral untreated defects. In

group 2, KLD+CF treatment elicited a tissue with higher aggrecan detected by

immunostaining vs. the contralateral untreated control (p=0.041). In addition, although

defects treated with KLD+CF received the highest collagen II immunostaining score of

all the groups, 3.4, this was not different from its contralateral untreated control

Page 62: In vitro and in vivo Growth Factor Delivery to

62

(p=0.110). Comparing all defects, treated defects had more collagen II (treatment

p=0.001) and increased aggrecan (treatment p=0.056) compared to untreated defects;

there were no differences among groups (group p=0.293). There were similar levels of

collagen I immunostaining found in all the defects (treatment p=0.471, group p=0.919),

consistent with the observation of mostly fibrocartilage seen in the gross scoring (Fig

3.5C).

3.4 Discussion

Treatment of full-thickness articular cartilage defects with the self-assembling peptide

KLD (group 1) markedly improved cartilage regeneration, as seen by significant

increases in cumulative histology score, Safranin-O staining, and collagen II

immunostaining, compared to critically sized contralateral untreated defects. KLD has

several advantages as a material due to its ability to be injected arthroscopically into a

defect, assemble on contact with tissue, and promote cartilage regeneration without

inducing an immune response. Adding TGF-β1, dexamethasone, and IGF-1 to KLD

(group 2) resulted in increased aggrecan immunostaining, but in general did not result in

any additional beneficial or deleterious effects compared to KLD alone. Motivated by in

vitro results supporting BMSC chondrogenesis27-29, we delivered these factors with

allogeneic BMSCs in KLD in vivo (group 3). However, this treatment resulted in a poorer

repair than with KLD or KLD+CF. This is similar to other reports of fibrous tissue

formation after BMSC treatment35, 43, 48, interpreted as a negative result.

Page 63: In vitro and in vivo Growth Factor Delivery to

63

In our rabbit model, the presence of a full-thickness defect allowed bone marrow to

infiltrate the defect41 and act as a cell source, similar to abrasion arthroplasty and

subchondral bone microfracture surgical techniques. The positive healing response seen

with KLD treatment (group 1) demonstrates that the scaffold supports cell migration and

further chondrogenesis of these cells in vivo. The high porosity of this scaffold (~99.6%

water content26) is in agreement with a recent in vivo study in rabbits by Ikeda et al.21

demonstrating that scaffolds with >85% porosity promoted migration of bone marrow

cells into polymer scaffolds and with an in vitro study by Wang et al.45 demonstrating

endothelial cell migration into several types of self-assembling hydrogels.

Regarding group 2, KLD+CF treatment, our results raise the issues of whether the CF

dose was appropriate to improve endogenous cell response and whether cells migrating

into the scaffold in vivo respond differently to these CFs than in vitro and thereby require

different stimulation. The inclusion of CFs did not show increased chondrogenesis over

KLD alone (group 1) as determined by cumulative histology score, Safranin-O, aggrecan,

and type II collagen immunostaining. Similar to these results, Holland et al.17 reported

that TGF-β1 and IGF-1 delivered in gelatin microparticles within an acellular

oligo(poly(ethylene glycol) fumarate) (OPF) scaffold resulted in repair no different than

in empty defects, despite positive results in vitro. That study also delivered IGF-1 alone

and found that this growth factor alone resulted in a significantly improved repair. In

addition, other studies have looked at delivering IGF-1 alone in vivo and have reported

similar positive results36, 40. While we did not test the effects of IGF-1 alone in this study,

Page 64: In vitro and in vivo Growth Factor Delivery to

64

our results emphasize the need for further understanding of the combination of IGF-1

with other CF in vivo in order to take advantage of its characteristic anabolic properties.

The intra-articular use of TGF-β1 remains controversial due to the pro-inflammatory

response by the synovium seen at certain concentrations. Van der Kraan and van den

Berg et al. have studied the interaction of TGF-β1 with various joint tissues and have

shown that while TGF-β1 can stimulate proteoglycan production in cartilage, when it is

exposed to synovial tissue, synovial fibrosis can occur; TGF-β1-induced osteophyte

formation is also common2. Mi et al.32 showed that injection of recombinant adenoviral

vector for hTGF-β1 into the knee joint space through the patellar tendon (resulting in

~8.75 ng TGF-β1) dramatically increased joint inflammation, though this dose is more

than 6-fold higher than our total dose of 1.4 ng. Accordingly, we did not observe these

effects when TGF-β1 was added to the peptide alone (group 2). In contrast, when TGF-

β1 was added in combination with BMSCs (group 3), osteophyte formation increased,

suggesting an interaction of TGF-β1 with these cells was responsible for the increased

osteogenesis rather than the presence of TGF-β1 alone in the joint (group 2). This finding

is similar to results recently published by Guo et al.15, in which 600 ng of TGF-β1 per mL

of hydrogel was used in combination with BMSCs in an OPF scaffold. Furthermore, we

observed a detrimental increase in osteophyte formation and distortion of normal joint

anatomy when, in the same rabbit, 1.4 ng TGF-β1+BMSCs were placed in one joint and

0.7 ng TGF-β1+BMSCs in the contralateral joint33. The authors were surprised at this

result given the total body dose of TGF-β1 was only 1.5x what was used in group 2 or

group 3 and the cell numbers 2-fold greater. While the amount of TGF-β1 delivered in

Page 65: In vitro and in vivo Growth Factor Delivery to

65

group 3 of our study was lower than that in the Mi32 study, and we attempted to deliver

TGF-β1 in a controlled fashion, it is still possible that this TGF-β1 level was too high to

beneficially induce chondrogenesis when combined with BMSCs, despite the inclusion of

dexamethasone, which has been shown to enhance chondrogenesis compared to TGF-β1

alone25, 34. A study by Fan et al. using only 0.8 ng of TGF-β1 with BMSCs in a gelatin-

chondroitin-hyaluronate tri-copolymer scaffold reported improved rabbit cartilage defect

healing compared to treatment with BMSCs implanted in a scaffold without TGF-β1.

Taken together, these studies suggest that while the amount of TGF-β1 we chose

combined with BMSCs effectively induces chondrogenesis in vitro28, additional research

is needed to determine a successful strategy for optimizing chondrogenesis of BMSCs in

vivo, and interactions with synovial tissues must be considered.

Due to the often noted ability of cartilage defects in young/skeletally immature rabbits to

heal well naturally41, we used skeletally mature rabbits with average age of 11 months

and critically sized defects in order to test the ability of treatment to improve adult animal

healing. Although full-thickness defects were used, defects only entered the

subchondral/trabecular bone, resulting in contralateral untreated defects that do not heal

spontaneously by 12 weeks. These results are similar to other studies using similar-aged

rabbits and defect sizes. A limitation of our model is the lack of enough tissue to perform

direct biochemical assessment in addition to histological and immunohistochemical

measurements in this study. However, the purpose of the present study was to perform an

initial trial prior to a larger animal study where such additional measures will be utilized,

since ample tissue will be available.

Page 66: In vitro and in vivo Growth Factor Delivery to

66

In summary, the self-assembling peptide hydrogel KLD offers a new material suitable for

further testing in a clinically relevant defect in a large animal. We demonstrated

improved filling of osteochondral defects and improved cartilage repair, as seen by

cumulative histology score, Safranin-O staining, and type II collagen immunostaining. In

this small animal model, the full-thickness defect provided access to the marrow, similar

in concept to abrasion arthroplasty or spongialization in large animal models (goat, sheep,

horse, human), and suggests that combining KLD with these techniques may offer an

improvement over current practice. Ongoing studies include the evaluation of KLD in a

clinically relevant sized equine defect co-treated with microfracture and subjected to

strenuous exercise, compared to defects treated with microfracture alone.

3.5 Acknowledgements

I would like to thank Alan J. Grodzinsky, Sc.D. and Eric J. Vanderploeg, Ph.D. for their

helpful discussions in planning and analyzing this study. I would like to thank my

collaborators at Colorado State University for their animal model expertise: Christina Lee,

Ph.D., Dora J. Ferris, DVM, Myra F. Barrett, DVM, John D. Kisiday, Ph.D., and David

D. Frisbie, DVM, Ph.D., Diplomate ACVS. Finally, this work would not be possible

without the following funding sources: NIH-NIBIB Grant EB003805; NDSEG and NSF

graduate fellowships.

Page 67: In vitro and in vivo Growth Factor Delivery to

67

3.6 References

1. Anonymous, The GLIMMIX Procedure. 2006, SAS Institute Inc: Cary, NC. p. 943.

2. Davidson ENB, van der Kraan P, and van den Berg W. TGF-β and osteoarthritis. Osteoarthritis and Cartilage. 2007; 15:597-604.

3. Davis ME, Hsieh PCH, Takahashi T, Song Q, Zhang S, Kamm RD, Grodzinsky AJ, Anversa P, and Lee RT. Local myocardial insulin-like growth factor 1 (IGF-1) delivery with biotinylated peptide nanofibers improves cell therapy for myocardial infarction. Proc Natl Acad Sci USA. 2006; 103:8155-8160.

4. Diao H, Wang J, Shen C, Xia S, Guo T, Dong L, Zhang C, Chen J, Zhao J, and Zhang J. Improved cartilage regeneration utilizing mesenchymal stem cells in TGF-β1 gene-activated scaffolds. Tissue Eng Part A. 2009; 15:2687-2698.

5. Elisseeff J, McIntosh W, Fu K, Blunk T, and Langer R. Controlled-release of IGF-1 and TGF-β1 in a photopolymerizing hydrogel for cartilage tissue engineering. J Orthop Res. 2001; 19:1098-1104.

6. Erickson IE, Huang AH, Chung C, Li RT, Burdick JA, and Mauck RL. Differential maturation and structure-function relationships in mesenchymal stem cell- and chondrocyte-seeded hydrogels. Tissue Eng Part A. 2009; 15:1041-52.

7. Fan H, Hu Y, Qin L, Li X, Wu H, and Lv R. Porous gelatin-chondroitin-hyaluronate tri-copolymer scaffold containing microspheres loaded with TGF-β1 induces differentiation of mesenchymal stem cells in vivo for enhancing cartilage repair. J Biomed Mater Res A. 2006; 77:785-794.

8. Florine E, Vanderploeg E, Kopesky P, Miller R, and Grodzinsky AJ. Dexamethasone Suppresses Aggrecan Catabolism in BMSC-seeded Peptide Hydrogels. in 56th Trans Orthop Res Soc. 2010. New Orleans.

9. Fortier LA, Mohammed HO, Lust G, and Nixon AJ. Insulin-like growth factor-I enhances cell-based repair of articular cartilage. J Bone Joint Surg Br. 2002; 84-B:276-288.

10. Frisbie D, Ghivizzani S, Robbins PD, Evans C, and McIlwraith C. Treatment of experimental equine osteoarthritis by in vivo delivery of the equine interleukin-1 receptor antagonist gene. Gene Ther. 2002; 9:12-20.

11. Frisbie D, Bowman S, Colhoun H, DiCarlo E, Kawcak C, and McIlwraith C. Evaluation of autologous chondrocyte transplantation via a collagen membrane in equine articular defects-results at 12 and 18 months. Osteoarthritis and Cartilage. 2008; 16:667-679.

12. Frisbie D, Kisiday J, Kawcak C, Werpy N, and McIlwraith C. Evaluation of adipose-derived stromal vascular fraction or bone marrow-derived mesenchymal stem cells for treatment of osteoarthritis. J Orthop Res. 2009; 27:1675-1680.

13. Getgood A, Brooks R, Fortier L, and Rushton N. Articular cartilage tissue engineering. J Bone Joint Surg Br. 2009; 91-B:565-576.

14. Guo C-A, Liu X-G, Huo J-Z, Jiang C, Wen X-J, and Chen Z-R. Novel gene-modified-tissue engineering of cartilage using stable transforming growth factor-β1-transfected mesenchymal stem cells grown on chitosan scaffolds. J Biosci Bioeng. 2007; 103:547-556.

Page 68: In vitro and in vivo Growth Factor Delivery to

68

15. Guo X, Park H, Young S, Kretlow J, van den Beucken J, Baggett L, Tabata Y, Kasper F, Mikos AG, and Jansen JA. Repair of osteochondral defects with biodegradeable hydrogel composites encapsulating marrow mesenchymal stem cells in a rabbit model. Acta Biomaterialia. 2010; 6:39-47.

16. Hoemann C, Sun J, McKee M, Chevrier A, Rossomacha E, Rivard G-E, Hurtig M, and Buschmann M. Chitosan-glycerol phosphate/blood implants elicit hyaline cartilage repair integrated with porous subchondral bone in microdrilled rabbit defects. Osteoarthritis and Cartilage. 2007; 15:78-89.

17. Holland TA, Bodde EWH, Cuijpers VMJI, Baggett LS, Tabata Y, Mikos AG, and Jansen JA. Degradable hydrogel scaffolds for in vivo delivery of single and dual growth factors in cartilage repair. Osteoarthritis Cartilage. 2007; 15:187-197.

18. Homandberg G, Kang Y, Zhang J, Cole A, and Williams J. A single injection of fibronectin fragments into rabbit knee joints enhances catabolism in the articular cartilage followed by reparative responses but also induces systemic effects in the non-injected knee joints. Osteoarthritis and Cartilage. 2001; 9:673-683.

19. Hunziker E. Growth-factor-induced healing of partial-thickness defects in adult articular cartilage. Osteoarthritis and Cartilage. 2001; 9:22-32.

20. Hwang NSP and Elisseeff JP. Application of Stem Cells for Articular Cartilage Regeneration. J Knee Surg. 2009; 22:60-71.

21. Ikeda R, Fujioka H, Nagura I, Kokubu T, Toyokawa N, Inui A, Makino T, Kaneko H, Doita M, and Kurosaka M. The effect of porosity and mechanical property of a synthetic polymer scaffold on repair of osteochondral defects. Int Orthop. 2009; 33:821-828.

22. Indrawattana N, Chen G, Tadokoro M, Shann LH, Ohgushi H, Tateishi T, Tanaka J, and Bunyaratvej A. Growth factor combination for chondrogenic induction from human msc. Biochem Biophys Res Commun. 2004; 320:914-919.

23. Jaklenec A, Hinckfuss A, Bilgen B, Ciombor DM, Aaron R, and Mathiowitz E. Sequential release of bioactive IGF-I and TGF-β1 from PLGA microsphere-based scaffolds. Biomaterials. 2008; 29:1518-1525.

24. Janjanin S, Li W-J, Morgan MT, Shanti RM, and Tuan RS. Mold-Shaped, Nanofiber Scaffold-Based Cartilage Engineering Using Human Mesenchymal Stem Cells and Bioreactor. J Surg Res. 2008; 149:47-56.

25. Johnstone B, Hering TM, Caplan AI, Goldberg VM, and Yoo JU. In Vitro Chondrogenesis of Bone Marrow-Derived Mesenchymal Progenitor Cells. Experimental Cell Research. 1998; 238:265-272.

26. Kisiday J, Jin M, Kurz B, Hung H, Semino C, Zhang S, and Grodzinsky AJ. Self-assembling peptide hydrogel fosters chondrocyte extracellular matrix production and cell division: Implications for cartilage tissue repair. Proc Natl Acad Sci USA. 2002; 99:9996-10001.

27. Kisiday J, Kopesky P, Evans C, Grodzinsky AJ, McIlwraith C, and Frisbie D. Evaluation of adult equine bone marrow- and adipose-derived progenitor cell chondrogenesis in hydrogel cultures. J Orthop Res. 2008; 26:322-331.

28. Kopesky P, Vanderploeg E, Kisiday J, Frisbie D, Sandy J, and Grodzinsky AJ. A Single-Dose of TGF-β Induces Chondrogenesis in MSC-seeded Peptide and Agarose Hydrogels. in 55th Trans Orthop Res Soc. 2009. Las Vegas.

