investigations on keloid pathogenesis and therapy · interactions in keloid scar pathogenesis and...

168
i INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY Anandaroop Mukhopadhyay (B.Pharm), The Tamilnadu Dr M.G.R Medical University, INDIA) THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF PHARMACY NATIONAL UNIVERSITY OF SINGAPORE 2007

Upload: others

Post on 07-Aug-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

i

INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY

Anandaroop Mukhopadhyay (B.Pharm), The Tamilnadu Dr M.G.R Medical University, INDIA)

THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

DEPARTMENT OF PHARMACY

NATIONAL UNIVERSITY OF SINGAPORE

2007

Page 2: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

ii

ACKNOWLEDGEMENT

It is my pleasure to thank the many people who made this thesis possible.

To start with, I would like to thank my Ph.D. supervisor Associate Professor Chan Sui

Yung for her excellent guidance and support. Had it not been for her, I wouldn’t have had

the courage to explore new horizons. Her critical evaluation of my research and constant

encouragement invigorated me to push the limits.

It is difficult to overstate my gratitude to my co-supervisor Assistant Professor Phan Toan

Thang. He took me in his research team knowing fully well that I had no experience in

skin biology. I thank him for having confidence in me and providing me the opportunity

to be a part of the cutting edge research in skin pathogenesis in his laboratory. With his

enthusiasm, his inspiration, and his great efforts to explain things clearly and simply, he

helped to make research fun for me. Throughout the period of writing my thesis, he

provided encouragement, sound advice, good teaching, good company, and lots of good

ideas. I would have been lost without him.

I am deeply indebted and thankful to my parents Subhash Chandra Mukhopadhyay and

Gopa Mukhopadhyay for instilling in me, values, that helped me comprehend the

importance of knowledge and pursue it. My brothers Abhiroop Mukhopadhyay and

Ritobrata Banerjee have always been my mentors and I am thankful to them for their

invaluable advice. I would also like to thank my uncle Dr M.G Banerjee for always

believing in me, and the rest of my family members for their support. Finally I am

grateful to all my friends and lab mates for making my graduate life enjoyable.

Page 3: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

iii

TABLE OF CONTENTS

Acknowledgement

List of Publications

List of Figures

Abbreviations

Summary

1 BACKGROUND AND INTRODUCTION........................................................................................4 1.1 OVERVIEW OF THE WOUND HEALING PROCESS..............................................................5 1.2 KELOID SCAR............................................................................................................................7

1.2.1 Clinical Characteristics of Keloid ...........................................................................................7 1.2.2 Epidemiology...........................................................................................................................9 1.2.3 Histopathology ........................................................................................................................9 1.2.4 Molecular Pathogenesis ........................................................................................................11 1.2.5 Current Treatment .................................................................................................................16

1.3 OBJECTIVES OF THE PRESENT STUDY..............................................................................19

2 MATERIALS AND METHODS ......................................................................................................21 2.1 MEDIA AND CHEMICALS .....................................................................................................22 2.2 RECOMBINANT GROWTH FACTORS AND ANTIBODIES................................................22 2.3 PREPARATION OF NORMAL AND KELOID TISSUE EXTRACTS..............23 2.4 IMMUNOHISTOCHEMISTRY ................................................................................................24

2.4.1 Preparation of Paraffin Sections of Normal and Keloid Tissues...........................................24 2.4.2 Pretreatment of Paraffin Sections for Immunohistochemistry...............................................24 2.4.3 Probing with Antibody and Developing ................................................................................25

2.5 CELL CULTURE.......................................................................................................................25 2.5.1 Keloid Keratinocyte and Fibroblast Database......................................................................25 2.5.2 Keloid and Normal Keratinocytes from Keloid scar and Normal Skin .................................26 2.5.3 Keloid and Normal Fibroblast from Keloid Scar and Normal Skin ......................................26 2.5.4 Keratinocyte-fibroblast Coculture.........................................................................................27

2.6 CELL COUNTING ....................................................................................................................29 2.7 TREATMENT OF FIBROBLASTS WITH GROWTH FACTOR.............................................29 2.8 TREATMENT OF CELLS WITH GLEEVEC...........................................................................29 2.9 SERUM STIMULATION..........................................................................................................30 2.10 MTT ASSAY .............................................................................................................................30 2.11 FIBROBLAST-POPULATED COLLAGEN LATTICE (FPCL) ..............................................31

2.11.1 Preparation of Fibroblast-Populated Collagen Lattices ..................................................31 2.11.2 Macroscopic Evaluation of FPCL Contraction ................................................................32

2.12 WESTERN BLOTTING ............................................................................................................32 2.13 IMMUNOASSAY......................................................................................................................33

Page 4: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

iv

2.14 FLOW CYTOMETRY...............................................................................................................33 2.15 RNASE PROTECTION ASSAY.................................................................................................34 2.16 RATE OF ATP SYNTHESIS.....................................................................................................34 2.17 MEASUREMENT OF INTRACELLULAR ATP......................................................................35 2.18 STATISTICAL ANALYSIS ......................................................................................................35

3 ROLE OF EPITHELIAL-MESENCHYMAL INTERACTIONS IN WOUND CONTRACTION AND SCAR CONTRACTURE ..................................................................................................................36

3.1 INTRODUCTION......................................................................................................................37 3.2 RESULTS ..................................................................................................................................39 3.3 DISCUSSION ............................................................................................................................46

4 ROLE OF ACTIVIN-FOLLISTATIN SYSTEM IN KELOID PATHOGENESIS.....................50 4.1 INTRODUCTION......................................................................................................................51 4.2 RESULTS ..................................................................................................................................53 4.3 DISCUSSION ............................................................................................................................67

5 ROLE OF PROTEOGLYCANS IN KELOID PATHOGENESIS................................................73 5.1 INTRODUCTION......................................................................................................................74 5.2 RESULTS ..................................................................................................................................77 5.3 DISCUSSION ............................................................................................................................94

6 ROLE OF STEM CELL FACTOR AND C-KIT SYSTEM IN KELOID PATHOGENESIS..101 6.1 INTRODUCTION....................................................................................................................102 6.2 RESULTS ................................................................................................................................104 6.3 DISCUSSION ..........................................................................................................................117

7 INVESTIGATION OF GLEEVEC AS A THERAPEUTIC AGENT FOR KELOID SCARS 124 7.1 INTRODUCTION....................................................................................................................125 7.2 RESULTS ................................................................................................................................127 7.3 DISCUSSION ..........................................................................................................................134

General Conclusion

Appendix 1

Page 5: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

v

LIST OF PUBLICATIONS

Publication Papers

1. Mukhopadhyay A, Tan EK, Khoo YT, Chan SY, Lim IJ, Phan TT. Conditioned medium

from keloid keratinocyte/keloid fibroblast coculture induces contraction of fibroblast-

populated collagen lattices. Br J Dermatol 2005 Apr; 152(4):639-45.

2. Phan TT, Lim IJ, Aalami O, Lorget F, Khoo A, Tan EK, Mukhopadhyay A, Longaker

MT Smad3 signaling plays an important role in keloid pathogenesis via epithelial-

mesenchymal interactions. J Pathol 2005 Oct; 207(2):232-42.

3. Khoo A, Ong CT, Tan EK, Mukhopadhyay A, IJ Lim, TT Phan. The role of Connective

Tissue Growth Factor (CTGF) in the biology of epithelial-mesenchymal interactions of

keloid pathogenesis. J Cell Physiol 2006 Aug; 208(2):336-43

4. Mukhopadhyay A, Chan SY, Lim IJ, Phillips DJ, Phan TT. The role of Activin System in

Keloid Pathogenesis. Am J Physiol-Cell Physiol 2006 Sep 13(in press)

5. Ong CT, Khoo A, Tan EK, Mukhopadhyay A, Do DV, Han CH, IJ Lim, TT Phan.

Epithelial-mesenchymal interactions in keloid pathogenesis modulate vascular

endothelial growth factor expression and secretion. J Pathol 2007; 211:95-108

6. Do DV, Mukhopadhyay A, Lim IJ, Phan TT. Roles of epithelial-mesenchymal

interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal

Transduction Therapy (invited review paper, in press).

7. Mukhopadhyay A, Fan S, Do DV, Khoo A, Ong CT, Lim IJ, Phan TT. Role of HGF /c-

Met system in keloid pathogenesis. Br J Dermatol, (in press).

Page 6: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

vi

Submitted manuscripts

1. Mukhopadhyay A, Khoo A, Chan SY, Aalami O, Lim IJ, Phan TT. Specific Target of

Sp1 Transcription Factor: A Novel Therapeutic Approach for Keloids.

2. Mukhopadhyay A, Wong MY, Chan SY , Do DV , Khoo A , Ong CT , Cheong HH , Lim

IJ , Phan TT. Syndecan-2 and Decorin - Proteoglycans with a difference: Implications in

keloid pathogenesis.

Presentations

1. NHG Annual Scientific Congress, Singapore, October 2004

2. AAPS (American Association of Pharmaceutical Scientists) Annual Meeting and

Exposition, Baltimore, USA, November 2004

3. 17th Pharmacy Congress and Intervarsity Symposium, Singapore, July 2005

4. 3rd Meeting of the WHSS (Wound Healing Society of Singapore), Singapore, August

2005

5. Inaugural AAPS-NUS Student Chapter Symposium, Singapore, September 2005

6. Controlled Release Society Conference, Vienna, Austria, July 2006

7. Biostar Congress, Stuttgart, Germany, October 2006

Page 7: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

vii

LIST OF FIGURES

FIGURE 1: SCHEMATIC REPRESENTATION OF DIFFERENT STAGES OF WOUND REPAIR (ADAPTED FROM

WERNER ET AL., 2003). ...........................................................................................................................6

FIGURE 2: KELOID FORMATION IN DIFFERENT PATIENTS (ADAPTED FROM MARNEROS ET AL., 2004)...............8

FIGURE 3: H&E STAINING OF NORMAL SKIN AND KELOID SCAR TISSUE .........................................................10

FIGURE 4: SCHEMATIC REPRESENTATION OF PATHWAYS POTENTIALLY RESULTING IN ACCUMULATION OF

COLLAGEN IN FIBROTIC SKIN DISEASES (ADAPTED FROM UITTO ET AL., 2007)......................................16

FIGURE 5: COCULTURE OF EPIDERMAL KERATINOCYTES AND DERMAL FIBROBLASTS AS AN IN VITRO MODEL

TO STUDY EPITHELIAL-MESENCHYMAL INTERACTION.....................ERROR! BOOKMARK NOT DEFINED.