Page 69: In vitro and in vivo Growth Factor Delivery to

69

29. Kopesky P, Vanderploeg E, Sandy J, Kurz B, and Grodzinsky AJ. Self-assembling peptide hydrogels modulate in vitro chondrogenesis of bovine bone marrow stromal cells. Tissue Eng Part A. 2009; In press.

30. Li W, Chiang H, Kuo T, Lee H, Jiang C, and Tuan R. Evaluation of articular cartilage repair using biodegradable nanofibrous scaffolds in a swine model. J Tissue Eng Regen Med. 2009; 3:1-10.

31. Longobardi L, O'Rear L, Aakula S, Johnstone B, Shimer K, Chytil A, Horton WA, Moses HL, and Spagnoli A. Effect of IGF-I in the Chondrogenesis of Bone Marrow Mesenchymal Stem Cells in the Presence or Absence of TGF-β1 Signaling. J Bone Miner Res. 2006; 21:626-636.

32. Mi Z, Ghivizzani S, Lechman E, Glorioso J, Evans C, and Robbins P. Adverse effects of adenovirus-mediated gene transfer of human TGF-β1 into rabbit knees. Arthritis Res Ther. 2003; 5:R132 - R139.

33. Miller R, Grodzinsky AJ, Vanderploeg E, Lee C, Ferris DJ, Barrett MF, Kisiday J, and Frisbie D. Repair of Full-Thickness Articular Cartilage Defect Using Self-Assembling Peptide, Growth Factors, and BMSCs. in 56th Trans Orthop Res Soc. 2010. New Orleans.

34. Mouw JK, Connelly JT, Wilson CG, Michael KE, and Levenston ME. Dynamic Compression Regulates the Expression and Synthesis of Chondrocyte-Specific Matrix Molecules in Bone Marrow Stromal Cells. Stem Cells. 2007; 25:655-663.

35. Murphy J, Fink D, Hunziker E, and Barry F. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003; 48:3464-3474.

36. Nixon A, Fortier L, Williams J, and Mohammed H. Enhanced repair of extensive articular defects by insulin-like growth factor-I-laden fibrin composites. J Orthop Res. 1999; 17:475-487.

37. Noth U, Steinert AF, and Tuan RS. Technology Insight: adult mesenchymal stem cells for osteoarthritis therapy. Nat Rev Rheumatol. 2008; 4:371-380.

38. Park K-H, Park W, and Na K. Synthetic matrix containing glucocorticoid and growth factor for chondrogenic differentiation of stem cells. J Biosci Bioeng. 2009; 108:168-173.

39. Rutgers M, van Pelt M, Dhert W, Creemers L, and Saris D. Evaluation of histological scoring systems for tissue-engineered, repaired, and osteoarthritic cartilage. Osteoarthritis and Cartilage. 2010; 18:12-23.

40. Schmidt M, Chen E, and Lynch S. A review of the effects of insulin-like growth factor and platelet-derived growth factor on in vivo cartilage healing and repair. Osteoarthritis and Cartilage. 2006; 14:403-412.

41. Shapiro F, Koide S, and Glimcher M. Cell origin and differentiation in the repair of full-thickness defects of articular cartilage. J Bone Joint Surg Am. 1993; 75:532-553.

42. Sieminski AL, Semino CE, Gong H, and Kamm RD. Primary sequence of ionic self-assembling peptide gels affects endothelial cell adhesion and capillary morphogenesis. J Biomed Mater Res A. 2008; 87:494-504.

43. Wakitani S, Mitsuoka T, Nakamura N, Toritsuka Y, Nakamura Y, and Horibe S. Autologous bone marrow stromal cell transplantation for repair of full-thickness articular cartilage defects in human patellae: two case reports. Cell Transplant. 2004; 13:595 - 600.

Page 70: In vitro and in vivo Growth Factor Delivery to

70

44. Wang D-A, Varghese S, Sharma B, Strehin I, Fermanian S, Gorham J, Fairbrother D, Cascio B, and Elisseeff J. Multifunctional chondroitin sulphate for cartilage tissue-biomaterial integration. Nat Mater. 2007; 6:385-392.

45. Wang X, Horii A, and Zhang S. Designer functionalized self-assembling peptide nanofiber scaffolds for growth, migration, and tubulogenesis of human umbilical vein endothelial cells. Soft Matter. 2008; 4:2388-2395.

46. Wegener B, Schrimpf F, Pietschmann M, Milz S, Berger-Lohr M, Bergschmidt P, Jansson V, and Muller P. Matrix-guided cartilage regeneration in chondral defects. Biotechnol Appl Biochem. 2009; 53:63-70.

47. Wei X and Messner K. Age- and injury-dependent concentrations of transforming growth factor-β1 and proteoglycan fragments in rabbit knee joint fluid. Osteoarthritis and Cartilage. 1998; 6:10-18.

48. Wilke M, Nydam D, and Nixon A. Enhanced early chondrogenesis in articular defects following arthroscopic mesenchymal stem cell implantation in an equine model. J Orthop Res. 2007; 25:913-925.

49. Worster AA, Brower-Toland BD, Fortier LA, Bent SJ, Williams J, and Nixon AJ. Chondrocytic differentiation of mesenchymal stem cells sequentially exposed to transforming growth factor-b1 in monolayer and insulin-like growth factor-I in a three-dimensional matrix. J Orthop Res. 2001; 19:738-749.

50. Zhang S, Holmes T, Lockshin C, and Rich A. Spontaneous Assembly of a Self-Complementary Oligopeptide to Form a Stable Macroscopic Membrane. Proc Natl Acad Sci USA. 1993; 90:3334-3338.

Page 71: In vitro and in vivo Growth Factor Delivery to

71

3.7 Tables

Table 3.1. Treatment groups with amounts delivered per animal.

Group n KLD (µg)

bKLD (µg)

TGF-β1 (ng)

bIGF (ng)

Streptavidin (ng)

Dex (ng)

BMSCs (x103)

1 (KLD) 6 48 0.48 0 0 0 0 0 2 (KLD+CF) 6 48 0.48 1.4 4.1 30.7 0.6 0

3 (KLD+CF+BMSCs) 7 48 0.48 1.4 4.1 30.7 0.6 150

Page 72: In vitro and in vivo Growth Factor Delivery to

72

Table 3.2. Gross Observations Scoring System.

Gross Observations Scoring System Feature Score I. Joint Observation Normal appearance 0 Slight inflammation 1 Moderate inflammation 2 Severe inflammation 3 II. Incision Appearance Normal appearance 0 Slight inflammation 1 Moderate inflammation 2 Severe inflammation 3 Slight dehiscence (incision basically intact) 4 Marked dehiscence (requires intervention) 5 Active infection 6 Healing infections 7 III. Muscle wastage Circumferential measurement at the proximal aspect of the patella (cm) IV. Angle of Stifle: Change in normal angle (if any) None, within normal limits 0 Abnormal 1 V. Inflammation/Swelling None, within normal limits 0 Abnormal 1 VI. Trauma/Damage None, within normal limits 0 Abnormal 1 VII. Infection/Discharge None, within normal limits 0 Abnormal 1 VIII. Presence of Osteophytes and other Osteoarthritic Changes None, within normal limits 0 Abnormal 1 IX. Articular Surface Integrity, Contour, and Congruity Normal 0 Abnormal 1 X. Presence of Loose Bodies in Synovial Fluid None, within normal limits 0 Abnormal 1 XI. Injury to Apposing Articular Surface None, within normal limits 0 Abnormal 1 XII. Synovial Membrane Appearance Normal 0 Slight 1 Mild 2 Moderate 3

Page 73: In vitro and in vivo Growth Factor Delivery to

73

Severe 4 XIII. Cartilage Attachment This category describes on average the defect repair tissue attachment with the surrounding normal cartilage. Possible responses were: Normal Attachment 0 Moderate Attachment 1 Mild Attachment 2 Slight Attachment 3 No Attachment 4 XIV. Bone Attachment This category describes the firmness of the repair tissue attachment to the bone at the base of the defect. Possible responses were: Similar to Surrounding Cartilage 0 Slightly Soft-vs-Surrounding Cartilage 1 Mildly Soft-vs-Surrounding Cartilage 2 Moderately Soft-vs-Surrounding Cartilage 3 Marked Softening-vs-Surrounding Cartilage 4 XV. Firmness This category describes on average the firmness of the repair tissue to normal surrounding articular cartilage. Possible responses were: Similar Compared to Surrounding Cartilage 0 Slightly Soft Compared to Surrounding Cartilage 1 Mildly Soft Compared to Surrounding Cartilage 2 Moderately Soft Compared to Surrounding Cartilage 3 Marked Softening Compared to Surrounding Cartilage 4 XVI. Blood This category describes the presence or absence of hemorrhage associated with the defect area or its periphery. Possible responses were: Fresh Blood, Active Hemorrage at Time of Surgery 1 Old Blood, No Active Hemorrage at Time of Surgery 2 No Blood Visualized at Time of Surgery 3 XVII. Shape This category describes the margin of the defect as it relates to the original geometry at time 0 (creation). Possible responses were: No Apparent Change in Damage Tissue Beyond Defect 0 Degeneration in Tissue Beyond Defect 1 XVIII. Grade (Overall quality of repair) This category describes the overall subjective evaluation of the repair tissue by the evaluator. Criteria used to determine the grade were: (1) Attachment of repair tissue to the surrounding normal articular cartilage (2) Level (height) and undulation of the repair tissue surface as compared to the surrounding normal articular cartilage (3) Color of the repair tissue, where white homogenous tissue without a fibrous like appearance is used as the “gold standard.” Possible responses were: Tissue Not Present to Grade 0 Poor 1 Fair 2

Page 74: In vitro and in vivo Growth Factor Delivery to

74

Good 3 Excellent 4 XIX. Level This category describes the level of repair tissue filling in association with the surrounding normal articular cartilage. Possible responses in relation to the surrounding normal articular cartilage were: Mildly Recessed 1 Slightly Recessed 2 Leveled 3 Slightly Elevated 4 Mildly Elevated 5 Moderately Elevated 6 XX. Color This category describes the color of the repair tissue. When repair tissue is characterized by two colors, the predominate color is indicated first. Possible responses were: Red 1 White/Red 2 Yellow 3 Yellow/White 4 White/Yellow 5 White 6 XXI. Surface This category describes the relative undulation of the repair tissue surface. Possible responses were: Non-Undulating 1 Slightly Undulating 2 Mildly Undulating 3 Moderatley Undulating 4 XXII. Area Percent of Surface Area Filled in Defect (0-100%) XXIII. Volume Percent of Volume Filled in Defect (0-100%)

Page 75: In vitro and in vivo Growth Factor Delivery to

75

Table 3.3. Modified O’Driscoll Histological Scoring System

Feature Score I. Nature of the Repair Tissue Some Fibrocartilage, mostly nonchondrocytic cells 0 Mostly Fibrocartilage 1 Mixed Hyaline and Fibrocartilage 2 Mostly Hyaline Cartilage 3 Hyaline Cartilage 4 II. Surface Regularity Severe Disruption, Including Fibrillation 0 Fissures 1 Superficial Horizontal Lamination 2 Smooth and Intact 3 III. Structural Integrity (morphologic zone reconstitution) Severe Disintegration 0 Slight Disruption, Including Cysts 1 Normal 2 IV. Thickness 0% - 50% of Normal Cartilage 0 50% - 100% of Normal Cartilage 1 100% of Normal Adjacent Cartilage 2 V. Bonding to Adjacent Cartilage Not Bonded 0 Bonded at One End or Partially at Both Ends 1 Bonded at Both Ends of Graft 2 VI. Hypocellularity Moderate Hypocellularity 0 Slight Hypocellularity 1 Normal Cellularity 2 VII. Chondrocyte Clustering 25% - 100% of the Cells 0 < 25% of the Cells 1 No Clusters 2 VIII. Freedom from Degenerative Changes in Adjacent Cartilage Severe Hypocellularity, Poor or No Staining 0 Mild or Moderate Hypocellularity, Slight Staining 1 Normal Cellularity, Mild Clusters, Moderate Staining 2 Normal Cellularity, No Clusters, Normal Staining 3 IX. Reconstitution of Subchondral Bone No Subchondral Bone Reconstitution 0 Minimal Subchondral Bone Reconstitution 1 Reduced Subchondral Bone Reconstitution 2 Normal 3 X. Inflammatory Response in Subchondral Bone Region Severe 0 Moderate 1 None / Mild 2 XI. Safranin-O Staining None 0

Page 76: In vitro and in vivo Growth Factor Delivery to

76

Slight 1 Moderate 2 Normal 3 XII. Cumulative Histology Score (sum of above scores) 0-28

Page 77: In vitro and in vivo Growth Factor Delivery to

77

Table 3.4. Gross Scores. One-way p-values for within group comparisons are listed in the untreated columns. Two-way p-values for differences among groups are in a separate column (TXT = treatment, Group = group, TXT*Group = interaction term). P-values are not shown when all the scores being compared are zero. Gross Scores KLD KLD+CF KLD+CF+BMSCs Feature Treated untreated treated untreated treated untreated

Two-way P-values

I. Joint Observation 0±0 0±0 0±0 0±0 0±0 0±0 II. Incision Appearance 0±0 0±0 0±0 0±0 0±0 0±0

III. Muscle wastage 12.8± 0.3

12.4±0.3, p=0.010

12.8± 0.3

12.7±0.3, p=0.530

13.0± 0.3

12.9±0.3, p=0.049

TXT: 0.001 Group: 0.553

TXT*Group: 0.032

IV. Angle of Stifle: Change in normal angle (if any)

0±0 0±0 0±0 0±0 0±0 0±0

V. Inflammation/ Swelling 0±0 0±0 0±0 0±0 0±0 0±0

VI. Trauma/Damage 0±0 0±0 0±0 0±0 0±0 0±0 VII. Infection/Discharge 0±0 0±0 0±0 0±0 0±0 0±0

VIII. Osteophytes and other Osteoarthritic Changes

0±0.2 0±0.2 0.2±0.2 0.2±0.2, p=1.000

0.6±0.2 0.6±0.2, p=1.000

TXT: 1.000 Group: 0.029 TXT*Group:

1.000 IX. Articular Surface Integrity, Contour, and Congruity

0±0 0±0 0±0 0±0 0±0 0±0

X. Presence of Loose Bodies in Synovial Fluid 0±0 0±0 0±0 0±0 0±0 0±0

XI. Injury to Apposing Articular Surface 0±0 0±0 0±0 0±0 0±0 0±0

XII. Synovial Membrane 0.5±0.2 0.3±0.2,

p=0.611 0.2±0.2 0.3±0.2,

p=0.562 0.1±0.2 0.3±0.2,

p=0.356

TXT: 0.750 Group: 0.685 TXT*Group:

0.605

XIII. Cartilage Attachment to surrounding cartilage

0.7±0.3 1.0±0.3, p=0.530

0.5±0.3 0.3±0.3, p=0.611

0.4±0.3 1.0±0.3, p=0.172

TXT: 0.299 Group: 0.541 TXT*Group:

0.423

XIV. Bone Attachment 0.3±0.3 0.7±0.3, p=0.363

0.7±0.3 0.5±0.3, p=0.695

0.3±0.3 0.9±0.3, p=0.030

TXT: 0.192 Group: 0.970 TXT*Group:

0.262

XV. Firmness 0.7±0.4 0.8±0.4, p=0.741

1.0±0.4 0.5±0.4, p=0.296

0.4±0.4 1.0±0.4, p=0.172

TXT: 0.750 Group: 0.995 TXT*Group:

0.222

XVI. Blood 3.0±0.1 3.0±0.1, p=1.000

3.0±0.1 3.0±0.1, p=1.000

3.0±0.1 2.9±0.1, p=0.356

TXT: 0.384 Group: 0.450 TXT*Group:

0.450 XVII. Shape 0±0 0±0 0±0 0±0 0±0 0±0

XVIII. Grade (Overall quality of repair) 3.2±0.4 2.2±0.4,

p=0.076 2.7±0.4 2.8±0.4,

p=0.818 3.3±0.4 2.4±0.4,

p=0.017

TXT: 0.061 Group: 0.896 TXT*Group:

0.220

XIX. Level 2.7±0.3 2.0±0.3, p=0.102

2.5±0.3 2.3±0.3, p=0.695

2.7±0.3 2.3±0.3, p=0.200

TXT: 0.050 Group: 0.897 TXT*Group:

0.613

XX. Color 5.8±0.5 4.7±0.5, p=0.126

4.8±0.5 4.8±0.5, p=1.000

5.9±0.5 5.3±0.5, p=0.231

TXT: 0.182 Group: 0.363 TXT*Group:

0.547

Page 78: In vitro and in vivo Growth Factor Delivery to

78

XXI. Surface 1.8±0.3 2.2±0.3, p=0.465

1.5±0.3 2.0±0.3, p=0.296

1.7±0.3 2.0±0.3, p=0.457

TXT: 0.127 Group: 0.753 TXT*Group:

0.925

XXII. Area 94.2±6.6 90.0±6.6, p=0.383

95.7±6.6 90.0±6.6, p=0.441

97.1±6.1 82.9±6.1, p=0.276

TXT: 0.138 Group: 0.902

TXT*Group: 0.681

XXIII. Volume 91.7±7.6 71.7±7.6, p=0.058

88.2±7.6 80.8±7.6, p=0.477

94.3±7.0 82.1±7.0, p=0.297

TXT: 0.032 Group: 0.706

TXT*Group: 0.667

Page 79: In vitro and in vivo Growth Factor Delivery to

79

Table 3.5. Histological Scores. One-way p-values for within group comparisons are listed in the untreated columns. Two-way p-values for differences among groups are in a separate column. Histology Scores KLD KLD+CF KLD+CF+BMSCs Feature treated untreated treated untreated treated untreated

Two-way p-values

I. Nature of the Repair Tissue 1.0±0.3 0.7±0.3,

p=0.363 0.7±0.3 0.7±0.3,

p=1.000 0.9±0.3 0.4±0.3,

p=0.134

TXT: 0.264 Group: 0.749

TXT*Group: 0.710

II. Surface Regularity 2.5±0.3 2.2±0.3, p=0.175

2.0±0.3 1.7±0.3, p=0.465

1.4±0.3 0.9±0.3, p=0.298

TXT: 0.130 Group: 0.005 TXT*Group:

0.906

III. Structural Integrity 1.3±0.2 1.0±0.2 1.2±0.2 1.2±0.2,

p=1.000 0.9±0.2 0.7±0.2,

p=0.563

TXT: 0.284 Group: 0.118

TXT*Group: 0.655

IV. Thickness 1.7±0.2 1.2±0.2, p=0.237

1.5±0.2 1.7±0.2, p=0.611

1.3±.2 1.3±0.2, p=1.000

TXT: 0.583 Group: 0.478 TXT*Group:

0.396

V. Bonding to Adjacent Cartilage 0.7±0.3 1.0±0.3,

p=0.363 1.3±0.3 1.3±0.3,

p=1.000 0.6±0.3 1.0±0.3,

p=0.200

TXT: 0.161 Group: 0.226

TXT*Group: 0.580

VI. Hypocellularity 1.7±0.2 1.3±0.2, p=0.175

1.7±0.2 1.3±0.2, p=0.175

2.0±0.2 2.0±0.2, p=1.000

TXT: 0.031 Group: 0.081 TXT*Group:

0.259

VII. Chondrocyte Clustering 1.7±0.2 1.8±0.2,

p=0.363 1.7±0.2 1.7±0.2,

p=1.000 1.9±0.2 2.0±0.2,

p=0.356

TXT: 0.364 Group: 0.397

TXT*Group: 0.809

VIII. Freedom from Degenerative Changes in Adjacent Cartilage

1.7±0.2 1.8±0.2, p=0.611

1.8±0.2 2.2±0.2, p=0.363

1.3±0.2 1.0±0.2, p=0.172

TXT: 0.658 Group: 0.015

TXT*Group: 0.270

IX. Reconstitution of Subchondral Bone 1.8±0.3 1.5±0.3,

p=0.363 2.2±0.3 2.0±0.3,

p=0.695 1.6±0.3 0.9±0.3,

p=0.074

TXT: 0.074 Group: 0.030

TXT*Group: 0.556

X. Inflammatory Response in Subchondral Bone Region

2.0±0.1 1.8±0.1, p=0.363

2.0±0.1 2.0±0.1, p=1.000

2.0±0.1 2.0±0.1, p=1.000

TXT: 0.306 Group: 0.358

TXT*Group: 0.358

XI. Safranin-O Staining 1.2±0.2 0.2±0.2,

p=0.012 0.3±0.2 0.5±0.2,

p=0.611 0.3±0.2 0.6±0.2,

p=0.337

TXT: 0.286 Group: 0.604

TXT*Group: 0.011

XII. Cumulative Histology Score (sum of above scores)

17.2±1.1 14.5±1.1, p=0.034

16.3±1.1 16.2±1.1, p=0.915

14.0±1.0 12.7±1.0, p=0.417

TXT: 0.140 Group: 0.030

TXT*Group: 0.541

Page 80: In vitro and in vivo Growth Factor Delivery to

80

3.8 Figures

Figure 3.1. Gross necropsy photographs of treated and untreated joints in KLD, KLD+CF, and KLD+CF+BMSCs.

Page 81: In vitro and in vivo Growth Factor Delivery to

81

Figure 3.2. Gross pathologic observation of joints comparing KLD, KLD+CF, and KLD+CF+BMSCs treated and contralateral untreated defects. ; * p<0.05, † p=0.0292. (A) Gross osteophytes (scored as 0-1, where 0 = normal, 1 = abnormal); (B) Volume % Filled (scored as 0-100%); (C) Level of treated defects (scored as 1-6, where <3 is recessed, 3 is level with surrounding cartilage, and >3 is elevated); (D) Muscle wastage (circumference proximal to patella).

Page 82: In vitro and in vivo Growth Factor Delivery to

82

Figure 3.3. Histological scores comparing KLD, KLD+CF, and KLD+CF+BMSCs treated and untreated defects. Higher scores indicate more similar to native cartilage. (A) Cumulative histology (scored as 0-28); (B) Surface regularity (scored as 0-3); (C) Freedom from degenerative change of cartilage surrounding defect (scored as 0-3); (D) Reconstitution of subchondral bone (scored as 0-3); * p<0.05.

Page 83: In vitro and in vivo Growth Factor Delivery to

83

Figure 3.4. Safranin-O staining. (A) Scored as 0-3; * p<0.05. (B) Images showing histological evaluation of representative treatment groups and representative contralateral control knees. Treatment and contralateral pictures were representative of the mean scores and were taken from different animals for KLD and KLD+CF+BMSCs, and from the same animal for KLD+CF. Scale bar = 1 mm.

Page 84: In vitro and in vivo Growth Factor Delivery to

84

Figure 3.5. Immunohistochemistry scores (0-4). A) Aggrecan. B) Collagen II. C) Collagen I. * p<0.05.

Page 85: In vitro and in vivo Growth Factor Delivery to

85

Chapter 4. Intra-articular Injection of HB-IGF-1 Sustains Delivery of IGF-1 to Cartilage through Binding to Chondroitin Sulfate

IGF-1 stimulates cartilage repair but is not a practical therapy due to its short half-life.

We have previously modified IGF-1 by adding a heparin-binding domain and have

shown that this fusion protein (HB-IGF-1) stimulates sustained proteoglycan synthesis in

cartilage. Here, we first examined the mechanism by which HB-IGF-1 is retained in

cartilage. We then tested whether HB-IGF-1 provides sustained growth factor delivery to

cartilage in vivo and to human cartilage explants. Retention of HB-IGF-1 and IGF-1 was

analyzed by Western blotting. The requirement of heparan sulfate (HS) or chondroitin

sulfate (CS) glycosaminoglycans for binding was tested using enzymatic removal and

cells with genetic deficiency of HS. Binding affinities of HB-IGF-1 and IGF-1 proteins

for isolated glycosaminoglycans were examined by surface plasmon resonance and

ELISA. In cartilage explants, chondroitinase treatment decreased binding of HB-IGF-1,

whereas heparitinase had no effect. Furthermore, HS was not necessary for HB-IGF-1

retention on cell monolayers. Binding assays showed that HB-IGF-1 bound both CS and

HS, whereas IGF-1 did not bind either. After intra-articular injection in rat knees, HB-

IGF-1 was retained in articular and meniscal cartilages, but not in tendon, consistent with

enhanced delivery to CS-rich cartilage. Finally, HB-IGF-1 but not IGF-1 was retained in

human cartilage explants. After intra-articular injection in rats, HB-IGF-1 is specifically

retained in cartilage through its high abundance of CS. Modification of growth factors

with heparin-binding domains may be a new strategy for sustained and specific local

delivery to cartilage.

Page 86: In vitro and in vivo Growth Factor Delivery to

86

4.1 Introduction

Insulin-like growth factor-I (IGF-1) is known to be an important anabolic factor in

cartilage homeostasis14. IGF-1 not only promotes synthesis of aggrecan, link protein, and

hyaluronan3, 6, 34, it also inhibits proteoglycan degradation19, 29, 47. IGF-1 is primarily

produced by the liver and reaches cartilage through the synovial fluid33, 42, 43, acting on

chondrocytes through both autocrine and paracrine mechanisms27, 36. In multiple animal

models of cartilage injury, viral delivery of IGF-1 has been successfully used to enhance

cartilage repair15, 30.

While IGF-1 may therefore be a potential therapeutic for cartilage repair, a clinically

useful technique for non-viral IGF-1 delivery to cartilage has yet to be developed. A

successful IGF-1 delivery strategy must overcome two major obstacles. First, IGF-1 has a

short half-life of 8-16 hours in vivo when delivered systemically23. Second, systemic

delivery of IGF-1 must be minimized since long-term excess circulating IGF-1 has been

linked to increased risk for cancer5 and high-dose systemic IGF-1 administration causes

significant adverse events21. Studies delivering IGF-1 directly to the joint through fibrin

constructs11, 20, 37 have been promising, but rapid clearance of IGF-1 from the joint has

prevented intra-articular injections of IGF-1 without a carrier from being effective42, and

has been a limiting factor in delivery methods proposed to date.

We have focused on the family of heparin-binding growth factors as a model for

sequestration and sustained local delivery of growth factors to cartilage. Basic fibroblast

growth factor (bFGF or FGF-2), vascular endothelial growth factor (VEGF), heparin-

Page 87: In vitro and in vivo Growth Factor Delivery to

87

binding epidermal growth factor-like growth factor (HB-EGF), pleiotrophin, midkine,

and platelet-derived growth factor (PDGF) are all members of the heparin-binding

growth factor family and have been extensively studied for their ability to be retained in

the extracellular matrix (ECM) of various tissues through their highly positively charged

heparin-binding domains9, 22. Heparin-binding domains may be particularly relevant for

localizing growth factors in cartilage. In particular, FGF-2 has been shown to bind to

isolated highly negatively charged small leucine rich proteoglycan fibromodulin18 and to

the heparan sulfate proteoglycan perlecan17, 50 in cartilage. Binding to ECM maintains a

reservoir of FGF-2 that is released from the tissue upon cartilage injury or degradation18,

48, 49, and binding to perlecan has been shown to protect FGF-2 from proteolytic

degradation10, 41.

Motivated by these considerations, we have designed a new strategy for local delivery of

IGF-1 in various tissues: we added the heparin-binding domain of HB-EGF to the

amino-terminus of IGF-1 to create a new heparin-binding IGF-1 fusion protein, HB-IGF-

146. We have previously shown that HB-IGF-1 produces long-term delivery of

bioavailable IGF-1 to bovine cartilage explants and a single dose stimulates a sustained

increase in proteoglycan synthesis compared to IGF-1. However, the mechanism by

which HB-IGF-1 is retained in tissues is not yet clear. Heparin-binding domains are all

highly positively charged but the rigidity of their secondary structure varies, leading to

different specificities for binding to heparan sulfate as opposed to other negatively

charged sulfated glycosaminoglycans4, 45. Cartilage extracellular matrix (ECM) contains

Page 88: In vitro and in vivo Growth Factor Delivery to

88

primarily chondroitin sulfate (CS), while the pericellular matrix is rich in heparan sulfate

(HS)9, 50.

We hypothesized that HB-IGF-1 is retained in cartilage by binding heparan sulfate

proteoglycans in the matrix and at the cell surface. In the present study, we tested this

hypothesis by measuring release of bound HB-IGF-1 following chondroitinase or

heparitinase treatment of cartilage explants, binding of HB-IGF-1 to cells unable to

produce heparan sulfate, and the binding affinities of HB-IGF-1 for isolated heparan

sulfate and chondroitin sulfate. Surprisingly, we found that HB-IGF-1 was retained

primarily by binding to chondroitin sulfate, whereas heparan sulfate was not required.

This result led us to test whether intra-articular injection of HB-IGF-1 allows sustained in

vivo delivery preferentially to CS-rich rat knee cartilage and whether HB-IGF-1 can bind

adult human cartilage.

4.2 Materials and Methods

Protein Production: HB-IGF-1 and IGF-1 were expressed in E. coli as Xpress and

hexahistidine tagged proteins and were purified by Ni-NTA affinity followed by reverse-

phase chromatography, as previously described in detail46.

Binding in Bovine Cartilage with GAG-ase Treatments: Cartilage disks (3 mm diameter,

0.5 mm thick) from calf femoropatellar grooves were cultured in serum-free low glucose-

DMEM with 500 nM HB-IGF-1 or IGF-1 for 2 days. At Day 2, disks were washed with

PBS and treated with either no enzyme, chondroitinase ABC (E.C. 4.2.2.4, Associates of

Page 89: In vitro and in vivo Growth Factor Delivery to

89

Cape Cod, Inc., East Falmouth, MA) (0.4 U/mL), or heparitinase (4:1 mixture of

heparitinase I and II, E.C. 4.2.2.8, Associates of Cape Cod, Inc.) (0.036 U/mL). These

enzymes have been shown to be specific for chondroitin sulfate38 and heparan sulfate44,

respectively. At Day 4, half of the chondroitinase-treated disks were treated with

heparitinase (0.036 U/mL); all other disks were incubated in enzyme-free medium (n=4).

On Day 6, disks were flash frozen and protein was extracted by pulverization and by

incubation with 100 mM NaCl, 50 mM Tris, 0.5% TritonX-100, pH 7.0 with protease

inhibitor cocktail (Roche, Basel, Switzerland) rotating at 4 ˚C overnight. Protein was

quantified by BCA assay (Thermo Fisher Scientific Inc., Rockford, IL) and equal

amounts of protein were loaded on a 4-12% SDS-PAGE gel and analyzed by Western

blot with anti-IGF-1 (1:500, Abcam Inc., Cambridge, MA, recognizes both HB-IGF-1

and IGF-146). 5 ng of recombinant HB-IGF-1 was loaded as a protein standard and equal

protein of an explant incubated with 500 nM IGF-1 for 2 days without enzyme treatment

or washing was loaded as a positive control. HB-IGF-1 released to the medium was

analyzed by ELISA as previously described46. Heparitinase activity was confirmed by

assaying conditioned medium using an anti-HS-stub antibody 3G10 (1:500, Associates of

Cape Cod, Inc.) by Western blot. This antibody has been shown previously to only detect

neoepitopes generated by heparitinase cleavage and not by chondroitinase7.

Chondroitinase activity was confirmed by assaying for GAG loss in treated explants

using the DMMB dye binding assay, which showed that >75% sGAG was removed after

48 h.