FIGURE 6: EFFECT OF CONDITIONED MEDIA COLLECTED FROM KELOID KERATINOCYTE (KK)/KELOID

FIBROBLAST (KF) CO CULTURES ON COLLAGEN CONTRACTION BY KFS ...............................................42

FIGURE 7: COMPARISON OF CONTRACTION OF COLLAGEN GEL LATTICE INCORPORATED WITH KELOID

FIBROBLASTS (KFS) AND NORMAL FIBROBLASTS (NFS). ......................................................................43

FIGURE 8: Α-SMOOTH MUSCLE ACTIN (Α-SMA) EXPRESSION BY KELOID FIBROBLASTS (KFS) (A) OR NORMAL

DERMAL FIBROBLASTS (NFS) (B). .........................................................................................................44

FIGURE 9: EFFECT OF ANTI TRANSFORMING GROWTH FACTOR (TGF)-Β1 NEUTRALIZING ANTIBODY ON

COLLAGEN CONTRACTION BY KELOID FIBROBLAST (KF) STRAIN 48 (KF48) INDUCED BY CONDITIONED

MEDIA COLLECTED FROM KELOID KERATINOCYTE (KK) STRAIN 48 (KK48)/KF48 COCULTURE. .........45

FIGURE 10: INCREASED LOCALIZATION OF ACTIVIN-A AND FOLLISTATIN IN THE BASAL LAYER OF EPIDERMIS

IN NORMAL AND KELOID TISSUES..........................................................................................................57

FIGURE 11: EXPRESSION OF ENDOGENOUS ACTIVIN-ΒA MRNA DERIVED FROM NORMAL AND KELOID TISSUES.

.............................................................................................................................................................58

FIGURE 12: ELEVATED LEVELS OF ACTIVIN-A OBTAINED FROM KELOID FIBROBLAST CULTURES...................59

FIGURE 13: INCREASED PROLIFERATION OF NORMAL FIBROBLASTS WHEN COCULTURED WITH ACTIVIN-A

OVEREXPRESSING HΒAHACAT CELLS. .................................................................................................60

Page 8: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

viii

FIGURE 14: INCREASED PROLIFERATION OF NORMAL AND KELOID FIBROBLASTS TREATED WITH RHACTIVIN-

A. .........................................................................................................................................................61

FIGURE 15: EFFECT OF ACTIVIN-A, FOLLISTATIN OR TGF-Β1 ON COLLAGEN, FIBRONECTIN AND Α-SMA

EXPRESSION..........................................................................................................................................63

FIGURE 16: ACTIVIN-A EXPRESSION INCREASED BY EPITHELIAL-MESENCHYMAL INTERACTIONS RESULTING

IN A FIBROTIC PHENOTYPE. ...................................................................................................................72

FIGURE 17: ELEVATED LEVELS OF SYNDECAN-2 IN TISSUE EXTRACTS OBTAINED FROM KELOID TISSUE. .......81

FIGURE 18: SERUM GROWTH FACTORS UPREGULATED SYNDECAN-2 EXPRESSION IN NORMAL AND KELOID

FIBROBLASTS. .......................................................................................................................................82

FIGURE 19: INCREASED ECTODOMAIN SHEDDING OF SYNDECAN-2 IS OBSERVED IN KELOID COCULTURES AS

COMPARED TO MONOCULTURES............................................................................................................83

FIGURE 20: EXOGENOUS RHFGF-2 STIMULATES SHEDDING OF SYNDECAN-2 FROM FIBROBLAST CELL

SURFACE INTO THE CONDITIONED MEDIA..............................................................................................84

FIGURE 21: INCREASED LOCALIZATION OF FGF-2 IN THE BASAL LAYER OF EPIDERMIS AND DERMIS IN KELOID

TISSUE. .................................................................................................................................................85

FIGURE 22: ELEVATED LEVELS OF FGF-2 IN TISSUE EXTRACTS OBTAINED FROM KELOID TISSUE. .................86

FIGURE 23: COCULTURED NORMAL AND KELOID KERATINOCYTES EXPRESS INCREASED LEVELS OF FGF-2 AS

COMPARED TO MONOCULTURED KERATINOCYTES. ...............................................................................87

FIGURE 24: COCULTURED NORMAL AND KELOID FIBROBLASTS EXPRESS INCREASED LEVELS OF FGF-2 AS

COMPARED TO CELLS IN MONOCULTURE...............................................................................................88

FIGURE 25: DOWNREGULATION OF DECORIN IN TISSUE EXTRACTS OBTAINED FROM KELOID TISSUE..............89

FIGURE 26: SERUM GROWTH FACTORS DOWNREGULATE DECORIN EXPRESSION IN NORMAL AND KELOID

FIBROBLASTS. .......................................................................................................................................90

FIGURE 27: INCREASED SECRETORY DECORIN IN COCULTURED NORMAL FIBROBLASTS AND NOT KELOID

FIBROBLASTS. .......................................................................................................................................91

FIGURE 28: EFFECT OF DECORIN ON COLLAGEN, FIBRONECTIN AND Α-SMA EXPRESSION..............................92

FIGURE 29: ECTODOMAIN SHEDDING OF SYNDECAN-2 BY FGF-2 STIMULATED BY EPITHELIAL-MESENCHYMAL

INTERACTIONS RESULTING IN A FIBROTIC PHENOTYPE........................................................................100

Page 9: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

ix

FIGURE 30: ELEVATED LEVELS OF SCF IN TISSUE EXTRACTS OBTAINED FROM KELOID TISSUE. ...................107

FIGURE 31: SERUM GROWTH FACTORS UPREGULATED SCF EXPRESSION IN NORMAL AND KELOID

FIBROBLASTS. .....................................................................................................................................108

FIGURE 32: INCREASED LEVELS OF SCF IN CONDITIONED MEDIA OBTAINED FROM KELOID COCULTURES AS

COMPARED TO MONOCULTURES..........................................................................................................109

FIGURE 33: ELEVATED LEVELS OF C-KIT AND PHOSPHO C-KIT (TYR 703, TYR 721) IN TISSUE EXTRACTS

OBTAINED FROM KELOID TISSUE. ........................................................................................................110

FIGURE 34: INCREASED LOCALIZATION OF C-KIT IN THE BASAL LAYER OF EPIDERMIS OF KELOID TISSUE. ..111

FIGURE 35: C-KIT UPREGULATED IN KELOID KERATINOCYTES AS COMPARED TO NORMAL KERATINOCYTES.

...........................................................................................................................................................112

FIGURE 36: INCREASED ECTODOMAIN SHEDDING OF C-KIT IN KELOID KERATINOCYTE/KELOID FIBROBLAST

COCULTURES. .....................................................................................................................................112

FIGURE 37: TACE OVEREXPRESSED IN KELOID SCAR AS COMPARED TO NORMAL SKIN................................115

FIGURE 38: UPREGULATION OF TACE IN KELOID FIBROBLAST COCULTURES AS COMPARED TO NORMAL

COCULTURES. .....................................................................................................................................116

FIGURE 39: C-KIT SYSTEM IN KELOID PATHOGENESIS..................................................................................122

FIGURE 40: NO SIGNIFICANT DIFFERENCE IN THE PROLIFERATIVE POTENTIAL OF KELOID KERATINOCYTES

AND KELOID FIBROBLASTS..................................................................................................................129

FIGURE 41: GLEEVEC DOWNREGULATES EXPRESSION OF PHOSPHO AKT, PHOSPHO MTOR, PHOSPHO C-KIT

(TYR 721). ..........................................................................................................................................130

FIGURE 42: GLEEVEC DOWNREGULATES EXPRESSION OF COLLAGEN, FIBRONECTIN, Α-SMA, VEGF, HDGF,

TGF-Β1, SCF, FGF-2. ........................................................................................................................131

FIGURE 43: GLEEVEC SIGNIFICANTLY REDUCES THE CONTRACTION OF COLLAGEN GEL LATTICE

INCORPORATED WITH KELOID FIBROBLASTS. ......................................................................................132

FIGURE 44: GLEEVEC SIGNIFICANTLY REDUCES THE INTRACELLULAR ATP AND THE RATE OF ATP SYNTHESIS

IN KELOID FIBROBLASTS. ....................................................................................................................133

FIGURE 45: SUMMARY OF VARIOUS GROWTH FACTORS INVOLVED IN KELOID PATHOGENESIS AND THE

POSSIBLE THERAPEUTIC TARGETS. ......................................................................................................139

Page 10: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

x

ABBREVIATIONS

ALK

Activin receptor – like kinase

BMP

Bone morphogenetic protein

CTGF Connective tissue growth factor

DMEM

Dulbecco’s modified Eagle medium

ECM

Extracellular matrix

EGF

Epidermal growth factor

ELISA

Enzyme-Linked Immunosorbent Assay

ERK

Extraregulated Kinase

FCS

Fetal calf serum

FGF-2

Basic fibroblast growth factor

FPCL

Fibroblast populated collagen lattice

FPD Fibroproliferative disorder

GADPH

glyceraldehyde-3-phosphate dehydrogenase

HBSS

Hank’s balanced salt solution

HDGF Hepatocyte derived growth factor

HSPG

Heparan Sulfate Proteoglycan

IgG

Immunoglobulin G

IgM

Immunoglobulin M

Page 11: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

xi

JNK

Jun-Amino terminal-Kinase

KF

Keloid fibroblasts

KGM

Keratinocyte growth medium

KK

Keloid keratinocytes

MAPK

Mitogen-Activated-Protein-Kinase

mTOR Mammalian target of rapamycin

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

NF

Normal fibroblasts

NK

Normal keratinocytes

PBS

Phosphate buffered saline

SCF Stem cell factor

TGF-β1

Transforming growth factor-beta1

VEGF

Vascular endothelial growth factor

α-SMA

Alpha-Smooth Muscle Actin

Page 12: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

1

SUMMARY

Keloid scars are a result of such an aberration in wound healing, and are characterized by

overabundant collagen/ECM deposition. Several treatment modalities have been explored

but none of the treatments are effective. In spite of the large number of growth factors,

cytokines and chemokines, that have been studied and are now known to play a role in its

pathogenesis, the precise pathobiology leading to this scar formation remains largely

unknown. Thus to identify new targets for therapeutic intervention, it is imperative to

identify key modulators which are aberrantly expressed in keloids and to understand and

assess the role of these modulators towards this fibrotic phenotype.

This study aims to identify key modulators of cellular dynamics in keloid scars and

investigate the effect of epithelial-mesenchymal interactions on the expression profile of

these modulators using various in vitro models. This study further explores different

growth factors or chemical drugs as potential therapeutic agents for keloid scars.