Page 90: In vitro and in vivo Growth Factor Delivery to

90

Binding to Chinese hamster ovary (CHO) cell surfaces: Mutant CHO cells unable to

produce heparan sulfate (strain pgsD-677)26 and wildtype CHO (K1) cells were cultured

in F12 medium supplemented with 10% FBS. At confluence, cells were washed with

PBS and incubated in serum-free medium with 100 nM HB-IGF-1 or IGF-1. After 3

hours, cells were washed 3x10 min with PBS and lysed with 50 mM Tris/HCl, 150 mM

NaCl, 10 mM EDTA, 1% Triton X-100, 1% Igepal CA-630 (Sigma-Aldrich, St. Louis,

MO), 2 mM sodium orthovanadate, 1 mM phenylmethylsulfonyl fluoride, and protease

inhibitor cocktail. Protein was quantified by BCA assay, and equal amounts of protein

were analyzed by Western blot using anti-IGF-1 as described above. 5 ng of recombinant

IGF-1 was loaded as a control.

Biotinylation of Glycosaminoglycans (GAGs): Heparan sulfate (HS) (bovine kidney,

Sigma, #H7640, 0.88 sulfates/disaccharide25) and chondroitin sulfate C (CS) (CS-C from

shark cartilage, Sigma, #C4384, 0.99 sulfates/disaccharide25) (0.5 mg) were biotinylated

mid-chain with Ez-Link-biotin hydrazide (Thermo Fisher Scientific Inc.) as previously

described12 and purified following manufacturer instructions. Biotinylation was

confirmed by dot blot using anti-biotin (1:500, Cell Signaling Technology, Danvers, MA).

Binding Analysis via Surface Plasmon Resonance: All binding experiments were

performed at room temperature at a flow rate of 20 µL/min on a Biacore2000 system (GE

Healthcare, Buckinghamshire, UK). Biotinylated HS and CS were immobilized on

separate flow cells of a streptavidin-coated Biacore chip (GE Healthcare, Piscataway, NJ)

and coated with ~600 RU. Another flow cell was left untreated as a control. HB-IGF-1 or

Page 91: In vitro and in vivo Growth Factor Delivery to

91

IGF-1 was injected in running buffer consisting of 0.01M HEPES, 0.15M NaCl, 3mM

EDTA, 0.005% Tween20, pH 7.4. KinInject was used to inject each IGF-1 over the chip

with association and dissociation times of 5 min. The surface was regenerated by flowing

1 M NaCl over the chip between experiments. Three to four concentrations of each IGF-1,

with three repeats at each concentration, were performed for kinetic analyses. Control

flow cell curves were subtracted from all binding curves in order to account for non-

specific binding and refractive index change. Association and dissociation rate constants

(ka and kd respectively) were determined by fitting the measured binding curves globally

with a 1:1 binding model using BIAevaluation software v4.1 and floating Rmax as a local

parameter40. The equilibrium dissociation constant (KD) was calculated as kd/ka.

ELISA Analysis of Binding to Biotinylated GAGs: Coating, blocking and washing buffers,

secondary antibody, substrate, and stop solutions were purchased from KPL, Inc.

(Gaithersburg, MD). Streptavidin-coated microplates (R&D Systems, Inc., Minneapolis,

MN) were coated with biotinylated HS and CS at 20 µg/mL overnight at 4 ˚C. Plates

were blocked for 15 min at room temperature before incubation with 0-500 nM HB-IGF-

1 or IGF-1 for one hour. Plates were washed three times and incubated with rabbit anti-

IGF-1 (10 µg/mL) (Abcam Inc.) for one hour at room temperature to detect protein bound

to the biotinylated GAGs. After additional washes, anti-rabbit HRP (1:500) was applied

for one hour at room temperature. Following final washes, color was developed by

addition of ABTS peroxidase substrate solution. Absorbance was measured at 405 nm

after quenching the wells with stop solution.

Page 92: In vitro and in vivo Growth Factor Delivery to

92

Rat Intra-articular Injection: 10 µg HB-IGF-1 in 50 µl saline, 10 µg IGF-1 in 50 µl saline,

or 50 µl saline alone was injected into the right knee joint of 2-month-old male Sprague-

Dawley rats. After one day, joint tissues were harvested, extracted, and analyzed by

Western blot. 5 ng of recombinant HB-IGF-1 or IGF-1 was loaded as standards. All

animal procedures were approved by the Harvard Medical Area Standing Committee on

Animals.

Human Cartilage Binding Assay: Joints from 4 human subjects were obtained

postmortem from the Gift of Hope Organ and Tissue Donor Network (Elmhurst, IL).

Cartilage disks (3 mm diameter, 0.8 mm thick) were harvested from femoropatellar

grooves of 26-year old (Collins grade 0)35, 49-year old (Collins grade 0), and 42-year old

female (Collins grade 2) knee joints and from a 28-year old (Collins grade 0) male knee

joint and cultured in 1% ITS serum-free high glucose DMEM supplemented with 500 nM

HB-IGF-1 or IGF-1. After 48 h (Day 0), disks were washed with PBS and incubated in

IGF-1 free medium. Disks were collected on Days 0, 1, 2 and 4, and protein was

extracted and analyzed for IGF-1 bound by Western blot using anti-IGF-1. 5 ng of

recombinant HB-IGF-1 or IGF-1 was loaded as standards. Procedures were approved by

the Office of Research Affairs at Rush–Presbyterian–St. Luke’s Medical Center and the

Committee on Use of Humans as Experimental Subjects at Massachusetts Institute of

Technology.

Statistical Analyses: All data are presented as mean ± standard error of the mean. Surface

plasmon resonance binding constants were log-transformed and evaluated by Student’s t-

Page 93: In vitro and in vivo Growth Factor Delivery to

93

test without assuming equal variances. Densitometry data for HB-IGF-1 binding to

human cartilage were rank-transformed and evaluated by paired t-test. Densitometry data

for HB-IGF-1 binding to CHO cells were rank-transformed and evaluated by Student’s t-

test without assuming equal variances. HB-IGF-1 loss to medium ELISA data were

analyzed by one-way ANOVA. Immobilized GAG ELISA data were analyzed by two-

way ANOVA followed by Posthoc Tukey tests. All tests were performed with acceptance

level α=0.05.

4.3 Results

HB-IGF-1 Is Retained in Cartilage Explants through Binding to Chondroitin Sulfate: To

determine which GAG was more important for HB-IGF-1 retention in cartilage, we first

tested whether removal of HS or CS affects HB-IGF-1 retention in cartilage explants.

Explants were first incubated with 500 nM HB-IGF-1 or IGF-1 for 48 h (days 0-2, Fig

4.1A). After washing the explants, they were then treated with either chondroitinase ABC,

heparitinase, or no enzyme for another 48 h (days 2-4). To rule out the possibility that the

high abundance of CS in the tissue could sterically block penetration of heparitinase into

cartilage and therefore block heparitinase action, at day 4 half of the explants treated with

chondroitinase were incubated with heparitinase for another two days. All other explants

were incubated in medium with no added enzymes during this time (days 4-6). All

explants were collected after this final incubation period (on day 6) and were analyzed by

Western blot for HB-IGF-1 or IGF-1 remaining in the explant.

Page 94: In vitro and in vivo Growth Factor Delivery to

94

As shown previously46, cartilage explants incubated with IGF-1 in the medium for two

days retained no IGF-1 in the tissue after 4 days of incubation in the absence of

exogenous IGF-1 (Fig 4.1B). In contrast, in the absence of enzymatic treatment, HB-IGF-

1 was strongly retained in cartilage explants. Unexpectedly, treatment with heparitinase

had no effect on the retention of HB-IGF-1 in the cartilage, whereas treatment with

chondroitinase ABC caused a substantial decrease in HB-IGF-1 remaining in the tissue.

While some HB-IGF-1 remained in the explants after treatment with chondroitinase,

treatment of these explants with heparitinase did not reduce retention of HB-IGF-1 any

further (Fig 4.1B, “C’ase+H’ase”). ELISA of conditioned medium confirmed that

significantly more HB-IGF-1 was released from chondroitinase-treated cartilage than

from heparitinase-treated cartilage, indicating comparatively low amounts of HB-IGF-1

bound to heparan sulfate ex vivo (Fig 4.1C).

Heparan Sulfate is Not Required for Retention of HB-IGF-1 on Cell Monolayers: To

confirm that heparan sulfate is not required, we tested retention of HB-IGF-1 and IGF-1

to mutant Chinese hamster ovary (CHO) cells (strain pgsD-677) that are unable to make

HS due to a genetic defect in HS chain polymerization26. The mutant cells upregulate

synthesis of CS and thus produce similar amounts of total sulfated GAG as the wild-type

cells26. After incubation of CHO cells with 100 nM HB-IGF-1 or IGF-1 in serum-free

medium, cells were washed and lysed for analysis by Western blot. HB-IGF-1 was

retained in the wildtype CHO cells after washing, whereas no IGF-1 was retained (Fig

4.2A,B). However, HB-IGF-1 was also retained in the HS-deficient cells, confirming that

HB-IGF-1 can be retained by binding to CS in the absence of HS (Fig 4.2A,B).

Page 95: In vitro and in vivo Growth Factor Delivery to

95

HB-IGF-1 but not IGF-1 Binds Immobilized Glycosaminoglycans (GAGs): In order to

quantify the binding affinities of HB-IGF-1 and IGF-1 for immobilized CS and HS, we

used surface plasmon resonance. Comparable levels of biotinylated HS and CS were

attached to a streptavidin-coated Biacore chip. A representative sensorgram is shown in

Fig 4.3A, demonstrating that while IGF-1 does not bind to either GAG (response units

(RU) <10), HB-IGF-1 binds to both HS and CS. Kinetic analysis of the surface plasmon

resonance curves confirmed that although HB-IGF-1 bound HS significantly more

strongly (KD = 21 ± 6 nM) than CS (KD = 160 ± 12 nM) (p = 0.012), the KD for the

binding of HB-IGF-1 to CS is well within the range of reported receptor-ligand

affinities24. This difference in KD values resulted from significantly different association

rate constants, (ka = 16x104 ± 6.6x104 (1/M·s) for HS vs. ka = 1.5x104 ± 0.06x104 (1/M·s)

for CS, p=0.035); the dissociation rates kd were similar (2.7x10-3 ± 0.93x10-3 (1/s) for HS

vs. 2.5x10-3 ± 0.16x10-3 (1/s) for CS), p=0.85).

The relative differences in binding affinities of HB-IGF-1 and IGF-1 to HS and CS were

confirmed by a sandwich ELISA. Binding of HB-IGF-1 to immobilized HS and CS

increased with concentration, with more binding to HS at the same given concentration of

HS or CS contained in each well (Fig 4.3B). Therefore, although HB-IGF-1 binds HS

with higher affinity, the data suggest that binding to CS would dominate in cartilage

tissue, where CS is ~500-1000 more abundant than HS16, 50.

Page 96: In vitro and in vivo Growth Factor Delivery to

96

HB-IGF-1 Is Preferentially Retained in Cartilage after Intra-articular Injection: The

ability of HB-IGF-1 to bind CS led us to hypothesize that it would be retained

preferentially in the GAG-rich cartilage tissues if delivered by intra-articular injection.

Consistent with this hypothesis, we found that one day after intra-articular injection in a

rat knee, HB-IGF-1 remained strongly detectable in articular cartilage extracts (Fig 4.4,

Articular Cartilage), despite stronger immunoreactivity of the IGF-1. HB-IGF-1 was also

slightly detectable in extracts of the fibrocartilaginous meniscus (Fig 4.4, Meniscus). In

contrast, HB-IGF-1 was not detectable in patella, patellar tendon, or muscle extracts (Fig

4.4). IGF-1 was not detectable in any of the tissues one day after injection (Fig 4.4).

HB-IGF-1 Is Retained in Human Cartilage: To examine whether HB-IGF-1 may be

relevant as a strategy for clinical delivery of growth factors to cartilage, we tested

whether HB-IGF-1 could also be retained in post-mortem adult human cartilage explants.

Human knee cartilage obtained from three Collins grade 0 donor joints (both male and

female) and one Collins grade 2 joint (female) was incubated with 500 nM HB-IGF-1 or

IGF-1 for two days, washed with PBS, and incubated in no-IGF-1 medium for an

additional 0, 1, 2, or 4 days. Binding of IGF-1 was only detectable immediately after

washing out the IGF (Fig 4.5A, Day 0). In contrast, HB-IGF-1 was retained after further

incubation for up to 4 days (Fig 4.5A). Analysis of Western blots from the four donor

cartilages by densitometry demonstrated that retention of HB-IGF-1 was significantly

higher than IGF-1 after 1, 2, and 4 days of incubation (Fig 4.5B).

Page 97: In vitro and in vivo Growth Factor Delivery to

97

4.4 Discussion

We demonstrate here that adding a heparin-binding motif to IGF-1 converts it from a

short-acting growth factor to one that can be locally delivered and retained in articular

cartilage in vivo. Contrary to our initial expectations, HB-IGF-1 does not require heparan

sulfate for retention in either cell culture or in cartilage explants. While we show that HB-

IGF-1 does have a higher affinity for heparan sulfate, its affinity for chondroitin sulfate is

within an order of magnitude. Therefore, in cartilage, where CS concentrations are 500-

1000 times higher than HS16, 50, binding to CS dominates and retention of HB-IGF-1 is

independent of HS.

These results led us to hypothesize that HB-IGF-1 could be preferentially delivered and

retained in articular cartilage due to its high concentration of sulfated

glycosaminoglycans14. We tested this hypothesis in rats and showed that after intra-

articular injection, HB-IGF-1 remains bound to the CS-rich articular cartilage, but not to

the adjacent patella, patellar tendon, or muscle tissue. In contrast, unmodified IGF-1 is

not able to bind either CS or HS, and was not detectable in either tissue after intra-

articular injection. Finally, we demonstrated that HB-IGF-1 is retained in adult human

cartilage whereas unmodified IGF-1 is not. Taken together, the results suggest that

modification of growth factors with heparin-binding domains may be a clinically relevant

strategy for local delivery to cartilage.

Our finding that HB-IGF-1 is retained primarily by chondroitin sulfate contrasts with the

extensive work demonstrating that the heparin-binding domain of FGF-2 binds primarily

Page 98: In vitro and in vivo Growth Factor Delivery to

98

to heparan sulfate proteoglycans in cartilage17, 50. This is likely explained by differences

in the binding specificities of the heparin-binding domains. In general, specificity for HS

depends on not only the heparin binding domains, but also on the secondary and/or

tertiary structure of the native proteins. The heparin-binding domain we used here to

make HB-IGF-1 came from HB-EGF. Structure-function analysis of HB-EGF has shown

that in addition to the heparin-binding domain, a portion of the EGF-like domain45 is

required for binding specifically to heparan sulfate. Similarly, FGF-2 affinity for HS has

been shown to depend on the spatial distribution of basic amino acids within the heparin-

binding loops of this molecule and on the specific conformation and topological

arrangement of these loops39. Since we added only the heparin-binding domain of HB-

EGF to IGF-1, it is likely that charge plays a primary role in the interactions of HB-IGF-1,

allowing it to bind other negatively charged glycosaminoglycans such as CS.

FGF-2 also appears to be retained in pericellular matrix as a reservoir of the growth

factor that does not activate the chondrocytes until released by matrix mechanical

stimulation or trauma. While we have not shown specific binding locations within

cartilage, we have previously demonstrated that retention of HB-IGF-1 is accompanied

by a sustained increase in 35S-sulfate incorporation46, strongly suggesting that factors

retained by chondroitin sulfate represent a pool that remains available to stimulate

chondrocyte receptors at the cell surface.