Bearing in mind the importance of wound contraction and scar contracture in the wound

healing process and scar formation respectively, an in vitro coculture model and a

fibroblast-populated collagen lattice was employed to study the role of epithelial-

mesenchymal interaction towards a contractile phenotype. It was observed that

epithelial-mesenchymal interactions increased the contractile response of both normal

and Keloid fibroblasts in vitro. In addition KF’s were shown to have an elevated

contracile response to signals from the extracellular envirionment than normal

fibroblasts, due to the increased expression of α-SMA, a contractile protein.

Page 13: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

2

Further studies were performed to investigate the molecular effectors of keloid scars and

the effect of epithelial-mesenchymal interaction on the expression profile of these

effectors.

Activin-A, a dimeric protein and a member of the TGF-β superfamily, has been shown to

regulate various aspects of cell growth and differentiation in the repair of the skin

mesenchyme and the epidermis. Thus the study aimed at investigating the role of activin

and its antagonist, follistatin, in keloid pathogenesis. The findings strongly suggest that

activin-A is a potent inducer of fibroblast activation and involved in the pathogenesis of

keloids. They also emphasize the importance of follistatin in regulating activin-A

bioactivity and suggest a possible therapeutic potential of follistatin in the treatment and

prevention of keloids.

Most of the growth factors and cytokines involved in the wound healing process seem to

be immobilized at the cell surface and extracellular matrix via binding with

proteoglycans, making them important modulators of cell dynamics. Thus the expression

of two proteoglycans, namely syndecan-2 and decorin, were investigated in order to

elucidate their roles in keloid pathogenesis.

It was demonstrated that syndecan-2 and FGF-2 were not only overexpressed in keloid

tissues but could also interact with each other resulting in the shedding of syndecan-2

which could in turn activate a whole cascade of events resulting in keloidogenesis. In

addition, decorin appeared to be downregulated in keloid tissues, and this protein with

strong antifibrotic effects could have the potential to be used as a therapeutic agent for

keloids.

Page 14: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

3

Finally the ubiquity of SCF and its tyrosine kinase receptor c-KIT in different kinds of

cells and tissues, and their importance in wound healing and cancer led to their

investigation as potential players in keloid pathogenesis. The study demonstrated that

both SCF and c-KIT were upregulated in keloid scar tissues in vivo. They were also

upregulated in cultured fibroblasts on stimulation with serum highlighting their possible

role in the initial phases of the healing process when the wound is flooded with serum. In

addition, we demonstrated that epithelial-mesenchymal interactions, mimicked by

coculture of keratinocytes and fibroblasts in vitro, not only stimulated secretion of

soluble form of SCF in keloid cocultures but also brought about shedding of the

extracellular domain of c-KIT perhaps by upregulation of TACE which was also shown

to be elevated in keloid tissues and keloid cocultures. Although the increased

phosphorylation of c-KIT suggests the activation of SCF/c-KIT pathway in keloid

cocultures, the exact role of the shed c-KIT is yet to be understood.

Having identified a tyrosine kinase receptor system as a major player in keloid

pathogenesis, we investigated the role of an established tyrosine kinase inhibitor, gleevec,

as a possible therapeutic agent for keloid scars. It was observed that although gleevec did

not have a significant effect on the proliferative potential of KF and KK, it was as able to

downregulate the expression of ECM components as well as profibrotic factors in KF via

inhibition of the c-KIT mediated PI3 kinase signaling pathway. It was also able to render

the KF less bioenergetic by reducing the intracellular ATP levels and their rate of ATP

synthesis. Thus given its effects, it has a potential of being a therapeutic agent for keloids.

Page 15: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

4

1 BACKGROUND AND INTRODUCTION

Page 16: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

5

1.1 OVERVIEW OF THE WOUND HEALING PROCESS The wound healing process is a highly orchestrated process comprising of overlapping

phases of inflammation, granulation tissue formation, angiogenesis and remodeling of the

wound matrix (Clark et al., 1996). After wounding, the healing cascade immediately

ensues with the release of various growth factors and cytokines from the serum of the

disrupted blood vessels and degranulating platelets. The disruption of the blood vessels

results in the formation of a fibrin clot which not only works as a barrier for invading

microorganisms but also as a mesh for the binding of inflammatory cells, fibroblasts and

growth factors (Tuan et al., 1998). Within a few hours of injury, inflammatory cells like

neutrophils followed by monocytes and lymphocytes infiltrate the tissue and phagocytose

the bacteria. In addition to their role in defence against microorganisms, inflammatory

cells also serve as a pool for several growth factors and other macromolecules which are

required to intiate the proliferative phase of the healing process. The proliferative phase

involves the migration and proliferation of the keratinocytes from the wound edge

followed by the proliferation of fibroblasts in the area neighbouring the wound. These

fibroblasts then migrate into the wound area over a provisional wound matrix, deposit

large amounts of ECM and subsequently transform into myofibroblasts with a contractile

phenotype (Werner et al., 2003). Myofibroblasts are a group of actin rich fibroblasts and

are known to play an important role in wound contraction. Proto-myofibroblasts

differentiate to myofibroblasts by the production of α-SMA in order to generate more

force for contracture (Tomasek et al., 2002; Roy et al., 2001). At the same time,

angiogenic factors are released which promote angiogenesis by stimulating endothelial

cell proliferation leading to capillary tube formation. The resulting wound connective

Page 17: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

6

tissue is called granulation tissue with nerve sprouting occuring at the wound edge.

Finally, a transition from granulation tissue to mature scar occurs, characterized by

continued collagen synthesis and collagen catabolism. This process requires a balance

between matrix biosynthesis and matrix degradation. A disruption in this balance either

due to excessive matrix deposition or decreased matrix degradation leads to keloid and

hypertrophic scars (Nedelec et al., 1996; Raghow et al., 1994).

Figure 1: Schematic representation of different stages of wound repair (Adapted from Werner et al., 2003). A: 12–24 h after injury the wounded area is filled with a blood clot. Neutrophils invade into the clot. B: at days 3–7 after injury, neutrophils undergo apoptosis. Instead, macrophages are abundant in the wound tissue at this stage of repair. Endothelial cells migrate into the clot; they proliferate and form new blood vessels. Fibroblasts migrate into the wound tissue, where they proliferate and deposit extracellular matrix to form a granulation tissue. Keratinocytes proliferate at the wound edge and migrate down the injured dermis and above the provisional matrix. C: 1–2 wk after injury the wound is completely filled with granulation tissue. Fibroblasts have transformed into myofibroblasts, leading to wound contraction and collagen deposition. The wound is completely covered with a neoepidermis.

12–24 h

Day 3-7

1-2 Week

Page 18: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

7

1.2 KELOID SCAR

1.2.1 Clinical Characteristics of Keloid

Keloid scars represent a pathological response to cutaneous injury and occur only in

humans. The term ‘keloid’ is derived from a Greek word “khele” meaning crab claw

(Ladin et al., 1995). Keloids usually appear as firm broad nodules, often erythematous

and with a shiny surface (Marneros et al., 2004). A keloid scar extends beyond the

confines of the original wound, does not regress spontaneously, grows in pseudotumor

fashion with distortion of the lesion and tends to recur after excision. They are

notoriously resistant to therapy. Numerous treatment modalities are available, none of

which are consistently effective (Poochareon et al., 2003).

Fibroproliferative disorders (FPD) involve various pathologic fibrotic conditions which

constitute a leading cause of mortality in the United States (from a 5 year running

average of the US bureau of vital statistics analysis of cause of death, 45% of deaths in

1988 included death due to any form of fibrosis of any organ or tissue) (Kozak et al.,

1988; Bitterman et al., 1991).

The keloid is a dermal form of FPD and although it has much less morbidity as compared

to FPD of other organs, it causes considerable functional and aesthetic problems

particularly after thermal injury (Tredget et al., 1990; Engrav et al., 1987). They have a

propensity to occur in melanocyte-rich regions like face, neck, deltoid, presternal area

and ear lobes (Kim et al., 2000; Crockett et al., 1964; Bayat et al., 2004). They have also

Page 19: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

8

been reported to appear on the cornea (Shukla et al., 1975; Shoukrey et al., 1993; Lahav

et al., 1982).

Keloids are often compared with hypertrophic scars. Though their gross appearance is

similar, hypertrophic scars unlike keloid scars, remain confined to the borders of the

original wound and most of the times retain their shape (Mutalik et al., 2005) [21]. In

addition, though hypertrophic scars develop within a few weeks after skin injury, keloid

scars often show a delayed onset.

Figure 2: Keloid formation in different patients (Adapted from Marneros et al., 2004)

Page 20: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

9

1.2.2 Epidemiology

There is believed to be a genetic and racial predisposition for the development of keloids

in darker-skinned races compared to lighter skinned people. Some 15-20% of blacks,

hispanics, and asians are afflicted with the disorder. Studies have reported incidence

ratios between the two groups to range from 2:1 to 19:1 (Oluwasnami et al., 1974; Omo-

Dare et al., 1975). Both autosomal dominant and autosomal recessive genetic inheritance

have been proposed but not confirmed (Omo-Dare et al., 1975). Although keloids are

known to occur sporadically, some data suggest familial occurrence (Bloom et al., 1956).

Differences in occurrence of keloids based on age and gender has also been reported. It

has been shown to be predominant in females and in populations between the ages of 10

and 30 (Cosman et al., 1961; Murray et al., 1981). Until now, no susceptibility genes for

keloid formation have been identified. TGF-β1, β2, β3, and TGF-β receptor

polymorphisms have bene studies but has not yielded any statistically significant

associations with keloids in case control studies (Bayat et al., 2002, 2003, 2004,2005 ).

The Difficulty in identifying genes specific to keloids among patients with keloids may

reflect genetic heterogeneity, whereby different genes contribute to keloid formation in

different families (Robies et al., 2007)

1.2.3 Histopathology

Contrary to the appearance of organized fibres in normal skin, keloids have stretched

collagen fibres aligned in the epidermal plane (Kelly et al., 1988). Excessive deposition

of collagen and other extracellular matrix components along with increased blood vessels

Page 21: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

10

and cells are observed in the histological sections of keloids. However the microvessels

in keloids seem to be occluded perhaps to the increase proliferation of endothelial cells

(Kischer et al., 1992). Keloid scars are also characterized by presence of tongue-like

advancing edge underneath normal-appearing epidermis and papillary dermis (Lee et al.,

2004). Keloid sections are also characterized by a thickened epidermis and prominent

disarray of fibrous fascicles/nodules. Mast cells, macrophages, epidermal langerhan cells

have all been demonstrated to be elevated in keloids. However their role in keloid

pathogenesis is not well understood. At higher magnification using scanning electron

microscopy, the random nature of fiber orientation, varying fiber length and poor bundle

formation in keloids are observed.