There may be additional mechanisms that are contributing to the differences seen here

between HB-IGF-1 and un-modified IGF-1. In particular, previous work has shown that

Page 99: In vitro and in vivo Growth Factor Delivery to

99

IGF-1 binds primarily to IGF binding proteins (IGFBPs) in cartilage, and not to ECM

constituents2, 13. Cartilage from osteoarthritic patients is known to be less responsive to

IGF-114, in part due to increased levels of IGF binding proteins (IGFBPs)8, 14, 28, 31, 32.

IGFBPs are present at a binding site density of 30-150 nM in bovine cartilage13 and can

bind IGF-1 with an equilibrium affinity (KD) of ~5 nM13. The KD found for HB-IGF-1

binding to heparan sulfate is on the order of that found for IGF-1 binding to IGFBPs,

indicating that it may be able to compete for IGFBP binding, possibly resulting in more

bioactivity than normal IGF-1 in addition to better delivery. Moreover, the modification

of the amino-terminus of IGF-1 by inclusion of the heparin-binding domain may decrease

the affinity of HB-IGF-1 to IGFBPs, as the deletion of the first three amino acids in the

case of the mutated analog des(1-3)IGF-1 decreased binding to IGFBPs by 100-fold1.

In conclusion, HB-IGF-1 may be a new therapeutic for sustained and relatively specific

local delivery of IGF-1 to cartilage through its preferential retention in CS-rich tissues.

Modification of growth factors by addition of heparin binding domains may therefore be

a novel strategy for targeted delivery to cartilage after intra-articular injection.

4.5 Acknowledgements

I would like to thank Alan J. Grodzinsky, Sc.D., Richard T. Lee, M.D., and Parth Patwari,

M.D., Sc.D. for their scientific guidance, Kiersten Cummings, B.S. for her expertise in

manufacturing HB-IGF-1, and Anna H. K. Plaas, Ph.D. and Ada A. Cole, Ph.D. for

supplying human donor tissue. I would also like to thank Dr. Jeff Esko for helpful

discussions. Finally, I am grateful for the following funding sources: NIH-NIBIB Grant

Page 100: In vitro and in vivo Growth Factor Delivery to

100

EB003805 and NIH-NIAMS Grant AR045779; NDSEG and NSF graduate fellowships; a

collaborative research grant from the Massachusetts Life Sciences Center and funding

from Biomeasure, Inc.

4.6 References

1. Bagley CJ, May BL, Szabo L, McNamara PJ, Ross M, Francis GL, Ballard FJ, and Wallace JC. A key functional role for the insulin-like growth factor 1 N-terminal pentapeptide. Biochem J. 1989; 259:665-71.

2. Bhakta NR, Garcia AM, Frank EH, Grodzinsky AJ, and Morales TI. The Insulin-like Growth Factors (IGFs) I and II Bind to Articular Cartilage via the IGF-binding Proteins. J Biol Chem. 2000; 275:5860-5866.

3. Bonassar LJ, Grodzinsky AJ, Frank EH, Davila SG, Bhaktav NR, and Trippel SB. The effect of dynamic compression on the response of articular cartilage to insulin-like growth factor-I. J Orthop Res. 2001; 19:11-17.

4. Cardin AD and Weintraub HJ. Molecular modeling of protein-glycosaminoglycan interactions. Arterioscler Thromb Vasc Biol. 1989; 9:21-32.

5. Chan JM, Stampfer MJ, Giovannucci E, Gann PH, Ma J, Wilkinson P, Hennekens CH, and Pollak M. Plasma Insulin-Like Growth Factor-I and Prostate Cancer Risk: A Prospective Study. Science. 1998; 279:563-566.

6. Curtis AJ, Devenish RJ, and Handley CJ. Modulation of aggrecan and link-protein synthesis in articular cartilage. Biochem J. 1992; 288:721-726.

7. David G, Bai XM, Van der Schueren B, Cassiman JJ, and Van den Berghe H. Developmental changes in heparan sulfate expression: in situ detection with mAbs. J. Cell Biol. 1992; 119:961-975.

8. Dore S, Pelletier JP, DiBattista JA, Tardif G, Brazeau P, and Martel-Pelletier J. Human osteoarthritic chondrocytes possess an increased number of insulin-like growth factor 1 binding sites but are unresponsive to its stimulation. Possible role of IGF-1-binding proteins. Arthritis Rheum. 1994; 37:253-63.

9. Farach-Carson MC, Hecht JT, and Carson DD. Heparan Sulfate Proteoglycans: Key Players in Cartilage Biology. Critical Reviews in Eukaryotic Gene Expression. 2005; 15:29-48.

10. Flaumenhaft R, Moscatelli D, Saksela O, and Rifkin DB. Role of extracellular matrix in the action of basic fibroblast growth factor: Matrix as a source of growth factor for long-term stimulation of plasminogen activator production and DNA synthesis. J Cellular Physiology. 1989; 140:75-81.

11. Fortier LA, Mohammed HO, Lust G, and Nixon AJ. Insulin-like growth factor-I enhances cell-based repair of articular cartilage. J Bone Joint Surg Br. 2002; 84-B:276-288.

12. Friedrich U, Blom AM, Dahlback B, and Villoutreix BO. Structural and Energetic Characteristics of the Heparin-binding Site in Antithrombotic Protein C. J. Biol. Chem. 2001; 276:24122-24128.

Page 101: In vitro and in vivo Growth Factor Delivery to

101

13. Garcia A, Szasz N, Trippel SB, Morales T, Grodzinsky AJ, and Frank E. Transport and binding of insulin-like growth factor I through articular cartilage. Arch Biochem Biophys. 2003; 415:69-79.

14. Goldring MB. Update on the biology of the chondrocyte and new approaches to treating cartilage diseases. Best Practice & Research Clinical Rheumatology. 2006; 20:1003-1025.

15. Goodrich LR, Hidaka C, Robbins PD, Evans CH, and Nixon AJ. Genetic modification of chondrocytes with insulin-like growth factor-1 enhances cartilage healing in an equine model. J Bone Joint Surg Br. 2007; 89-B:672-685.

16. Govindraj P, West L, Koob TJ, Neame P, Doege K, and Hassell JR. Isolation and Identification of the Major Heparan Sulfate Proteoglycans in the Developing Bovine Rib Growth Plate. J Biol Chem. 2002; 277:19461-19469.

17. Govindraj P, West L, Smith S, and Hassell JR. Modulation of FGF-2 binding to chondrocytes from the developing growth plate by perlecan. Matrix Biol. 2006; 25:232-9.

18. Heinegard D. Proteoglycans and more - from molecules to biology. Int J Exp Path. 2009; 90:575-586.

19. Hui W, Rowan AD, and Cawston T. Modulation of the expression of matrix metalloproteinase and tissue inhibitors of metalloproteinases by TGF-β1 and IGF-1 in primary human articular and bovine nasal chondrocytes stimulated with TNF-α. Cytokine. 2001; 16:31-35.

20. Hunziker EB and Rosenberg LC. Repair of partial-thickness defects in articular cartilage: cell recruitment from the synovial membrane. J Bone Joint Surg Am. 1996; 78:721-33.

21. Jabri N, Schalch D, Schwartz S, Fischer J, Kipnes M, Radnik B, Turman N, Marcsisin V, and Guler H. Adverse effects of recombinant human insulin-like growth factor I in obese insulin-resistant type II diabetic patients. Diabetes. 1994; 43:369-374.

22. Kirn-Safran CB, Gomes RR, Brown AJ, and Carson DD. Heparan sulfate proteoglycans: Coordinators of multiple signaling pathways during chondrogenesis. Birth Defects Research Part C: Embryo Today: Reviews. 2004; 72:69-88.

23. Laron Z. Somatomedin-1 (Recombinant Insulin-Like Growth Factor-1): Clinical Pharmacology and Potential Treatment of Endocrine and Metabolic Disorders. BioDrugs. 1999; 11:55-70.

24. Lauffenburger DA and Linderman JJ. Receptors: Models for Binding, Trafficking, and Signaling. 1993, New York: Oxford University Press.

25. Ley K, Cerrito M, and Arfors K-E. Sulfated polysaccharides inhibit leukocyte rolling in rabbit mesentery venules. Am J Physiol. 1991; 260:H1667-H1673.

26. Lidholt K, Weinke JL, Kiser CS, Lugemwa FN, Bame KJ, Cheifetz S, Massague J, Lindahl U, and Esko JD. A single mutation affects both N-acetylglucosaminyltransferase and glucuronosyltransferase activities in a Chinese hamster ovary cell mutant defective in heparan sulfate biosynthesis. Proc Natl Acad Sci U S A. 1992; 89:2267-2271.

Page 102: In vitro and in vivo Growth Factor Delivery to

102

27. Loeser RF and Shanker G. Autocrine stimulation by insulin-like growth factor 1 and insulin-like growth factor 2 mediates chondrocyte survival in vitro. Arthritis Rheum. 2000; 43:1552-1559.

28. Loeser RF, Shanker G, Carlson C, Gardin J, Shelton B, and Sonntag W. Reduction in the chondrocyte response to insulin-like growth factor in aging and osteoarthritis. Arthritis Rheum. 2000; 43:2110-2120.

29. Luyten FP, Hascall VC, Nissley SP, Morales TI, and Reddi AH. Insulin-like growth factors maintain steady-state metabolism of proteoglycans in bovine articular cartilage explants. Arch Biochem Biophys. 1988; 267:416-425.

30. Madry H, Kaul G, Cucchiarini M, Stein U, Zurakowski D, Remberger K, Menger MD, Kohn D, and Trippel SB. Enhanced repair of articular cartilage defects in vivo by transplanted chondrocytes overexpressing insulin-like growth factor I (IGF-I). Gene Ther. 2005; 12:1171-1179.

31. Martel-Pelletier J, Di Battista JA, Lajeunesse D, and Pelletier JP. IGF/IGFBP axis in cartilage and bone in osteoarthritis pathogenesis. Inflamm Res. 1998; 47:90-100.

32. Martin JA, Ellerbroek SM, and Buckwalter JA. Age-related decline in chondrocyte response to insulin-like growth factor-I: The role of growth factor binding proteins. J Orthop Res. 1997; 15:491-498.

33. Matsumoto T, Gargosky SE, Iwasaki K, and Rosenfeld RG. Identification and characterization of insulin-like growth factors (IGFs), IGF-binding proteins (IGFBPs), and IGFBP proteases in human synovial fluid. J Clin Endocrinol Metab. 1996; 81:150-5.

34. McQuillan DJ, Handley CJ, Campbell MA, Bolis S, Milway VE, and Herinton AC. Stimulation of proteoglycan biosynthesis by serum and insulin-like growth factor-I in cultured bovine articular cartilage. Biochem J. 1986; 240:423-430.

35. Muehleman C, Bareither D, Huch K, Cole AA, and Kuettner KE. Prevalence of degenerative morphological changes in the joints of the lower extremity. Osteoarthritis Cartilage. 1997; 5:23-37.

36. Nixon A, Saxer R, and Brower-Toland BD. Exogenous insulin-like growth factor-I stimulates an autoinductive IGF-I autocrine/paracrine response in chondrocytes. J Orthop Res. 2001; 19:26-32.

37. Nixon AJ, Fortier LA, Williams J, and Mohammed H. Enhanced repair of extensive articular defects by insulin-like growth factor-I-laden fibrin composites. J Orthop Res. 1999; 17:475-87.

38. Oike Y, Kimata K, Shinomura T, Nakazawa K, and Suzuki S. Structural analysis of chick-embryo cartilage proteoglycan by selective degradation with chondroitin lyases (chondroitinases) and endo-beta-D-galactosidase (keratanase). Biochem J. 1980; 191:193-207.

39. Raman R, Venkataraman G, Ernst S, Sasisekharan V, and Sasisekharan R. Structural specificity of heparin binding in the fibroblast growth factor family of proteins. Proc Natl Acad Sci U S A. 2003; 100:2357-62.

40. Ricard-Blum S, Feraud O, Lortat-Jacob H, Rencurosi A, Fukai N, Dkhissi F, Vittet D, Imberty A, Olsen BR, and van der Rest M. Characterization of Endostatin Binding to Heparin and Heparan Sulfate by Surface Plasmon Resonance and Molecular Modeling. J Biol Chem. 2004; 279:2927-2936.

Page 103: In vitro and in vivo Growth Factor Delivery to

103

41. Saksela O, Moscatelli D, Sommer A, and Rifkin DB. Endothelial cell-derived heparan sulfate binds basic fibroblast growth factor and protects it from proteolytic degradation. J Cell Biol. 1988; 107:743-51.

42. Schmidt MB, Chen EH, and Lynch SE. A review of the effects of insulin-like growth factor and platelet derived growth factor on in vivo cartilage healing and repair. Osteoarthritis Cartilage. 2006; 14:403-412.

43. Schneiderman R, Rosenberg N, Hiss J, Lee P, Liu F, Hintz RL, and Maroudas A. Concentration and size distribution of insulin-like growth factor-1 in human normal and osteoarthritic synovial fluid and cartilage. Arch Biochem Biophys. 1995; 324:173-188.

44. Silverberg I, Havsmark B, and Fransson L-Å. The substrate specificity of heparan sulphate lyase and heparin lyase from Flavobacterium heparinum. Carbohydrate Research. 1985; 137:227-238.

45. Thompson SA, Higashiyama S, Wood K, Pollitt NS, Damm D, McEnroe G, Garrick B, Ashton N, Lau K, Hancock N, Klagsbrun M, and Abraham JA. Characterization of sequences within heparin-binding EGF-like growth factor that mediates interaction with heparin. J Biol Chem. 1994; 269:2541-2549.

46. Tokunou T, Miller R, Patwari P, Davis ME, Segers VF, Grodzinsky AJ, and Lee RT. Engineering insulin-like growth factor-1 for local delivery. FASEB J. 2008; 22:1886-93.

47. Tyler JA. Insulin-like growth factor I can decrease degradation and promote synthesis of proteoglycan in cartilage exposed to cytokines. Biochem J. 1989; 260:543-548.

48. Vincent T, Hermansson M, Bolton M, Wait R, and Saklatvala J. Basic FGF mediates an immediate response of articular cartilage to mechanical injury. Proc Natl Acad Sci U S A. 2002; 99:8259-64.

49. Vincent TL, Hermansson MA, Hansen UN, Amis AA, and Saklatvala J. Basic fibroblast growth factor mediates transduction of mechanical signals when articular cartilage is loaded. Arthritis Rheum. 2004; 50:526-33.

50. Vincent TL, McLean CJ, Full LE, Peston D, and Saklatvala J. FGF-2 is bound to perlecan in the pericellular matrix of articular cartilage, where it acts as a chondrocyte mechanotransducer. Osteoarthritis Cartilage. 2007; 15:752-63.

Page 104: In vitro and in vivo Growth Factor Delivery to

104

4.7 Figures

Figure 4.1. Retention of HB-IGF-1 in bovine cartilage explants following enzymatic digestion of glycosaminoglycans. (A) HB-IGF-1 (HB) or IGF-1 (I) was incubated with cartilage disks for two days (Day 0 to Day 2) followed by treatment with no enzyme (No Enz), chondroitinase (C’ase), or heparitinase (H’ase) for an additional two days (Day 2 to Day 4). At Day 4, a subset of chondroitinase-treated disks was incubated with heparitinase (C’ase+H’ase) for two days while all remaining disks were kept in enzyme-free medium. (B) Western analysis of HB-IGF-1 or IGF-1 remaining in the cartilage tissue at Day 6. The blot shown is representative of four repeats. (C) ELISA of HB-IGF-1 released to the medium following 48-hour enzyme treatment of cartilage explants (Days 2-4 for No enzyme, C’ase, and H’ase conditions; Days 4-6 for C’ase + H’ase). mean ± SEM, * vs. No enzyme, n=4, p<0.001.