Figure 3: H&E staining of normal skin and keloid scar tissues

Normal Skin Keloid Scar

Page 22: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

11

1.2.4 Molecular Pathogenesis

The wound healing process involves a complex interplay of cells, mediators, growth

factors, and cytokines, leading to inflammation, cell proliferation, ECM deposition,

contraction and remodeling. Thus a disruption in the cellular harmony leads to either a

delayed healing response or execessive healing. The over healing response is marked by

an over exuberant deposition of collagen and other extracellular matrix components like

fibronectin by fibroblasts resulting in a scar which can either be keloidic or hypertophic.

Previous studies have demonstrated that keloid-derived fibroblasts produced 20 times

more type1 collagen than normal fibroblasts in vitro (Ladin et al., 1995; Ala-kokko et al.,

1987; Di cesare et al., 1990).

Keloids are characterized by an increased proliferation of fibroblasts in vitro (Calderon et

al., 1996). Recent studies have highlighted the importance of regulation of apoptosis, in

scar establishment and the development of a pathological scar. Apoptosis-related genes

and proteins have been shown to be down-regulated or mutated in keloid tissues and

fibroblasts (Sayah et al., 1999; Chodon et al., 2000).

The bioactivity of fibroblasts in fibrogenesis or excessive scar formation is regulated by a

number of growth factors, of which the TGF-β family is thought to play a central role by

its regulation of fibroblast proliferation, differentiation and matrix production. It is

believed to enhance the production of ECM elements, such as fibronectin and collagen,

and to upregulate the cellular expression of matrix receptor integrin (Tuan et al., 1998;

Niessen et al., 1999; Kim et al., 2000).

Page 23: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

12

TGF-β1 in particular has been widely implicated as a key modulator of cellular dynamics

in fibrosis. Many groups have demonstrated increased levels of TGF–β in keloid tissues

and other fibrotic disorders (Lee et al., 1999; Younai et al., 1994; Polo et al., 1999).

Peltonen and co workers (1991) demonstrated that TGF-β1 mRNA and TGF-β1 protein

were associated with excessive collagen synthesis and ECM accumulation in keloids.

Shah’s group (1994) showed that blocking of TGF–β1 and TGF–β2 by the application of

antibodies to cutaneous wounds markedly reduced scarring. Liu and team (2002)

demonstrated, by an in vitro experiment, that the effects of TGF-β1 might be modulated

by an autocrine loop and that this autocrine regulation might play an essential role in

keloid formation and development. Further, Chodon’s group (2000) reported that

dysregulation in the Fas-mediated apoptosis, which occurred in normal wound healing,

may be a factor contributing to scar formation and suggested a role for TGF-β1 in this

resistance.

Various other molecular pathways have been invoked in keloid pathogenesis. Platelet-

derived growth factor (PDGF) α-receptor expression was demonstrated to be elevated in

KF and this corresponded to an increase in mitotic response of the fibroblasts on

exposure to the PDGF isoforms (Haisa et al., 1994). An over expression of insulin-like

growth factor-I (IGF-1) receptor in KF was also demonstrated to enhance their invasive

potential highlighting the role of IGF-1 in keloid pathogenesis (Yoshimoto et al., 1999).

An elevated expression of interleukin (IL)-6 (Xue et al., 2000) and VEGF (Wu et al.,

2004; Le et al., 2004) has also been reported in KF.

Page 24: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

13

As the wound milieu consists of various cell types, it would be foolhardy to implicate

fibroblasts alone as main players in fibrosis. Other cell types have also been shown to

play a role in keloid pathogenesis directly or indirectly by influencing the fibroblasts. An

elevated presence of mast cells was observed in keloids suggesting their possible role in

keloid pathogenesis (Russel et al., 1977). Upon further investigation it was observed that

an elevation of mast cell histamine resulted in an up-regulation of procollagen type 1

production in KF (Kikuchi et al., 1995; Tredget et al., 1998). Another cell type which has

been shown to play an important role in pathogenesis of scars is endothelial cells. Along

with fibroblasts they are known to be important in production of collagen (Milsom et al.,

1973; Sollberg et al., 1991; Diegelmann et al., 1977). It has been observed that

overproliferation of endothelial cells leads to occlusion of microvessel lumens in keloids

resulting in a severe hypoxic condition (Kischer et al., 1982). The resulting hypoxia

further stimulates endothelial cell proliferation and collagen production by fibroblasts by

releasing growth factors like HIF-1α and VEGF (Knighton et al., 1983; Kischer et al.,

1992; Zhang et al., 2006).

It is now becoming clear, that epithelial-mesenchymal interactions, initially applied to

normal skin homeostasis (Maas-Szabowski et al., 2000; Mackenzie et al., 1994; Fusenig

et al., 1994), are also instrumental in modulating fibroblast behaviour in keloids by

paracrine influences. Previous laboratory-based findings showed that keloid-derived

keratinocytes enhanced both normal and keloid-derived fibroblast proliferation and

collagen synthesis in a co-culture system (Lim et al., 2001; Funayama et al., 2003; Xia et

al., 2004). NF and KF cocultured with KK in defined serum-free media showed a higher

rate of proliferation compared to that of coculture with NK (Lim et al., 2001; Funayama

Page 25: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

14

et al., 2003; Xia et al., 2004). It was subsequently observed that KF co-cultured with KK

produced increased amounts of both collagens I and collagen III compared with coculture

with NK (Xia et al., 2004; Lim et al., 2002; Lim et al., 2003). Mitogen activation protein

kinase (MAPK) and phosphatidyl-inositol-3 kinase (PI-3K) pathway activation have been

observed in excessively proliferating KF cocultured with KK. When these fibroblasts

were treated with MAPK-p44/42 specific inhibitor and PI-3K specific inhibitor, it caused

complete nullification of collagen I and III production, and significantly decreased

fibronectin and laminin β2, all of which is produced in excess in keloidogenesis (Lim et

al., 2003). Electron microscopic analysis also demonstrated that the appearance of

collagen–extracellular matrix (ECM) produced by NF cocultured with KK in vitro,

approximated the morphological appearance of in vivo keloid scar tissue (Lim et al.,

2002). Epithelial-mesenchymal interactions have been demonstrated to influence the

expression of various transcription factors, growth factors and cytokines in vitro. Phan

and his group (2005) demonstrated that epidermal-dermal interactions in keloids resulted

in activation of the TGFβ-Smad axis which in turn played a crucial profibrotic role in

keloid pathogenesis. Lim and his co-workers (2006) further demonstrated the modulation

of yet another transcription factor STAT-3 by epithelial-mesenhcymal interactions in

keloidic cells. Ong’s team (2007) used a two chambered coculture system to mimic

epithelial-mesenchymal interaction and reported the expression and secretion of VEGF to

be upregulated in KF on being coculutred with KK. Biologically active CTGF was also

shown to be secreted by KF only in the presence of overlying keratinocytes (Khoo et al.,

2006).

Page 26: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

15

In addition to the increase of profibrotic molecules, anti fibrotic growth factors and

enzymes have also been demonstrated to be downregulated in keloids. The balance

between matrix metalloproteinase (MMP) and their inhibitors TIMP is critical for over all

ECM turnover. Concentrations of collagenase inhibitors, α-globulins and plasminogen

activator inhibitor -1 have been shown to be elevated in both in vitro and in vivo keloid

samples, whereas the levels of degradative enzymes are frequently decreased

(Diegelmann et al., 1977; Tuan et al., 1996). It has also been observed that MMP activity

differs between KF and NF and these differences affect the phenotype (Uchida et al.,

2003).

Although attempts are being made in understanding the molecular basis of keloid

pathogenesis, the complexity of the repair process and the lack of proper in vitro and in

vivo animal models have hindered progress in revealing the mechanisms underlying

keloid scar formation. Thus it is imperative that new in vitro and in vivo methods are

developed to assists researchers to delineate the dynamics of growth factors, deemed

important in normal wound healing process, in pathological scars.

Page 27: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

16

Figure 4: Schematic representation of pathways potentially resulting in accumulation of collagen in fibrotic skin diseases (Adapted from Uitto et al., 2007). The net deposition of collagen is a balance between the rate of synthesis and the rate of degradation, both of which can be modulated by a number of factors, such as cytokines. These factors can be evoked by stimuli such as trauma to the skin. When superimposed on the individual’s genetic background, an imbalance in the flux through these pathways can result in collagen accumulation manifesting as tissue fibrosis.

1.2.5 Current Treatment

Several modalities have been explored for either the treatment of keloid scars or

prevention of their recurrence after surgery. However the several treatment strategies

used, remain unsatisfactory. The current treatments available are as follows:

Surgery: Surgical excision of hypertrophic and keloid scars when combined with steroid

injection reduces the recurrence rate. However, excision alone in keloids is met with high

Page 28: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

17

recurrence rate of 45 to 100% (Berman et al., 1995; 1996). Due to this, various adjunct

therapies have been explored to reduce the problem of recurrence.

Silicone Gel Sheeting: Silicone gel is a crosslinked polymer of dimethylsoloxane and is

not only an effective adjunct to excision of keloids but also a prophylaxis to abnormal

scarring in some incisions (Al-Attar et al., 2006). Despite the wide appreciation about the

effect of silicone gel sheeting among the physicians, the mode of action of silicone is still

unknown. Although silicone gel is comfortable, it requires active patient compliance and

long-term application which is challenging on mobile and angled anatomical sites (Sproat

et al., 1992).

Corticosteroid Injection: Injection of triamcinolone is efficacious for the treatment of

keloid scars in which it is used as a first-line therapy (Shons et al., 1983; Tang et al.,

1992). Corticosteroids are usually combined with surgical excision of the scar (Berman et

al., 1996). However intralesional injections of corticosteroids are highly painful. In

addition other side effects are skin atrophy, loss of pigmentation, and telangiectasia

(Sproat et al., 1992).

Pressure Therapy: Pressure therapy is an effective therapy with minimal side effects for

keloids but its practical use is limited to ear lobes. It is generally used as a post operative

adjunct for earlobe keloids (Brent et al., 1978; Russell et al., 2001).

Radiotherapy: Radiotherapy has been used as monotherapy, and in combination with

surgery, for hypertrophic scars and keloids. It is effective in reducing the recurrence rate

of scars. However, monotherapy remains controversial because of anecdotal reports of

carcinogenicity following the procedure (Thomas et al., 2002; Norris et al., 1995).

Page 29: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

18

Cryotherapy: Cryotherapy is an option considered in treatment of very small scars like

those arising from very severe acne (Layton et al., 1994). It uses rapid, repeated cooling

and rewarming of tissue causing cell death and tissue sloughing (Al-Attar et al., 2006).

Side-effects produced are slow healing of the wound along with tissue necrosis, altered

pigmentation, moderate skin atrophy and pain. The response rate is around 51 to 74

percent and may require more than one application (Thomas et al., 2002).

New emerging therapies: Recently, a number of novel experimental therapeutic

approaches have been explored, based on results from in vitro experiments from cells

derived from keloid tissue [eg. Interferon γ (Granstein et al., 1990; Larrabee et al., 1990),

imiquimod (Berman et al., 2002), 5-fluorouracil (Manuskiatti et al., 2002 ; Fitzpatrick et

al., 1999), bleomycin (Yamamoto et al., 2006).

Although new treatment modalities are constantly been explored, most of them are

accepted on a broad consensus rather than backed by a good knowledge of the

pathogenesis of keloid and hypertrophic scars. Adding to the problem, there is an evident

shortage of randomized controlled trials to assess the efficacies of existing therapeutic

interventions. It is also not possible to incorporate variables which widely exist among

the patients in actual clinical setting, like differences in age, race, involved body part,

associated systemic diseases like diabetes, local infection, in a volunteer-based study

using artificial methods of induction of wound. Results based on animal/animal cell

studies may not be reliable because animal tissue/cell behaviors are much different from

that of humans and more over, diseases like keloid exclusively occur in humans. These

Page 30: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

19

limitations make the assessment and comparison of different treatment methods very

difficult.

Thus, for more effective and specific treatments, it is imperative that molecular

mechanisms that cause keloid formation are elucidated and better understood. Increased

understanding at each level of pathogenesis may lead to development of new therapies, in

form of synthetic drugs or specific growth factors, to downregulate profibrotic molecules.

Monoclonal antibodies, growth factor receptor antagonists, antisense oligonucleotides are

some of the treatment modalities that hold promise.

1.3 OBJECTIVES OF THE PRESENT STUDY We hypothesise that epithelial–mesenchymal interactions have an important governing

role in keloid formation, and that keloid formation may be the consequence of abnormal

keratinocyte control over fibroblasts, mediated by the key factors like activin-A,

proteoglycans, Stem cell factor , rather than a defect of fibroblasts themselves.

The broad objective of this study is to identify key modulators of cellular dynamics in

keloid scars and investigate the effect of epithelial-mesenchymal interactions on the

expression profiles of these modulators using various in vitro models. Depending on the

profibrotic or antifibrotic effects of these modulators, they could either be targets for

therapeutic intervention using chemical agents or themselves be used as natural

therapeutic agents for keloid scars, respectively.

Page 31: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

20

The specific objectives of this study are as follows:

1. To investigate the role of epithelial-mesenchymal interactions, especially

fibroblast response to keratinocyte paracrine stimulation in wound contracture.

2. To investigate the role of various growth factor systems in keloid pathogenesis

a) Activin-A/ follistatin system

b) Proteoglycans

c) SCF/c-Kit system

3. Finally to investigate both growth factors and synthetic agents which could be

used as therapeutic agents for keloids

a) Natural modulators: decorin, follistatin

b) Synthetic modulators: gleevec (Imatinib mesylate)

Page 32: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

21

2 MATERIALS AND METHODS

Page 33: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

22

2.1 MEDIA AND CHEMICALS

Dulbecco’s modified eagle medium (DMEM), Hank’s balanced salt solution (HBSS),

fetal calf serum (FCS), streptomycin, penicillin, gentamicin and fungizone were

purchased from Gibco. Keratinocyte growth medium (KGM) was purchased from

Clonetics (USA). Phosphate buffered saline without Ca2+ and Mg2+ (PBS), epidermal

growth factor (EGF), cholera toxin and hydrocortisone were purchased from Sigma

Chemical Co (USA). Dispase II was purchased from Boehringer Mannheim (USA).

Rhodamine counter stain was obtained from Difco (USA). Tris base was purchased from

J.T Baker. Triton X-100, ethylenediaminetetraacetic acid (EDTA), 30% acrylamide/bis

solution (37.5:1 2.6%C) and glycine were purchased from Biorad. Sodium Chloride

(NaCl), nonidet P-40 (NP-40), sodium dodecyl sulphate, hydrogen peroxide (H2O2),

bovine serum albumin (BSA), tween-20, potassium chloride (KCl), potassium phosphate

(K3PO4), magnesium chloride (MgCl2), MTT [3-(4,5-Dimethylthiazol-2-yl)-2,5-

diphenyltetrazolium bromide], N,N –dimethylformamide (DMF) and paraformaldehyde

were all purchased from Sigma Chemical Co (USA). Methanol and acetic acid were

purchased from Lab-Scan.

2.2 RECOMBINANT GROWTH FACTORS AND ANTIBODIES

Mouse anti-α- SMA monoclonal antibody (Sigma), mouse anti-TGF- β1 neutralizing

antibody (R&D systems), Mouse anti-Collagen I,III monoclonal antibody

(Monosan®antibodies-The Netherlands), rabbit anti-fibronectin monoclonal antibody

(BD Transduction Laboratories), mouse anti-activin-A monoclonal antibody (Oxford

Page 34: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

23

Bio-Innovations, Oxford, UK) and mouse anti-follistatin (Gift from Dr David Phillips,

Monash University, Melbourne, Australia), rabbit-anti syndecan-2 polyclonal antibody

(Santa Cruz Biotechnology), rabbit-anti FGF-2 polyclonal antibody (Santa Cruz

Biotechnology), mouse anti-decorin monoclonal antibody (R&D systems), rabbit-anti

TACE polyclonal antibody (Santa Cruz Biotechnology), rabbit anti-c-Kit (tyr 703)

polyclonal antibody (Affinity BioReagents), rabbit-anti c-Kit (tyr 721) polyclonal

antibody (Affinity BioReagents), mouse-anti c-Kit monoclonal antibody (Neomarkers),

mouse-anti c-Kit monoclonal antibody (R&D systems), rabbit-anti VEGF polyclonal

antibody (Neomarkers), HDGF (Gift from M.D Anderson Cancer Center, U.S) , mouse-

anti Phospho Akt (Cell signaling), mouse-anti Phospho mTOR (Cell signaling), Labeled

chicken- anti mouse IgG antibody (Molecular probes), goat-anti rabbit IgG H&L (HRP)

polyclonal antibody (abcam), rabbit-anti mouse IgG H&L (HRP) polyclonal antibody

(abcam), rabbit-anti mouse IgM H&L (HRP) polyclonal antibody (abcam), recombinant

human activin-A (R&D systems), recombinant human follistatin (R&D systems),

recombinant human decorin (R&D systems), recombinant human TGF-β1 (R&D

systems), recombinant human FGF-2 (R&D systems).

2.3 PREPARATION OF NORMAL AND KELOID TISSUE EXTRACTS

Normal and keloid skin tissues were cut into small pieces using a scalpel to about 120

mg. 200 µl of lysis buffer containing 20 mM Tris-HCl (pH 7.5), 1% v/v Triton X-100,

100 mM NaCl, 0.5% w/v Nonidet P-40 and 1mg/ml protease inhibitor cocktail

(Boehringer Mannheim, Mannheim, Germany) were added to the tissue sections and

Page 35: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

24

sonicated. This was followed by centrifugation at 13000 x g for 10 min. Supernatant was

collected while the pellet was discarded. Protein concentration of the tissue extracts were

determined by the Bradford method.

2.4 IMMUNOHISTOCHEMISTRY

2.4.1 Preparation of Paraffin Sections of Normal and Keloid Tissues

Normal and keloid tissue samples were fixed in 4% v/v formalin in PBS for 24 hr (longer

for larger samples but never more than a week) followed by immersion in 70% v/v

ethanol overnight, in histocassettes. This was followed by paraffin embedding as per

standard protocol (BD biosciences). Sections were cut at 5 microns onto slides.

Tissue sections were dried onto the slides at 37 oC overnight.

2.4.2 Pretreatment of Paraffin Sections for Immunohistochemistry

The paraffin sections were dewaxed or deparaffinized in two changes of xylene followed

by re-hydration at 100%, 95% and 70% v/v ethanol gradients. The antigens were then

retrieved by immersing slides in 0.01 M citrate buffer, pH 6.0, heating in a microwave

oven (high for 2.5 min, low for 5 min), cooling at 4°C for 20 min, and washing in water

for 5 min. Endogenous peroxidase was blocked in 3% v/v H2O2, and non-specific binding

blocked for 1 hr (CAS block, Zymed Laboratories, South San Francisco, CA, USA).

Page 36: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

25

2.4.3 Probing with Antibody and Developing

Sections were then incubated in specific antibodies for 1-2 hrs at room temperature. After

washing, slides were incubated in universal secondary antibody provided in the vectastain

kit (Vector Labs). Slides were washed in Tris-buffered NaCl (TBS) 0.05% w/v Tween-20

pH 7.5, then MilliQ H2O. The sections were then treated with “vectastain elite RTU elite

reagent” for five minutes. The reaction product was developed with a 3, 3’-

diaminobenzidine tetrahydrochloride substrate kit (Dako), and sections counterstained

with hematoxylin. All wash steps were in TBS/0.05% w/v Tween-20. Antibodies were

diluted in 1% w/v BSA/TBS.

2.5 CELL CULTURE

2.5.1 Keloid Keratinocyte and Fibroblast Database

Keratinocytes and fibroblasts were randomly selected from a specimen bank of

keratinocyte/fibroblast strains derived from excised keloid specimens. All patients had

received no previous treatment for keloids before enrollment. Before informed consent

was obtained, a full history was taken and an examination performed, complete with

photographic documentation. The tisssues were separated into normal and keloids by an

approved surgeon and pathologist. Approval by the National University of Singapore-

Institutional Review Board (NUS-IRB) was obtained for this study.