Page 105: In vitro and in vivo Growth Factor Delivery to

105

Figure 4.2. Retention of HB-IGF-1 on cells lacking heparan sulfate. (A) HB-IGF-1 (“HB”) and IGF-1 (“I”) were incubated with mutant CHO cells unable to produce heparan sulfate (“no HS”) and wildtype CHO cells (“WT”), then washed in PBS. Western analysis of the cell lysates for IGF showed that HB-IGF-1 remained bound to cells with or without the presence of heparan sulfate, whereas IGF-1 binding was not detectable. (B) Densitometry of Western blots from four repeated experiments, each normalized to wild-type HB-IGF-1 binding. mean ± SEM, * vs. WT HB, p<0.005.

Page 106: In vitro and in vivo Growth Factor Delivery to

106

Figure 4.3. Binding analysis of HB-IGF-1 and IGF-1 to isolated glycosaminoglycans. (A) Representative sensorgram for Biacore kinetic analysis over HS or CS surfaces using 250 nM HB-IGF-1 or IGF-1. HB-IGF-1 is shown in the top two curves (black) with corresponding equilibrium binding constants, KD, determined from a minimum of three concentrations used during three experimental repeats. IGF-1 (bottom two curves, grey) was unable to bind either surface (RU < 10). (B) Sandwich ELISA detecting absorbance at a given HB-IGF-1 (solid line, black) or IGF-1 (dashed line, grey) concentration resulting from binding HS or CS. Representative of two repeats, each with duplicate wells. n=2, mean ± SEM, * vs. CS, p<0.05.

Page 107: In vitro and in vivo Growth Factor Delivery to

107

Figure 4.4. Retention of HB-IGF-1 in vivo. Western blot showing retained IGF-1 (I) or HB-IGF-1 (HB) in rat cartilage, meniscus, patella, patellar tendon, or muscle extracts one day after intra-articular injection of IGF-1, HB-IGF-1, or saline.

Page 108: In vitro and in vivo Growth Factor Delivery to

108

Figure 4.5. Retention of HB-IGF-1 in human cartilage explants. Human cartilage (grade 0 to 2) was incubated with HB-IGF-1 or IGF-1 for two days (Day -2 to 0), washed, and incubated in IGF-free medium for 4 days (Days 0-4). (A) Amount of HB-IGF-1 or IGF-1 remaining in cartilage after 0-4 days. The experiment was performed on cartilage from four donors and a representative Western blot is shown. (B) Analysis of four Western blots by densitometry, normalized to the density of 5 ng of the respective protein standard. mean ± SEM, * vs. IGF-1, p<0.05.

Page 109: In vitro and in vivo Growth Factor Delivery to

109

Chapter 5. Conclusions and Future Directions

The inability of articular cartilage to repair itself after acute injury has been implicated in

the development of osteoarthritis. Providing anabolic stimulation through delivery of

growth factors may improve the ability of endogenous and exogenous cell sources to

initiate repair. Additionally, providing a chondrogenic microenvironment may further

stimulate repair by native or delivered bone-marrow-derived stromal cells. The objective

of this work was to develop methods for delivering growth factors to cartilage and to test

the ability of a self-assembling peptide scaffold, (KLDL)3, with or without growth factors

to augment repair. Delivery methods included growth factor adsorption, scaffold-

tethering, and modification of growth factor structure.

In Chapter 2, two different modes of growth factor delivery were investigated: adsorption

and tethering. IGF-1 and TGF-β1 were pre-mixed with the self-assembling peptide

hydrogel, (KLDL)3 or KLD, solution to adsorb the growth factors to the scaffold during

the assembly process. Biotinylated versions of the growth factors were tethered to the

peptide through streptavidin and biotinylated KLD. Adsorption was able to deliver TGF-

β1 in sufficient quantities to induce chondrogenesis of bone marrow derived stromal cells

(BMSCs); IGF-1 diffused too quickly out of the scaffold to stimulate extracellular matrix

production by chondrocytes. Additionally, while tethering these factors through a biotin-

streptavidin bond provided long-term sequestration, tethered growth factors were not

effective in stimulating proteoglycan production. Therefore, while self-assembling

peptide sequences are readily functionalized, the manner in which growth factors are

Page 110: In vitro and in vivo Growth Factor Delivery to

110

delivered affects bioactivity and varies for specific growth factors and biological systems

of interest.

To improve tethering, one could use a lower affinity tethering system to allow reversible

tethering, or one could incorporate cleavable links in the peptide sequence. For instance,

one could use an MMP- or ADAMTS- cleavable link before the growth factor linkage, so

that upon upregulation of these enzymes the growth factor would be released.

In Chapter 3, the self-assembling peptide hydrogel KLD with or without chondrogenic

factors and allogeneic BMSCs was evaluated in a full-thickness, critically sized rabbit

cartilage defect model for ability to stimulate cartilage repair. Delivering KLD alone

resulted in improved filling of osteochondral defects and improved cartilage repair, as

seen by cumulative histology score, Safranin-O staining, and type II collagen

immunostaining. In this small animal model, the full-thickness defect provided access to

the marrow, similar in concept to abrasion arthroplasty or spongialization in large animal

models (goat, sheep, horse, human), and suggests that combining KLD with these

techniques may offer an improvement over current practice. Ongoing studies include the

evaluation of KLD in a clinically relevant sized equine defect co-treated with

microfracture and subjected to strenuous exercise, compared to defects treated with

microfracture alone.

Even though the chondrogenic factors and BMSCs selected for this study proved

sufficient to induce chondrogenesis in vitro, they did not have the intended effect when

Page 111: In vitro and in vivo Growth Factor Delivery to

111

delivered in vivo, highlighting the necessity of further in vivo testing. The length of time

the peptide hydrogel is present in the defect and the degradation kinetics should be also

addressed in future in vivo and in vitro studies.

In Chapter 4, it was shown that adding a heparin-binding motif to IGF-1 converts this

protein from a short-acting growth factor to one that can be locally delivered and retained

in articular cartilage in vivo. HB-IGF-1 does not require heparan sulfate for retention in

either cell culture or in cartilage explants. While HB-IGF-1 does have a higher affinity

for heparan sulfate, its affinity for chondroitin sulfate is within an order of magnitude.

Therefore, in cartilage, where CS concentrations are found to be 500-1000 times higher

than HS, binding to CS dominates and retention of HB-IGF-1 is independent of HS.

These results led to the hypothesis that HB-IGF-1 could be preferentially delivered and

retained in articular cartilage due to its high concentration of sulfated

glycosaminoglycans. This hypothesis was tested in rats in vivo, and it was shown that

after intra-articular injection, HB-IGF-1 remained bound to the CS-rich articular

cartilage, but not to the adjacent patella, patellar tendon, or muscle tissue. In contrast,

unmodified IGF-1 was not able to bind either CS or HS, and was not detectable in either

tissue after intra-articular injection. Finally, it was demonstrated that HB-IGF-1 was

retained in adult human cartilage, whereas unmodified IGF-1 is not. Taken together, the

results suggest that modification of growth factors with heparin-binding domains may be

a clinically relevant strategy for local delivery to cartilage. Ongoing studies focus on

Page 112: In vitro and in vivo Growth Factor Delivery to

112

evaluating the ability of HB-IGF-1 to promote proteoglycan synthesis in rat cartilage ex

vivo following intra-articular injection.

While injection of the protein alone has been shown to be adequate to deliver HB-IGF-1

to cartilage for up to four days, and sustained proteoglycan synthesis for up to eight days,

to attain longer-term delivery, a carrier may be necessary. One option would be to use a

hydrogel such as KLD with incorporation of chondroitin sulfate or heparan sulfate to

increase binding and modulate the release kinetics. Another option would be to

encapsulate HB-IGF-1 in polymer microspheres and deliver the microspheres in a

hydrogel to the joint. In addition, radiolabel studies with 125I-HB-IGF-1 will enable better

characterization of the uptake into KLD and the further delivery into cartilage tissue.

In conclusion, the work in this thesis provides a basis for future growth factor delivery

studies in the context of cartilage and suggests that the self-assembling peptide KLD and

the fusion protein HB-IGF-1 may be further developed to aid in cartilage repair.

Page 113: In vitro and in vivo Growth Factor Delivery to

113

Appendix A – Protocols and data related to Chapter 2 Encapsulating cells in KLD with or without biotin-streptavidin tethering of growth factors Use tissue-culture treated 24-well plates if using chondrocytes, non-tissue cultured if using BMSCs (BMSCs tend to crawl out of the gels and stick to the bottom of tissue-culture treated plates). Agarose ring molds: Fill wells of a 24-well plate with 0.6 mL of 2-3% sterile low-melting point agarose. Let agarose solidify at 4 °C for a few hours. In the hood, use a sterile 6 mm punch to make holes in the center of each well. Use a sterile spatula to remove these holes. Use a sterile 12 mm punch to make holes centered around the 6 mm holes you just

created. Again, use a sterile spatula to remove the agarose around the hole you just punched so

you are left with a ring with an outer diam of 12 mm and an inner diam of 6 mm. Casting with KLD: You will need the following solutions: 10% sterile sucrose 10% sterile sucrose + 2.5 mM HEPES HGDMEM + 25 mM HEPES Culture medium (usually 1% ITS+1 with HGDMEM unless doing IGF-1 experiments,

then mini ITS+1 (0.003% ITS+1)) And the following materials: KLD (6 mg per round of casting) For tethering: biotinylated KLD (bKLD), streptavidin, biotinylated IGF-1 (bIGF-1) 2 mL tubes with 0.9 mL Tris buffer in them Repeat pipettor with a 1 mL sterile syringe for each round of casting Fill the wells of the 24-well plate with HGDMEM+HEPES so that the rings get saturated

with medium in order to aid in self-assembly of the peptide.

Resuspend KLD at 4.5 mg/mL with sterile 10% sucrose. If tethering, you may want to increase this to 4.6 mg/mL to account for the extra volume you will be adding. For this protocol, I am assuming a final concentration of ~3.5 mg/mL. You can safely use a final concentration in the range of 3.2 – 4 mg/mL. Sonicate for about 60 min, vortexing and spinning down intermittently.

If tethering, resuspend bKLD at 2 mg/mL with sterile 10% sucrose. Sonicate for at least 30 min – bKLD at this concentration goes into solution much faster than KLD.

When KLD is in solution, transfer 1.2 mL to a polypropylene 5 mL tube for each round of casting you plan to do. (1.165 mL if tethering). Put tube in sonicator.

Page 114: In vitro and in vivo Growth Factor Delivery to

114

If tethering, add 27 uL bKLD to each tube (1:100 concentration of bKLD:KLD). Pre-mix streptavidin and bIGF-1 in equimolar amounts. If you want 300 ng/mL bIGF-1

inside the peptide, combine 4.5 uL of bIGF at 100 ug/mL and 3.34 uL of streptavidin at 1 mg/mL. Add this mixture to your peptide tube with bKLD in it about 5-15 minutes before casting. (If you want to adsorb growth factors, you would just add your growth factor directly to the peptide mixture at this point without including the streptavidin or bKLD.)

When all peptide tubes look like they have a low viscosity and few bubbles, spin down your tube of cells (200g for 8 min for chondrocytes, 100g for 5 min for BMSCs).

While the cells are spinning, aspirate the medium out of the centers of your agarose rings in your 24-well plates. Make sure that the rings are not leaking. If they are, just remove all of the medium from the well.

When the cells are finished, aspirate the supernatant and resuspend your cells in 0.3 mL of sucrose+HEPES. Make sure the pellet is broken up completely as cell clumps generally result in death.

Pull up all of the cells and put them into your peptide tube. Use your thumb to cover the top of the tube and vortex gently and briefly (~ speed 4) to homogenize the cells and peptide mixture.

Pull up the cell/peptide solution in your repeat pipettor set to 50 µL. Click the dispenser button once so that the volume numbers stop blinking.

Now quickly put 50 µL of cells/peptide in the middle of each ring (generally get 19-20 gels per round of casting).

When you are finished, use a P200 to put 50 µL of leftover cell/peptide solution in tubes containing 900 µL Tris buffer for use as day 0 DNA standards.

About 2-5 minutes after casting, you should put either HGDMEM+HEPES or culture medium in all of your wells so that the gels finish assembling and the pH is neutralized.

Repeat this process for the remaining rounds of casting.

Page 115: In vitro and in vivo Growth Factor Delivery to

115

Dose-response with soluble IGF-1 and bIGF-1 on chondrocytes encapsulated in KLD

Figure A1. Effect of soluble IGF-1 and bIGF-1 on sGAG and DNA content of chondrocytes encapsulated in KLD. Chondrocytes were encapsulated in KLD and cultured for 4 days in soluble IGF-1, soluble bIGF-1, or 1% ITS+1. Treatments were started on day 1 post encapsulation. sGAG and DNA content per gel are shown. mean±SEM. n=4.

Page 116: In vitro and in vivo Growth Factor Delivery to

116

Figure A2. Effect of soluble IGF-1 and bIGF-1 on 35S-sulfate incorporation of chondrocytes encapsulated in KLD. Chondrocytes were encapsulated in KLD and cultured for 4 days in soluble IGF-1, soluble bIGF-1, or 1% ITS+1. Treatments were started on day 1 post encapsulation; Radiolabeled on day 4; Ended experiment on day 5. mean±SEM. n=4.

Page 117: In vitro and in vivo Growth Factor Delivery to

117

Effect of tethered IGF-1 on BMSCs encapsulated in KLD

Figure A3. Effect of tethered bIGF-1 on BMSCs encapsulated in KLD. TGFβ-1 (100 ng/mL) and dexamethasone (100 nM) were adsorbed to KLD gels with or without 300 ng/mL biotinylated-IGF-1/streptavidin/biotinylated-KLD. (A) sGAG retained in the hydrogels. (B) DNA content of the hydrogels. (C) 35S-sulfate incorporation over the last 24 hours. (D) 3H-proline incorporation over the last 24 hours. n=16 (3 animals, 4 experiments, n=4 per experiment). mean ± sem. * vs. Day 14, † vs. Day 21, p<0.05 (linear mixed model with animal as a random factor on log transformed data).

Page 118: In vitro and in vivo Growth Factor Delivery to

118

Flow Cytometry Invitrogen - #V13241 – Vybrant Apoptosis Assay Kit #2 Accuri C6 Flow Cytometer – Schauer and Lauffenburger labs each have this machine Can reuse and autoclave syringe filters and plastic filter holders PI (propidium iodide, red fluorescence): >670 nm (FL3) AlexaFluor 488-Annexin V: 488 nm (FL1) Some annexin binding buffer comes with this kit, but you can make more of your own: ABB Recipe (500mL) - 10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl2, pH 7.4 1.192g HEPES 4.0908g NaCl 0.184g CaCl2*2H2O ~2mL of 2M NaOH 1. Assemble syringes/filters by putting a filter between a top and bottom plastic part and screwing them together. Put this on the bottom of a 1 mL syringe with the plunger removed. 2. Transfer 1-2 gels from the same condition to 1 mL syringe. 3. Add 300-500 µL PBS to wash the gel(s) down and push them through the syringe with the plunger. Collect the output in a large Eppendorf tube. You can use more PBS if necessary, the important part is to get the gel(s) washed down and in liquid so that when you push the plunger through, the gel is actually pushed through the filter and not getting stuck to the sides of the syringe. 4. Centrifuge the large Eppendorf tube at 200g for 8 min. 5. Keep pellet (cells). Some of the peptide will collect near the pellet but still in the supernatant, so you can discard that. 6. Reconstitute with ~400 µL annexin binding buffer to get ~1x106 cells/mL. You should count the cells the first time you do this and optimize it for your experiment. 7. Put 100 µL of each sample in new Eppendorf tube. 8. Add 1 µL of working stock PI to each tube. 9. Add 3 µL AlexaFluor488-Annexin V to each tube. 10. Use leftover cells to make up 3 types of controls:

a) Combine equal amounts of each type of sample into 3 tubes for total of 100 uL/tube. b) Keep one control unstained. Stain one control with red only. Stain one control with green only.