Page 37: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

26

2.5.2 Keloid and Normal Keratinocytes from Keloid scar and Normal Skin

Excised keloid scar and normal skin specimens were repetitively washed in PBS

containing 150 µg/ml gentamicin and 7.5 µg fungizone, until the washing solution

became clear. The tissue was then divided into pieces of approximately 5 mm × 10 mm

and the epidermis was scored. Dispase 5mg/ml in HBSS was added and skin was

incubated overnight at 4ºC. The epidermis was carefully scraped off with a scalpel the

next day and placed in trypsin 0.25% w/v /glucose 0.1% w/v /EDTA 0.02% for 10 min in

the incubator. Trypsin action was quenched by DMEM/10% FCS. The suspended cells

were transferred into tubes and centrifuged at 1000 rpm for 8 min. The cells were seeded

in Keratinocyte Culture Medium (80 ml DMEM supplemented with 20 ml FCS, EGF 10

ng/ml, cholera toxin 1 × 10-9 M and hydrocortisone 0.4 µg/ml) at 1 × 105 cells/cm2 for 24

hrs before changing to Keratinocyte Growth Medium (KGM). The cell strains were

maintained and stored at -150ºC. Only cells from second and third passages were used for

the experiments.

2.5.3 Keloid and Normal Fibroblast from Keloid Scar and Normal Skin

Remnant dermis from the keloid scar and normal skin were either minced or incubated in

a solution of collagenase type 1 (0.5 mg/ml) and trypsin (0.2 mg/ml) at 37ºC for 6 hrs.

Cells were pelleted and grown in tissue culture flasks. Alternatively the skin tissue

samples were chopped into pieces of 1-2 mm2. The pieces were then transferred to a 100

mm tissue culture dish previously coated with a thin layer of DMEM/10% v/v FCS.

Culture medium enough to cover the explants were then added and topped up after 2-3

Page 38: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

27

days. After 4-7 days the fibroblasts outgrew from the tissue. Fibroblast cell strains were

maintained and stored at -150ºC until use. Only cells from the second and third passages

were used for the experiments.

2.5.4 Keratinocyte-fibroblast Coculture KK obtained from randomly selected keloid strains were seeded at density of 1 × 105

cells/cm2 on 6-well transwell clear polyester membrane inserts with 0.4 µm pore size and

area 0.3 cm2 area (Costar Corp, USA). Ten days before co-culture, cells were maintained

in serum-free Keratinocyte Growth Medium (KGM) until 100% confluent in monolayer.

The medium was then changed to Dulbecco’s modified Eagle’s medium (DMEM)

supplemented with 10% heat inactivated fetal bovine serum (FBS) and

penicillin/streptomycin. The cells were raised to air-liquid interface to allow

keratinocytes to stratify and reach terminal differentiation.

KF obtained from randomly selected keloid strains were seeded in 6-well plates at a

density of 1 × 104 cells/ml in DMEM/10% v/v FCS for 24 hrs and then in serum-free

medium for another 48 hrs. Cells on both the membrane inserts and the wells were

washed twice with phosphate buffer saline (PBS) to remove the old medium before

combination of the inserts and plates for co-culture in serum-free DMEM. Controls

comprised of one series of non co-cultured keloid fibroblasts and one series non co-

cultured keloid keratinocytes. At day 5, the inserts with the cultured keratinocytes were

removed, and the conditioned media were collected, pooled and stored at -80ºC for later

Page 39: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

28

analysis. The fibroblasts and keratinocytes were also harvested for protein extraction for

western blot analysis. (Fig. 5)

Figure 5: Coculture of epidermal keratinocytes and dermal fibroblasts as an in vitro model to study epithelial-mesenchymal interaction

FFiibbrroobbllaasstt ssuubbccuullttuurree KKeerraattiinnooccyyttee ssuubbccuullttuurree

KKeerraattiinnooccyyttee-- FFiibbrroobbllaasstt CCooccuullttuurree

Page 40: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

29

2.6 CELL COUNTING

Before the cells were seeded into culture flasks for experiments, aliquots of the cell

suspension were mixed with trypan blue in a ratio of (1:4) and counted in a Neubauer’s

haemocytometer. All the cells (non-viable cells stained blue, viable cells became opaque)

were counted in the four corner squares of the hemocytometer. Since the volume of each

square is 10-4 cm3 use the following formula was used to calculate the number of cells in

the cell suspension

Cells per ml = the average count per square x the dilution factor x 104

Total cell number = cells per ml x the original volume of fluid from which cell sample

was removed.

2.7 TREATMENT OF FIBROBLASTS WITH GROWTH FACTOR

NF and KF from different patients were seeded in 6-well plates at a density of 1 × 104

cells/ml in DMEM/10% v/v FCS for 24 hrs and then in serum-free medium for another

48 hrs. The cells were subsequently treated with varying concentrations of activin-A (100

ng/ml; (R&D Systems), follistatin (FS-288, 100 ng/ml; R&D Systems) or TGF-β1 (5, 10

ng/ml; R&D Systems), decorin (0, 250, 500, 1000, 2000 ng/ml; R&D Systems), FGF-2

(10 ng/ml; R&D Systems) respectively, to test the effect of these growth factors on the

proliferation of fibroblasts and expression of key ECM proteins.

2.8 TREATMENT OF CELLS WITH GLEEVEC

Page 41: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

30

NF and KF from different patients were seeded in 6-well plates at a density of 1 × 104

cells/ml in DMEM/10% v/v FCS for 24 hrs and then in serum-free medium for another

48 hrs. The cells were subsequently treated with varying concentrations of gleevec (0.5

µg/ml, 1 µg/ml, 2.5 µg/ml, 5 µg/ml, 10 µg/ml, and 20 µg/ml), to test its effect on the

proliferation of fibroblasts and expression of key ECM proteins.

2.9 SERUM STIMULATION

When fibroblasts are exposed to serum, the signal is interpreted to be a physiological

wounding signal. The serum stimulation model is an in-vitro model used to mimic the

early phases of wounding. NF and KF were seeded in 6-well plates at a density of 1 × 104

cells/ml in 10% v/v FCS for 24 hrs and subsequently seeded in serum-free medium for

another 48 hrs. The fibroblasts were exposed to either DMEM supplemented with 10%

v/v FCS or DMEM alone for 24 hrs before being harvested for analysis.

2.10 MTT ASSAY

The MTT assay is a commonly used colorimetric assay to quantify cell numbers. It is

widely used in studies involving cell proliferation or cell toxicity. MTT [3-(4,5-

Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium] is cleaved by an active succinate-

tetrazolium reductase system present in the mitochondrial respiratory chain of a living

and metabolizing cell into blue formazon crystals which can be solubilized and

absorbance measure. The relationship between cell number and absorbance is linear.

Page 42: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

31

In our experimental set up, the treated cells in 96-well plates were incubated with 10 µl of

MTT (5 mg/ml) in 100 µl of DMEM to give a final concentration of 0.5 mg/ml in each

well for about 2hrs. The medium was removed and the blue crystals were soubilized by

Hansen’s method (1989) using 20% w/v SDS in a solution of DMF: water (1:1 v/v) and

shaking in an orbital shaker. The absorbance of the solution was then measured directly

by using a plate reader at 570 nm.

2.11 FIBROBLAST-POPULATED COLLAGEN LATTICE (FPCL)

2.11.1 Preparation of Fibroblast-Populated Collagen Lattices

Collagen lattices were prepared from type 1 collagen extracted from rat tail tendons in

0.1% (v/v) acetic acid. A working solution was constituted by diluting the collagen to 1.6

mg/ml on the day of experiment. Both NF and KF cell strains were trypsinised,

resuspended in medium and counted using a hemocytometer. A collagen-fibroblast (for

both NF and KF) suspension was prepared in serum-free DMEM and 2 ml of the

suspension was dispensed into 32-mm petri dishes. The final concentration of collagen

was 0.8 mg/ml with a cell density of 6.7 × 104 cells/ml. The collagen lattices were

subjected to different treatments and the area of the lattice calculated.

Page 43: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

32

2.11.2 Macroscopic Evaluation of FPCL Contraction

The lattice diameters were assessed at various time-points for several days by placing the

dish on transparent graph paper and measuring two diameters at right angles to one

another. The area of collagen gel populated with fibroblasts was photographed. The

average area was calculated from the mean of the diameters and expressed as a

percentage of initial lattice area.

Percentage of initial area = A2/A1 × 100,

where A1 = initial gel area and A2 = area at the observed interval

2.12 WESTERN BLOTTING

To study the changes in the expression of investigative proteins in keratinocytes and

fibroblasts under different culture conditions and under treatment with different drugs,

cells were lysed in lysis buffer containing 20 mM Tris-HCL (pH 7.5), 1% v/v Triton X-

100, 100 mM NaCl, 0.5% w/v Nonidet P-40 and 1 mg/ml protease inhibitor cocktail

(Antipaindihydrochloride, Aprotinin, Bestatin, Chymostatin, E-64, EDTA-Na, Leupeptin,

Pefabloc-SC, Pefstatin, Phosphormidon; Boehringer Mannheim, Mannheim, Germany).

100 µg of protein was extracted and electrophoresed on 8% w/v and 14% w/v sodium

dodecyl sulphate-polyacrylamide gels using the Protein II system (Bio-Rad). The proteins

were subsequently transferred to a nitrocellulose membrane and blotted for antibodies

against the proteins under investigation. This was followed by treatment with respective

Page 44: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

33

secondary antibodies. The blots were then visualized with a chemiluminescence-based

photoblot system (Amersham Biosciences).

2.13 IMMUNOASSAY

An immunoassay is a biochemical test generally used to measure the level of antigens

present in a biological fluid like serum or urine. It has an advantage of being highly

specific as well as sensitive. Due to this reason, often very low volume of sample is

required to detect the antigen.

In our study, ELISA kits for detection of investigative proteins were purchased and

assays performed on conditioned media (100 µl) obtained from NF, NK, KF and KK.

2.14 FLOW CYTOMETRY

FACS analysis was performed to detect the investigative protein’s expression. Briefly,

about 5*105 to 1*106cells were washed twice with ice-cold PBS at 250 g for 5 min. The

cells were then incubated with antibodies against the proteins for 30 min at 4°C followed

by incubation with an FITC-conjugated secondary antibody in for 30 minutes at 4°C in

the dark. Cells treated with an isotype control or without primary antibody were used as

negative controls. All washing steps involved washing with flow cytometry buffer

containing PBS, 2% v/v FBS and 0.1% v/v sodium azide. Thereafter, the cells were fixed

in 2% w/v paraformaldehyde in PBS for 15 minutes at 4°C in the dark. A FACScan

(Becton Dickinson, Sunnyvale, CA) flow cytometer was used for the analysis.

Page 45: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

34

2.15 RNase PROTECTION ASSAY

The RNase protection assay is a highly sensitive method used to detect and quantify

specific mRNA transcripts in a mixture of total RNA or mRNA. The assay involves

hybridising a radioactive probe complementary to the gene sequence of interest, and then

adding RNase to the mixture. The RNase digests the entire single stranded RNA leaving

behind the double stranded probe-target hybrid. This hybrid is then electrophoresed on a

denaturing TBE-urea polyacylamide gel and detected.