11. Vortex lightly to mix. 12. Stand at room temp for 15 min in the dark. 13. Add 0.4 mL annexin-binding buffer to each tube. Keep samples on ice. 14. Run through flow cytometer. Run controls first to set gates and adjust the compensation. You should not have to adjust the compensation more than a few % to get the green control samples all in the lower right quadrant instead of some in the upper right quadrant. Keep some cell only samples (cells that were never encapsulated in peptide) if possible to identify what is peptide and what is cells in the side scatter vs forward scatter plot.

Page 119: In vitro and in vivo Growth Factor Delivery to

119

15. Population should separate into 3 quadrants: lower left = alive; lower right = green, apoptotic; upper right = red and green, dead. Example pictures of side vs forward scatter plots:

Three controls, FL3 vs FL1 plots:

And looking at the chondrocyte gate for the staurosporine positive control:

Figure A4. Example flow cytometry images.

Page 120: In vitro and in vivo Growth Factor Delivery to

120

Figure A5. Effect of casting order on cell death. There was a slight increase with time, but overall it seems the largest variability is between different rounds of casting and with cell density.

Page 121: In vitro and in vivo Growth Factor Delivery to

121

Figure A6. Chondrocytes encapsulated in KLD with or without tethered IGF-1 were evaluated for cell death and apoptosis by staining with propidium iodide and annexin v and performing flow cytometry at days 1, 2, 3, and 6 post encapsulation. Medium was supplemented with No IGF-1, soluble IGF-1 (300 ng/mL), or staurosporine (1 µM). mean ± SEM. n=5 experiments (2 gels averaged for each experiment) for day 1. n=2 for day 2. n=4 for day 3. n=1 for day 6. Day 1 values ranged from 20-41% death for no IGF-1. Intra-experiment variation was <3% per condition. Stats: ANOVA on days 1 and 3 data with Tukey post-hoc, * vs. staurosporine, p<0.05.

Page 122: In vitro and in vivo Growth Factor Delivery to

122

Casting Alginate Beads with Cells Adapted from Delphine Dean and Bobae Lee. Autoclave:

• 50 mL beaker • 3-500 mL bottles (for solutions below) • 1 spatula (to move beads)

Solutions:

• 2% alginate in 0.9% NaCl • 0.9% NaCl: 9 g in 1000 mL DI water • 150 mM NaCl (FW 58.44): 4.38 g in 500 mL DI water • 102 mM CaCl2 (FW 111) in 0.9% NaCl: 5.66 g CaCl2 in 500 mL of 150 mM

NaCl • 55 mM Na Citrate (FW 294.10) in 150 mM NaCl: 8.09 g Na Citrate in 500 mL of

150 mM NaCl Filter all solutions through a 200 µm filter and into the sterile bottles. To make the beads

1. Fill the sterile 50 mL beaker with 30 mL of 102 mM CaCl2 solution. 2. Get a cell count of cells using the hemocytometer. Spin cells down (if needed).

1800 rpm, 10 min. Suction off supernatant (being careful not to suck up any cells.) Gently tap bottom of vial to break up cells. May need to add 1 mL sterile PBS (no Mg++ and Ca++, pH 7) to help break up the cells.

3. Add appropriate amount of alginate. [ideal conc: 10-15x106 cells/mL; can be as high as 20x106 cells/mL] (I did 20 million cells/mL and it worked fine.)

4. Use 16G needle and 3 mL syringe to suction up the alginate-chondrocyte solution. Remove as many air bubbles as possible from the syringe.

5. Remove 16G needle and replace with 22G needle. Hold the syringe at a 45° angle with the hole facing downward.

6. With steady pressure, drop even-sized droplets into the CaCl2 solution. Halfway-through you may want to exchange the CaCl2 solution with fresh solution so the Ca++ does not get depleted.

7. After 10 minutes, suction off the CaCl2 and rinse twice with 20 mL sterile PBS (no Mg++ and Ca++). Let each rinse go for 1 min. (Note: CaCl2 + PBS causes a precipitate to form, so only do 1 min rinses or use DMEM instead of PBS.)

8. Replace PBS with culture medium. 9. Fill a 12-well plate with 2 mL culture medium in each well. Using a sterile

spatula, transfer 6-8 beads/well. 10. Do a cell viability assay on one of the beads. 11. Keep alginate-cell beads in an incubator.

To dissolve the beads – I used the first method

1. Put 4 beads into a 1.5 mL eppendorf tube. Add 1.2 mL NaCitrate. Place in a 37 °C water bath or incubator for 10 minutes. Shake gently to mix. Do not vortex.

Page 123: In vitro and in vivo Growth Factor Delivery to

123

2. Spin 3000-4000 rpm (~1000 rcf) in Eppendorf centrifuge 5415C for 5 minutes. 3. Remove supernatant. Add 1 mL 0.9% NaCl. Centrifuge 3000-4000 rpm, 5 min.

Repeat. 4. Resuspend final pellet in PBS (if doing a cell viability assay) or culture medium at

desired concentration (~0.5 ml -> gave me about 1 million cells/mL). 5. Do a cell viability assay – should get <5% dead.

OR

1. Put 15 beads into a 15 mL conical tube. Add 5 mL NaCitrate. Place in 37 °C water bath for 10 minutes. Shake gently to mix. 5 more minutes with frequent shaking if the beads are not dissolved. Do not vortex.

2. Spin <1000 rpm (with CBE centrifuge) for 6 min. 3. Remove supernatant. Add 3 mL of 0.9% NaCl to rinse. Spin <1000 rpm.

Resuspend in medium.

Page 124: In vitro and in vivo Growth Factor Delivery to

124

TUNEL staining (courtesy of Dr. Bodo Kurz, University of Kiel)

Figure A7. TUNEL staining on chondrocytes encapsulated in KLD. Percent chondrocytes stained positive for TUNEL when encapsulated in KLD with or without tethered bIGF-1. Cells were incubated in No IGF-1, IGF-1 (300 ng/mL), or staurosporine (1 µM). Day 3 post encapsulation. Gels were fixed in 10% formalin overnight, paraffin embedded, and shipped to Dr. Bodo Kurz for TUNEL staining. Stats: Mixed model with animal as a random factor, n=18 (3 animals), Tukey post-hoc test, mean ± SEM, * vs No IGF, † vs Staurosporine, p<0.05. References below state that TUNEL is not specific for apoptosis and that results must be confirmed with morphology. Dr. Kurz has done this by electron microscopy and we have determined that the results are more consistent with the flow cytometry data showing no difference with soluble IGF or tethered IGF compared to no IGF gels, indicating the TUNEL staining was not representative of apoptosis in this case. Grasl Kraupp B, et al. In situ detection of fragmented dna (tunel assay) fails to discriminate among apoptosis, necrosis, and autolytic cell death: A cautionary note. Hepatology. 21:1465-1468, 1995. de Torresa C, et al. Identification of necrotic cell death by the TUNEL assay in the hypoxic-ischemic neonatal rat brain. Neuroscience Letters. 230:1-4, 1997. Kelly K, et al. A novel method to determine specificity and sensitivity of the TUNEL reaction in the quantitation of apoptosis. Am J Physiol Cell Physiol. 284:C1309-C1318, 2003.

Page 125: In vitro and in vivo Growth Factor Delivery to

125

Western blots on cells encapsulated in peptide hydrogels Adapted from Diana Chai and Nora Szasz protocols Gel Lysis Protocol Lysis Buffer: 50mM Tris/HCl, pH 7.2 (0.151375g in 25mL) 150mM NaCl (0.21915g in 25mL) 10mM EDTA (0.09306g in 25mL) 1% Triton X-100 (0.25mL) 1% equiv of NP-40 (0.25mL) -Sonicate to dissolve. -Day of add the following to 0.85 mL of lysis buffer:

10 µL Na3VO4 (0.7356 g/20mL dH2O pH 10 for 20 vials of 100x) 100 µL NaF (0.8398 g/20mL dH2O for 20 vials of 10x) 40 µL protease inhibitor cocktail (stock at 25x)

1. Rinse each peptide gel with ice-cold PBS. 2. Put each gel in a large eppendorf tube with 30 µL lysis buffer in it and mechanically break up by pipetting up and down. 3. Sonicate briefly, sit on ice for 15 min. 4. Centrifuge at 10000g for 10 min. 5. Freeze supernatant. Run a BCA assay to determine the total protein content in the gel. Try up to a 1:10 dilution. Western blot Use 15 well (1.5mm thick) Invitrogen bis-tris gel. Choose the % gel and MOPS/MES buffer depending on the MW of your protein of interest. Can run up to 25 µL per well. For each sample: 5 µL – 4x running buffer 2 µL – 10x reducing agent up to 13 µL sample (load equal amounts of total protein, typically ~10-15 µg) - I like to make a stock of 4x running buffer and reducing agent and then add 7 µL to each of my samples. After prepping samples, boil for 5 minutes, spin down, and load into your gel. Run at 200 V for 30-45 min. If apparatus heats up, can surround with ice or run at 4 °C. Transfer for 60 minutes @ 70V (time and voltage may change depending on protein of interest) with stir bar in cold room in transfer buffer without SDS (3.03 g tris base + 14.4 g Glycine, 200 mL methanol). (Can add 0.1% SDS and reduce methanol to 10% if transferring large proteins >80 kDa). Use PVDF membrane in most cases.

Page 126: In vitro and in vivo Growth Factor Delivery to

126

Block for 1 hr in 5% milk+PBST (or TBST) at room temp on shaker plate.

Incubate gel in 1:500 to 1:1000 primary antibody overnight at 4 °C on shaker plate. The following day, wash in PBST 3x10 min at room temp on shaker plate.

Incubate in secondary anti-species of your primary antibody (1:2000) for 1 hr at room temp on shaker plate.

Wash in PBST 3x10 min or longer last rinse if high background is an issue at room temp on shaker plate.

Image with chemiluminescent kit.

Page 127: In vitro and in vivo Growth Factor Delivery to

127

Figure A8. Phospho AKT and phospho ERK Western blots. Chondrocytes were encapsulated in KLD with or without 300 ng/mL tethered IGF-1 (bIGF). Gels were cultured in No IGF-1 (No and bIGF) or 300 ng/mL IGF-1 (IGF) supplemented medium for 1, 2, 3, or 6 days post encapsulation. BMSCs were encapsulated in KLD with 100 ng/mL TGF-β1 and 100 nM dexamethasone adsorbed to the scaffold (SDT+dex) with or without 300 ng/mL tethered IGF-1 (bIGF). Gels were cultured in basal medium for 7 days post encapsulation. At each timepoint, gels were lysed and protein measured by BCA assay. Equal amounts of protein were loaded (5-10 µg) and a Western blot for either (A) pAKT (Cell Signaling #9271) or (B) pERK (Cell Signaling #9101) was performed. Membranes were stripped and reprobed for either AKT (Cell Signaling #9272) or ERK (Cell Signaling #9102).

Page 128: In vitro and in vivo Growth Factor Delivery to

128

Appendix B. Protocols and data related to Chapter 3

Figure B1. Circular dichroism spectroscopy to confirm secondary structure of the peptide and to monitor assembly. Top: Dissolved KLD in 10% sucrose to 0.35 mg/mL. Added 2 or 5 µL of 0.2 N NaOH to 200 µL KLD solution to get pH 7.5 and 9, respectively. As pH increases, the peptide self-assembles. Bottom: 25 µL of PBS was added to 275 µL of 0.35% RAD.

Page 129: In vitro and in vivo Growth Factor Delivery to

129

In situ peptide assembly Made 4 defects, front two were subchondral (5 mm diam), back two were 5 mm diam x 2 mm deep cartilage-only defects with vertical edges.

Figure B2. In situ peptide assembly and cell viability. Used 2 peptide concentrations for subchondral defects, front-most one was 6.3 mg/mL, back one was 3.5 mg/mL. At 5 min, starting to gel but still liquid. At 8 min, the 6.3 mg/mL gel was assembled. At 12 min, the 3.5 mg/mL gel was assembled. (Used 2 µL of Coomassie blue to make the peptide purple in order to see it in the defect.) These gels stayed in the defect even after tilting the joint to fill the other defects. PBS did not wash the gels out. Used 2 peptide concentrations for non-chondral defects, front one was 6.3 mg/mL but we added 100 µL more sucrose to it since starting to clump in falcon tube, ended up looking like original 6.3 mg/mL but actually probably more like 4 mg/mL. Back defect was 3.5 mg/mL. At 8 min, both defects had assembled nicely and did not get disrupted by stream of PBS or by articulation of joint. Chondrocytes in KLD survived this procedure (Lower right). Notes: Blood from subchondral defect did not affect self-assembly process but these were not actively bleeding defects, so this may not reflect the surgical situation. Able to self-assemble KLD in 8 min and flow PBS over gels without displacement.

Page 130: In vitro and in vivo Growth Factor Delivery to

130

Joint Articulation/KLD retention study

Figure B3. In situ filling and joint articulation using 4 mg/mL KLD with Trypan blue. Made 15 mm square defect, 1.5 mm deep on trochlear groove of bovine. Filled to edge of defect. Dripped in PBS to initiate self-assembly. Lubricated joint surface (not defect area) with PBS. Articulated joint 3 times and KLD remained in defect. Additional 3 articulations had no effect on KLD. Upper left, before fill; Upper right: fill; Lower left: after articulations.

Figure B4. In situ filling and joint articulation using 3.2 mg/mL KLD with Trypan blue. Did same as above and peptide stayed in defect, but didn’t seem as sticky as 4 mg/mL. Left: fill; Right: after articulation.

Page 131: In vitro and in vivo Growth Factor Delivery to

131

Other conditions of Rabbit Study Table B1. Treatment groups not included in Chapter 3. Condition n KLD BMSCs TGF-β1 bIGF/dex HA 4 (left) + + 1.4 ng + - 4 (right) 7 + + 0.7 ng + - 5 (left) - + - - + 5 (right) 6 - - - - +

Figure B5. Gross necropsy photographs of joints in Groups 4 and 5.

Page 132: In vitro and in vivo Growth Factor Delivery to

132

Figure B6. Gross and histologic effects comparing defects treated with KLD+BMSCs+1.4 ng TGF-β1 in group 3 to defects treated with the same combination in group 4. Defects in group 3 had contralateral empty defects while defects in group 4 had contralateral defects treated with KLD+BMSCs+0.7 ng TGF-β1.

Page 133: In vitro and in vivo Growth Factor Delivery to

133

Figure B7. Immunohistochemistry scores (0-4). A) Aggrecan. B) Collagen II. C) Collagen I. * p<0.05.