In this study, the total RNA was isolated from the tissue lysate. RNase protection assay

was performed as described by Werner’s group (1993) using a radiolabelled antisense

probe complementary to the activin-βA mRNA (Hübner et al., 1996). Hybridization of

the same RNA with a gapdh riboprobe served as a loading control.

2.16 RATE OF ATP SYNTHESIS

The measurement of ATP by the luciferin-luciferase reaction was done as previously

described. (Vincent et al., 2004; Zhang et al., 2004) and was extended in this study to

digitonin-permeabilized whole fibroblasts. KF were permeabilized with 65 µg

digitonin/106 cells in buffer (150 mM KCl, 25 mM Tris-HCl, 2 mM EDTA, 0.1% w/v

BSA, 10 mM potassium phosphate, 0.1 mM MgCl2, pH 7.4). The reaction mixture

contained 1 mM ADP in a total volume of 0.25 ml of the above buffer. The reaction was

started with 50,000 cells, treated with the drug or only treated with DMEM with 5% v/v

serum for 24 hrs, followed by incubation at 37oC for 5 min and stopped by boiling for 3

min. Following centrifugation, 25 µl supernatant was added to a well in a 96-well plate

Page 46: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

35

together with 100 µl of 250x-diluted FL-AAM (Sigma). The chemiluminescence

generated was read in a luminometer (PerkinElmer Victor) as described previously by

Ng’s group (2006). The amount of ATP biosynthesized in 5 min was extrapolated from a

standard curve of ATP of 5 to 100 ρmol, a range which produced a linear response.

2.17 MEASUREMENT OF INTRACELLULAR ATP Intracellular ATP was determined from the whole cell lysate following the luciferin-

luciferase assay described above. Normalization was done with the protein content of the

lysate determined by the Bradford method.

2.18 STATISTICAL ANALYSIS Statistical significance between groups was assessed using either Student’s t-test or a

one-way analysis of variance with Tukey’s post-hoc test. All tests were performed using

MiniTab® software. Except for ELISA measurements where the mean value of data

from 5 different cell populations in each group was used, for all other results the cell

population representative of other populations with at least 5 different clinical samples

studied was conducted. Differences at the 95% level ( P value < 0.05) were considered to

be significant.

Page 47: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

36

3 ROLE OF EPITHELIAL-MESENCHYMAL INTERACTIONS IN WOUND CONTRACTION AND

SCAR CONTRACTURE

Page 48: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

37

3.1 INTRODUCTION The process of wound contraction, which involves the centripetal movement of the

wound edges by means of a retractile activity of the granulation tissue, is very critical for

proper healing of any cutaneous wound. However, unlike in other mammals, the

contraction of wound is often less beneficial in humans leading to minimal scarring in

some cases to major body deformations (Hunt et al., 1979; Rudolph et al., 1992).

Although it is widely accepted that the contractile force resides in the granulation tissue

which fills the wound (Carell et al., 1916), the exact mechanism is still not clearly

understood.

Epithelial-mesenchymal interactions have recently been explored as a possible factor

contributing to excessive scar formation. Given their emerging role in keloid

pathogenesis ((Lim et al., 2001; 2002; 2003; Funayama et al., 2003; Xia et al., 2004)

they might well be an important player in modulating wound contraction and scar

contracture. During wound contraction, fibroblasts differentiate into myofibroblasts

expressing α-smooth muscle actin. In normal wound repair, these myofibroblasts undergo

apoptosis and disappear but under pathological conditions these myofibroblasts become

apoptosis resistant, remain in the extracellular matrix and continue to remodel, leading to

sustained wound contracture (Tomasek et al., 2002). Previous studies reported the

modulation of underlying mesenchymal cell apoptosis by the epithelium (Hurle et al.,

1986). It has been further reported that epithelial–mesenchymal interaction upregulates

TGF-β1 (Funayama et al., 2003; Xia et al., 2004), a growth factor with important roles in

Page 49: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

38

wound and scar contraction (Kamamoto et al., 2003; Spyrou et al., 2002). In addition,

TGF-β has also been found to inhibit apoptosis of different types of cells including

myofibroblasts (Smith et al., 1999; Kim et al., 1998; Genestier et al., 1998) by various

mechanisms including the downregulation of c-myc, preventing the decline of Bcl-2

(Zhang et al., 1999) and through activation of the protein kinase pathway (Chin et al.,

1999). Taken a step further, epithelial-mesenchymal interactions would similarly affect

wound contracture by modulating apoptosis of myofibroblasts. The exact mechanism for

this however remains unclear.

New investigative tools, arising from tissue bioengineering have stimulated new studies

related to scar formation. Bell’s group (1979) described an in vitro model for the study of

wound contraction using collagen gels populated by fibroblasts. The advantage of this

model is that it includes the two fundamental dermal participants in scar formation: the

ECM and the fibroblasts, unlike the traditional in vitro models where the cells are grown

as a monolayer. Harris and co-workers (1981) demonstrated that the contraction of the

collagen lattice occurred as a consequence of fibroblast migration through the matrix.

This process was termed ‘tractional remodeling’ (Grinell et al., 1994).

In this study, we hypothesized the role of epithelial-mesenchymal interactions, especially

fibroblast response to keratinocyte paracrine stimulation in wound contracture. To test

our hypothesis, we investigated the contractile response of collagen lattices populated by

KF and NF, respectively to the addition of conditioned media obtained from KK/KF

coculture. To complement this data, immunohistochemical analysis of the fibroblasts

exposed to KK/KF conditioned media was performed to assess the expression of alpha-

smooth muscle actin in order to test our second hypothesis that fibroblasts exposed to this

Page 50: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

39

abnormal media might differentiate into the myofibroblasts in keloid scar tissue. The

effect of anti-TGF β1 neutralizing antibody on collagen lattice contraction induced by

KK/KF coculture conditioned media was also studied to investigate the role of this

fibroinductive cytokine.

3.2 RESULTS • Keloid or normal dermal fibroblast populated collagen lattice exhibited

significant contraction when treated with KK/KF coculture conditioned media

Study groups comprised collagen matrices populated with KF or NF exposed to the

conditioned media collected from KK/KF coculture or non-cocultured KKs (KK48,

KK43, KK30, KK25) or KFs (KF48, KF43, KF30, KF25). Lattice areas were calculated

at intervals of 1, 2, 3, 6, 24 and 48 hrs to determine the contraction percentage.

Conditioned media from the non-cocultured KK and non-cocultured KF controls induced

slight contraction of collagen lattices populated by KFs (Fig. 6a), but no contraction in

the group of collagen lattices populated with normal dermal fibroblasts (Fig. 6b).

Conditioned media obtained from KK/KF coculture, however, induced significantly

greater collagen lattice contraction, with nearly 90% or 60% contraction of the collagen

gel at 48 hrs compared with approximately 20% or 5% contraction for the controls, with

KF or NF, respectively (Fig. 6a,b).

Page 51: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

40

• Contraction of collagen lattice populated by KF was more significant as

compared to that populated by NF

The FPCLs were photographed and the area calculated as previously mentioned, for side-

by-side analysis. It was observed that collagen lattices populated with KFs demonstrated

significantly greater contraction when treated with KK/KF conditioned media compared

with collagen lattices populated with NF (Figs 6 and 7)

• Expression of α-smooth muscle actin was increased in keloid-derived fibroblasts

treated with keloid keratinocyte/keloid fibroblast coculture conditioned media

KF and NF were seeded at a density of 1000 cells per well in eight-well Laboratory-tek

chamber slides in DMEM/10% FCS for 24 hrs. The medium was then replaced by serum-

free DMEM for 48 hrs. After 48 hrs, conditioned media pooled from KK/KF coculture

and non-coculture samples were added. This was followed by addition of anti-α-SMA

antibody (Sigma, A-2547). A rhodamine counterstain (Difco) was added to the

fluorescein isothiocyanate-labelled secondary antibodies to reduce the nonspecific

background staining. A bright green fluorescence marked the positive expression of α-

SMA, while cells demonstrating minimal or no expression were stained dull orange.

Compared with control groups treated with non-coculture conditioned media, increased

expression of α-SMA (marked by bright green fluorescence) was seen in both KF

(Fig. 8a) and NF (Fig. 8b) study groups treated with conditioned media obtained from

KK/KF coculture. Actin fibres were more elongated and stressed in the cells treated with

KK/KF coculture conditioned media compared with those treated with non-coculture

Page 52: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

41

conditioned media. KFs demonstrated significantly increased expression of α-SMA

compared with similarly exposed NF (Fig. 8).

• Contraction of KF populated collagen lattice by KK/KF coculture conditioned

media is attenuated by anti-TGF-β1 neutralizing antibody.

Study groups comprised collagen matrices populated by KFs from sample KF48 with

conditioned media collected from KK48/KF48 coculture, non-cocultured KKs, non-

cocultured KFs and KK48/KF48 coculture with anti-TGF-β1 antibody. At each interval

(6, 24 and 48 hrs) the percentage lattice contraction was calculated. Significant

contraction was seen in collagen lattice exposed to conditioned medium obtained from

KK/KF coculture compared with non-coculture samples. The addition of anti-TGF-β1

antibody to the conditioned medium obtained from KK/KF coculture, however,

significantly decreased the rate and extent of contraction of the collagen lattice (Fig. 9).

Page 53: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

42

Figure 6: Effect of conditioned media collected from (KK)/(KF) cocultures on collagen contraction by KFs Collagen matrices composed of KFs (a) or NFs (b) were treated with conditioned media collected from non-cocultured KKs ,non-cocultured KFs (□) and KK/KF coculture (■). At each interval of study (1, 2, 3, 6, 24 and 48 h) the area of the gel was calculated as a percentage of the initial area. Each bar shows mean ± SEM of eight lattices pooled from four independent experiments using four different KF (a) or NF (b) cell strains.

Page 54: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

43

Figure 7: Comparison of contraction of collagen gel lattice incorporated with KFs and NFs. The fibroblast-populated collagen lattices were collected after 48 hrs, stained with rhodamine B and photographed. (a) NF1 and (b) KF48 were treated with KK48/KF48 coculture conditioned media. (c) NF2 and (d) KF30 were treated with KK30/KF30 coculture conditioned media. (e) NF3 and (f) KF25 were treated with KK25/KF25 coculture conditioned media. (g) NF4 and (h) KF43 were treated with KK43/KF43 coculture conditioned media.