Page 134: In vitro and in vivo Growth Factor Delivery to

134

Results: Radiographic analyses pre- and post-treatment did not reveal any differences among groups 1-3, but comparing treated defects in group 3 to the 100 ng/mL TGF-β1 defects in group 4 showed more radiographic pathology for the group 4 knees (p<0.057) demonstrating higher amounts of lysis, bony proliferation, osteophyte formation and patellar luxation in that group. Upon necropsy, joints in groups 1-3 and 5 appeared normal, while three rabbits that received treatments in both knees (group 4) had mild to severe inflammation. Group 4 also demonstrated bony proliferation along the trochlear ridges and patellar luxation. Those receiving an intra-articular injection of BMSCs in one knee (group 5) also showed mild osteophyte formation, while rabbits with both knees treated (group 4) had mild to moderate osteophyte formation. Comparing group 3 and group 4 defects treated with the same amount of TGF-β1 (100 ng/mL), group 4 revealed significantly worse repair based on total grade (the overall quality of the repair tissue taking into account all observed factors) (p<0.019), color (p<0.019), and synovial membrane (p<0.0496), while incision appearance, inflammation and swelling, and articular surface integrity approached significance (p<0.055). Within group 4, comparing the defect with 50 ng/mL TGF-β1 to the contralateral defect receiving 100 ng/mL TGF-β1, there were no significant differences for any of the scores. Defects receiving an intra-articular injection of BMSCs (group 5) appeared more yellow in color than those receiving HA alone (p<0.028). Similar to gross evaluation, there were no significant differences within group 4. Knees with 100 ng/mL TGF-β1 in group 4 had worse repair tissue than treated defects in group 3 with more hypocellularity (p<0.055) and less repair tissue thickness (p<0.064). There were no significant differences within group 5, although the nature of predominant tissue and reconstitution of subchondral bone scores trended towards a worse repair for the BMSC-treated defects (p<0.07). Comparing group 3 treated defects to group 4 defects with 100 ng/mL TGF-β1, collagen II scoring was not significant, but trended (p<0.1) towards higher scores for group 3. Within groups 4 and 5 there were no significant differences. Discussion: In addition, treating two knees in one rabbit with BMSCs and different doses of TGF-β1 (group 4) resulted in an increased inflammatory response and bony reaction, as seen by the comparison between this group and the treated defects in group 3 in which only one knee received TGF-β1. This finding suggests possible systemic effects of treatment and that negative effects were caused by increasing the total body dose of TGF-β1 (2.25 ng total for rabbits receiving two treatments and 1.5 ng for those with one treatment). Finally, treating defects with an intra-articular injection of BMSCs in HA (group 5) offered no advantage over injection of HA alone.

Page 135: In vitro and in vivo Growth Factor Delivery to

135

Appendix C. Protocols and data related to Chapter 4 Western blot on medium following enzyme treatment of cartilage explants HS stub antibody detects cleavage following heparatinase treatment. Use 4-12% bis-tris 15 well (1.5mm thick) Invitrogen mini-gel. Used Novex sharp protein standard from Invitrogen. Can run up to 25 µL per well. 5 µL – 4x running buffer 2 µL – 10x reducing agent 13 µL conditioned medium Boil all samples for 5 min. Ran Gel in MOPS running buffer at 200V for 45-60 min Transferred for 80 minutes @ 75V with stir bar in cold room in transfer buffer with 0.1% SDS (1 g SDS) and 10% methanol (3.03 g tris base + 14.4 g Glycine, 100 mL methanol). Used PVDF membrane. Antibody Incubation: Blocked for 1 hour in 5% milk+PBST. Incubated gel 1:500 with 3G10ab (Seikagaku) O/N @ 4 °C . Following day, washed in PBST 3x10 min. Incubated in secondary: 1:1000 Anti-mouse 60 minutes @ RT. Wash in PBST 3x10 minutes Imaged using chemiluminescence kit.

Page 136: In vitro and in vivo Growth Factor Delivery to

136

Figure C1. Western blots of conditioned medium following no enzyme (N), heparitinase (H), chondroitinase (C), or chondroitinase followed by heparitinase (C+H) treatment of cartilage explants. Used anti-HS stub (3G10ab) antibody.

Page 137: In vitro and in vivo Growth Factor Delivery to

137

Biotinylation of GAGs Adapted protocol from: Ute Friedrich, Anna M. Blom, Björn Dahlbäck and Bruno O. Villoutreix. Structural and Energetic Characteristics of the Heparin-binding Site in Antithrombotic Protein C. J Biol Chem. 276:24122-24128, 2001. Biotin hydrazide biotinylates GAGs on carboxyl groups. I first attempted to use Sulfo-NHS-LC-Biotin, which normally biotinylates at the primary amine of proteins, but this didn’t seem to work for the GAGs even though others have reported doing it this way. Pierce EZ-Link Biotin-LC-Hydrazide kit: #21340 Pierce Zeba Desalt Spin Columns: #89891 Heparin: Sigma, #H3149 Heparan sulfate: Sigma, #H7640 Chondroitin sulfate: Sigma, #C4384 1. Dissolved GAG in 0.1 M MES at 2.5 mg/mL. Used 200 µL (0.5 mg). 2. Added 12.5 µL of 50 mM biotin hydrazide. (at least 1 mg biotin in 54 µL DMSO). 3. Added 0.5 µL of 0.5 M EDC. (at least 5 mg in 50 µL MES). 4. Mix with constant shaking (taped to rotary shaker plate and put on high) O/N at room temperature. 5. Desalt using spin columns and equilibrate in water. I would use 2 different columns to make sure you get rid of all the unincorporated biotin. I only used one column and one of the times I did this it seemed like I had a lot of free biotin left in my sample when I was finished. You can store at 4 °C for at least several weeks. An alternative would be to lyophilize and store in -80 °C for long-term storage. 6. Confirm biotinylation using HABA/Avidin assay or dot blot using anti-biotin. Sandwich ELISA (biotinylated GAGs attached to streptavidin-coated ELISA plate; HBIGF/XIGF in solution; amount bound detected by anti-IGF-1) Used KPL ELISA kit (54-62-15) 1. Use pre-coated R&D streptavidin plate (CP003). Coat ELISA plates with 100 µL of 20 µg/mL biotinylated GAGs in coating buffer. Incubate overnight at 4 °C. Need 6.5 mL coating buffer. 2. Block with kit for 15min

1. Make BSA diluent for rest of steps. 2. Add 300 µL BSA Diluent/Blocking Solution to each well. 3. Incubate 15 minutes at room temp, empty plate and tap out residual liquid.

3. Incubate with varying amounts of HBIGF. 1. Add 100 µL Standard/Sample to each well. (do 2 wells per sample)

2. React at room temperature for 1 hour. 3. Empty plate, tap out residual liquid.

4. Wash plates.

Page 138: In vitro and in vivo Growth Factor Delivery to

138

1. Fill each well with 1X Wash Solution. 2. Invert plate to empty, tap out residual liquid. 3. Repeat 3 - 5 times.

5. Incubate with anti-IGF for 1 h at 10 µg/mL at room temp.

1. Add 100 µL Detection Antibody Solution to each well. (anti-IGF at 10 µg/ml) 2. React 1 hour, room temperature. 3. Empty plate, tap out residual liquid and wash as above.

6. Wash plates. 7. Incubate with anti-rabbit HRP for 1 h at 1:500 at room temp.

1. Add 100 µL Secondary Antibody Solution to each well. 2. React 1 hour, room temperature, in dark. 3. Empty plate, tap out residual liquid and wash as above. 4. Give final 5 minute soak in Wash Solution; tap residual liquid from plate.

8. Wash plates. 9. Read Substrate

1. Dispense 100 µL Substrate Solution into each well. 2. After sufficient color development (read every 1 min), add 100 µL

Stop Solution to each well. 3. Read plate with plate reader at 405 nm.

Page 139: In vitro and in vivo Growth Factor Delivery to

139

Surface Plasmon Resonance (Biacore) Protocols Conditioning Buffer: 1M NaCl in 50 mM NaOH. Running Buffer: 0.01M HEPES pH 7.4 -> 1 mL in 100 mL 0.15M NaCl -> 0.8766 g 3mM EDTA -> 0.111672 g 0.005% Tween20 -> 5 µL Filter and Degas. Regeneration Buffer: 1M NaCl in PBS, pH 7.4 Start-up Protocol: Load chip. Put in Running Buffer. Dock. Prime, ignore errors. Shutdown Protocol: Undock. Take out chip, store in 50 mL conical with 2 mL water in bottom at 4 °C. Put in maintenance chip. Dock. Put tubes in water instead of running buffer. Prime into water (7 min). Fill 2 vials with 3 mL SDS (1) or 3 mL glycine (2). Put in rack. Run desorb (22 min). Prime water 2x. Take out water. Put in kim wipe. Prime with air. Undock. Take out maintenance chip. First run, attach GAGs to chip: Run sensorgram. Fc-1-2-3-4 measure all 4. Switch to Fc-4. Flow 20 µL/min. NaOH 3x 1min. Wash needle 2x. Manual inject. Load bHeparin, 26 µL in F4, saturated at 10 µL. Switch to Fc-3. Quick inject 20 µL NaOH 3x. Wash needle 2x. Switch to 5 µL/min. Load bHS, saturated in few µL. Switch to Fc-2. Flow 20 µL/min NaOH 3x1min. Wash needle 2x. Flow 10 µL/min. Manual inject. Load bCS, saturated at 5 µL. Switch to Fc-1.

Page 140: In vitro and in vivo Growth Factor Delivery to

140

Flow 20 µL/min. Quick inject 20 µL NaOH 3x. Wash needle 2x. Put 1 µM (90 µL) HBIGF. Switch to Fc-1-2-3-4. 5 µL/min. KinInject 25 µL = 300 s. Dissoc 300 s. Put in 0.1M NaCl in PBS. Change to 20 µL/min. Quick inject 20 µL x2. Wash needle 2x. Switch to 5 µL/min. Put in 100 nM (70 µL) HBIGF. Kininject 25 µL = 300 s. Dissoc 300 s. 20 µL/min. Quick inject 0.1M NaCl in PBS. 20 µL 1x. To acquire kinetic data: Docked chip. Primed into RB. Run 1: Flow rate 20 µL/min. Run with RB. Quick Inj 1M NaCl in PBS. Run with RB. Run 2: 100 nM fresh HBIGF at 20 µL/min for 300 s. Dissociation 300 s. Quick Inj 2x with 1M NaCl 20 µL. Wash needle 2x. Run 3: 250 nM HBIGF Etc.

Page 141: In vitro and in vivo Growth Factor Delivery to

141

Densitometry (semi-quantitative analysis of Western blots) with FluorChem software program: Hit cancel when it asks for a serial number.

Make sure your file name is short or the file will not open. On the contrast enhancement panel, hit reverse and auto contrast to view your gel. You

can further adjust the black, white, and gamma channels to improve your view. Doing this does not affect the pixels the software program uses to calculate density.

On the toolbox panel, choose the Analysis Tools tab. Then choose the Spot Denso tab. Make sure that if you select a band and select background that the background has a

lower value. If this is not the case, click the invert button. In most cases, when you are starting with a black gel with white bands, you should not have to select invert.

Use the object buttons to outline each band you wish to analyze separately. If you have different amounts of background on your gel, also draw small boxes under

each band of interest using the background buttons. Then select the background box and corresponding band box while holding the shift key. Hit link bkgd. This should change both boxes to the same color.

To copy your data, click output and choose copy to clipboard.

Paste data into excel. The average column = IDV/area. If your boxes are different sizes, you should use the

IDV column to analyze your data. In order to average data from multiple gels, you must have a common condition you can

normalize to on each gel.

Page 142: In vitro and in vivo Growth Factor Delivery to

142

Culture of CHO (Chinese Hamster Ovary) Cells Adapted from Robin Prince of Rich Lee’s lab CHO cells grow in F-12 media supplemented with 10% FBS. 677 cells do not produce heparan sulfate and appear more boxy compared to the CHOK1 (WT) cells which look more spindle shaped. Starting culture Thaw cells in 37 oC water bath. – Do this fast (2 min or less) Put one vial in 10 cm dish with 10 mL warmed F-12 + 10% FBS media. Change medium next morning. It may take several days for the cells to become confluent. (Because these cell lines are very stable it is not necessary to maintain them in drug-containing media). Splitting confluent cells Warm pbs, medium, and trypsin to 37 °C. Aspirate off the media from a 10 cm dish of cells. Add 2 mL trypsin and let incubate for 5 min at 37 °C. Add 8 mL medium to stop trypsin, pipette up and down to de-clump. Spin down cells at 125g for 5 min. Resuspend in 10 mL medium, pipet up and down. Typically split 1:4 or 1:5, can go up to 1:8 safely. Freezing cells You should do this during the first passage to keep your stock. You can take half the confluent cells in a 10 cm dish to freeze and replate the rest depending on your needs. Rinse 10 cm dish with PBS. Put in 2 mL trypsin (37 °C for 5 min). Add 8 mL F12+10% FBS to stop. Spin down, resuspend in 10 mL F12+10% FBS. Count undiluted and should get ~6 million cells. Take 5 mL (3 million cells) to freeze down and put in a separate tube. Spin that down and resuspend in 0.6 mL (5 million cells/mL) of F12+20%FBS+10%DMSO. Put in cryovial and put in Mr. Frosty in -80 °C overnight. Transfer to liquid nitrogen storage.

Page 143: In vitro and in vivo Growth Factor Delivery to

143

Appendix D. Luminex Pilot Study Adapted from Megan McBee and Arek Raczynski of David Schauer’s lab Joint Extraction on Mice Materials: We used the Biorad Lysis buffer kit. 1. 500 mM AEBSF (12 mg/100 µL water). Make 100 µL fresh. 2. Lysing Solution (from Biorad kit) = 40 µL Factor 1 + 20 µL Factor 2 + 9.9 mL Cell Lysis Buffer. Vortex gently. Add 40 µL 500 mM AEBSF per 10 mL Lysing Solution. Make 10 mL (for 20 samples) 3. Diluent = 0.05% Tween-20, 0.1% BSA in PBS. Make 50 mL fresh Methods: 1. The joints were harvested by careful dissection to remove all attached tissues (particularly muscle). The "whole joint" preparation includes bone ends, patellar without proximal muscle, and all intra-articular structures (cartilage, cruciates, synovium, meniscus etc) and fluid. This preparation was flash frozen and then pulverized as a single sample. If possible, weigh the amount of tissue you collect prior to flash freezing. 2. Pulverize by 12 hammer blows. 3. Recover powder to tube and immediately add 350 µL (record known volume) of "Lysing Solution" (Reagent 2 above) to sample. Parafilm top of closed tubes to prevent any possible leakage. Rotate samples for 1-2 h at 4 °C for extraction. 4. Centrifuge samples at 4500g for 20 min at 4 °C. 5. Collect supernatant without disturbing pellet and remove clear supernatant. Weigh how much you collect. 6. Store at -80 °C. Before doing Luminex Assay: 1. Thaw samples and perform BCA assay. Refreeze. (We used this to normalize and to get an idea of how much sample to load.) Luminex Assay: 1. We used the 23-plex mouse cytokine pro kit which includes magnetic beads to be used in combination with a magnetic plate washer. It comes with very detailed instructions which we followed. We used the same lysis buffer to dilute samples and make up standard curves as explained a bit more below. 2. From the BCA assay our samples were in the range of 2.3-4.7 mg/mL which is a little on the high side of what to load, but since this was our first try with this we used either 50 uL straight sample in duplicates or 25 uL sample + 25 uL lysis buffer in duplicates. We also used both the high and low PMT standard curves for the same reason that we didn’t know what to expect. To use both of the standard curves you have to run the plate twice, but we didn’t notice any issues with doing this.

Page 144: In vitro and in vivo Growth Factor Delivery to

144

In the end, the data we used came from using the undiluted samples and the high PMT curves. 3. To make sure that all of the standards and samples were incubated with the beads for about the same amount of time, we pipetted everything into a separate 96 well plate and then used a multichannel pipet to load the samples/stds onto the actual plate. This will make more sense when you read the biorad protocol. 4. Also, you should change tips for everything as the assay is very sensitive. 5. Finally, we used n=3 since this was a pilot study, but this was probably too low to get significance on some of the cytokines.

Figure D1. Methods: IACUC approval was given by Rush University. Ten week old C57Bl/6 male mice were subjected to a surgical Destabilization-Induced Joint Injury (DMM) model developed by Glasson et al. alone, to treadmill (TM) running alone (14 days 15° incline, 32 cm/sec, 20 min per day, for 2 weeks), or to DMM surgery followed by TM running beginning on day 4 post-surgery. For DMM surgery, the anterior medial meniscotibial ligament was completely severed. Control mice were subjected to only cage activity. At 3 weeks post surgery, mice were euthanized and joints were stained with India Ink for gross observations or protein was extracted for Luminex (23-plex mouse cytokine kit), n=3. Statistics: Data were log-transformed and analyzed by ANOVA with Tukey post-hoc test, p<0.05. Luminex data were normalized to the total protein extracted per joint and to the control mice.