Page 55: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

44

Figure 8: α-Smooth muscle actin (α-SMA) expression by KFs (a) or NFs (b). KFs (a) and NFs (b) were seeded at a density of 1000 cells per well in eight-well Lab-tek chamber slides in Dulbecco's modified Eagle's medium (DMEM)/10% fetal calf serum for 24 hrs. Medium was then replaced by serum-free DMEM for 48 hrs. After 48 hrs conditioned media pooled from keloid keratinocyte (KK)/KF coculture and non-coculture samples were added. This was followed by addition of anti-α-SMA antibody. A rhodamine counterstain was added to the fluorescein isothiocyanate-labelled secondary antibodies at a 1: 40 dilution. A bright green fluorescence marked the positive expression of α-SMA, while cells demonstrating minimal or no expression were stained dull orange. Each set represents KFs (a) or NFs (b) treated with conditioned media collected from KK/KF coculture.

Page 56: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

45

Figure 9: Effect of anti transforming growth factor (TGF)-β1 neutralizing antibody on collagen contraction by KF48 induced by conditioned media collected from KK48)/KF48 coculture. Collagen matrices composed of KF48 were treated with conditioned media collected from KK48/KF48 coculture (checked ■), KK non-coculture (■), KF non-coculture (□) or KK48/KF48 coculture with anti-TGF-β1 antibody ( ). At each interval of study (6, 24 and 48 hrs) the area of the gel was calculated as a percentage of the initial area. Results are shown as mean ± SEM.

Page 57: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

46

3.3 DISCUSSION In recent years, significant work has been done to support the hypothesis that epithelial–

mesenchymal interactions have an important governing role in keloid formation, and that

keloid formation may be the consequence of abnormal keratinocyte control over

fibroblasts rather than a defect of fibroblasts themselves (Yang et al., 2003). In this study

an established two-chamber coculture model was used to obtain conditioned media and

the effects of the latter was tested on FPCLs, an established three-dimensional wound

contraction and scar contracture model, to investigate the effect of paracrine secretions

from epithelial–mesenchymal interactions on collagen lattice contraction. Conditioned

media obtained from KK/KF coculture induced greater and faster contraction of collagen

lattices populated with KFs compared with lattices populated with NFs. It was also

observed that conditioned media obtained from non-cocultured KFs and KKs produced

decreased contraction of the collagen lattice. This underscores the importance of

paracrine secretions resulting from epithelial–mesenchymal interactions in wound repair,

and supports the role of overlying epidermal keratinocytes in modulation of KF

behaviour.

Transforming growth factor-beta has often been considered to be a key player in the

pathogenesis of keloids (Kim et al., 2000). Increased mRNA expression of TGF-β1 and

collagen has been observed in keloids (Kim et al., 2000). Previous studies demonstrated

that KFs produced increased TGF-β (Fusenig et al., 1994; Maas-Szabowski et al., 1999;

Mackenzie et al., 1994). Tissue fibrosis is thought to be the result of excessive ECM

deposition induced by TGF-β which signals fibroblasts to increase the synthesis of matrix

Page 58: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

47

proteins, decrease the production of matrix-degrading proteases and increase the

production of inhibitors of these proteases (Border et al., 1996). There is evidence

suggesting that TGF-β1 can inhibit apoptosis in various types of cells (Smith et al., 1999;

Kim et al., 1998; Genestier et al., 1998). Chodon’s group (2000) reported that

dysregulation in the Fas-mediated apoptosis which occurs in normal wound healing may

be a factor contributing to scar formation and suggested a role for TGF-β1 in this

resistance; specifically that of prolonged proliferative phase due to generation of an

apoptosis resistant phenotype. As the epithelium regulates underlying mesenchymal cell

apoptosis (Hurle et al., 1986), epithelial-mesenchymal interactions could be one factor

responsible for this decreased apoptosis. Funayama and his co-workers (2003)

demonstrated through a similar coculture model that the keloid derived fibroblasts

brought about a greater proliferation and diminished apoptosis when cocultured with NK

and KK. They also demonstrated an upregulation of TGF-β1 in the fibroblasts cocultured

with keloid derived keratinocytes. Xia and his team (2004) demonstrated that the

upregulation of TGF-β1 in a previous study and observed that this was the net effect of an

increased TGF-β1 mRNA transcription in response to paracrine signaling between KK

and KF which was amplified in keloid versus normal cells. Their group postulated that an

aberration in this paracrine regulation in Keloid-derived cells could have resulted in the

generation of a positive feedback loop between KKs and KFs even after reepithelisation,

increasing the expression of TGF-β1 and related factors such as CTGF, which potentiate

TGF-β signaling.

TGF-β1 has also been demonstrated to enhance the contraction of collagen gels in vitro

(Montesano et al., 1998; Finesmith et al., 1990) and wound contraction in vivo

Page 59: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

48

supporting its role in wound contraction. The effect of TGF-β is mainly through the Smad

family of proteins, although a Smad independent pathway involving signaling through

MAPK, ERK, JNK pathways has also been proposed (Derynck et al., 2003). In the

present study, we observed attenuation of FPCL contraction with the addition of anti-

TGF-β1 neutralizing antibody into conditioned media from KK/KF co-culture, indicating

the importance of TGF-β1 in keloid scar contracture.

Myofibroblasts, a group of actin rich fibroblasts, are known to play an important role in

wound contraction. Wound contraction is a very important step in wound repair involving

the conversion of proto-myofibroblasts to differentiated myofibroblasts by the production

of α-SMA in order to generate more force for contracture (Tomasek et al., 2002; Roy et

al., 2001). In normal wound repair, these myofibroblasts disappear by apoptosis but in

pathological conditions the myofibroblasts persist and continue to remodel the ECM,

resulting in connective tissue contracture (Tomasek et al., 2002). Since increased

expression of TGF-β1 directly induces α-SMA expression (Desmouliere et al., 1993;

1995; Ronnov-Jessen et al., 1993) and epithelial–mesenchymal interactions modulate

TGF-β1 expression, we hypothesized that epithelial–mesenchymal interactions could also

indirectly modulate α-SMA expression in vitro. In the present study

immunohistochemical analysis of α-SMA expression by KFs and NFs exposed to

conditioned media from KK/KF coculture and non-coculture conditioned media controls,

demonstrated increased expression of α-SMA in both KF and NF groups, supporting the

hypothesis.

Page 60: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

49

To summarize, it has been demonstrated that the paracrine output from epithelial-

mesenchymal interactions between KKs and KFs modulate contraction of FPCL, strongly

suggesting its role in scar and wound contraction. TGF-β1 is likely to play an important

role in contraction of FPCL induced by epithelial-mesenchymal interactions. These data

suggest a therapeutic approach involving targeting the epidermal layer of the keloid

rather than its deeper constituents for the treatment of this difficult condition.

Page 61: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

50

4 ROLE OF ACTIVIN-FOLLISTATIN SYSTEM IN KELOID PATHOGENESIS

Page 62: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

51

4.1 INTRODUCTION

The bioactivity of fibroblasts in fibrogenesis or excessive scar formation is regulated by a

number of growth factors. The transforming growth factor-β (TGF-β) family, comprising

the TGF-β’s, bone morphogenetic proteins (BMPs) and activins, is thought to play a

central role by regulating fibroblast proliferation, differentiation and matrix production

(Derynck et al., 2003; Kim et al., 2000; Niessen et al., 1999; Tuan et al., 2000). Although

a series of studies has demonstrated the importance of TGF-β as a key player in keloid

pathogenesis, the function and role of activin-A have yet to be established.

Activins, like other members of the TGF-β superfamily, are dimeric proteins consisting

of two βA subunits (activin-A), two βB subunits (activin-B) or a βA and a βB subunit

(activin-AB) (Massague et al., 1990; Vale et al., 1990). Activin-A signals through

heterodimeric complexes of two receptor types - type I receptors, also called activin

receptor-like kinases (ALKs), and type II receptors which are transmembrane serine–

threonine kinases (Mathews et al., 1993). The biological action of activin-A is also

regulated by follistatin, a soluble activin-binding glycoprotein, which inhibits activin’s in

vitro and in vivo functions (Massague et al., 1990; Phillips et al., 2003).

Activin-A was discovered initially as a regulator of pituitary function (Vale et al., 1986).

Subsequent research has shown it to affect proliferation and differentiation of various cell

types. For example, it has been shown to stimulate proliferation of lung fibroblasts and

their differentiation into myofibroblasts (Ohga et al., 1996). It has also been

demonstrated to play a role in the proliferation and differentiation of keratinocytes in

vitro (Seishima et al., 1999). Activin-A inhibits proliferation of vascular endothelial cells

Page 63: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

52

(Mathews et al, 1994; McCarthy et al., 1993; Kozian et al, 1997) and has been shown to

cause cell death in primary hepatocyte cultures (Schwall et al., 1993), B cell hybridomas

and mouse and human myeloma cells (Nishihara et al., 1993, 1995).

The first evidence for a role of activin-A in wound healing came from the studies by

Hübner’s group (1996). They demonstrated an increased induction of activin-A

expression within 24 hr after injury, which remained high until the end of the repair

process. Transgenic mice, which overexpressed the activin-βA subunit specifically in the

epidermis, had epidermal hyperthickening and dermal fibrosis (Munz et al., 1999). After

skin injury, these mice showed enhanced granulation tissue formation with strong

deposition of ECM below the hyperproliferative keratinocytes. By contrast, reduced

granulation tissue formation and thus delayed healing was observed in mice

overexpressing the activin antagonist, follistatin, in the epidermis (Wankell et al., 2000).

Several studies have implicated activin-A as an important player in fibrotic disorders.

Sugiyama’s team (1998) demonstrated an upregulation of activin-A in cirrhotic and

fibrotic rat livers. Furthermore, there was an increased expression of activin-A in various

pulmonary conditions associated with interstitial pulmonary fibrosis (Matsuse et al.,

1996). Activin-A has also been reported to act as a profibrotic cytokine in renal disease

(Gaedeke et al., 2005). Thus, although in vitro studies suggest that activin-A may play a

role in the repair of the mesenchyme and epithelium, a disruption in the control of

activin-A expression could lead to the development of tissue fibrosis.

The present study characterized the role of activin-A synthesis and signaling in keloid

pathogenesis. It evaluated the activin-follistatin system in regulating cellular proliferation

Page 64: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

53

and expression of ECM components in keloid scars, using an extensive array of

techniques such as RNAse protection assay, immunohistochemistry, and co-cultures.

Overall, the expression and production of activin-A and its biological regulator,

follistatin, were perturbed in the development of human keloids.

4.2 RESULTS • Increased activin-A and follistatin in the basal layer of epidermis in keloid

tissues

Immnohistochemical analysis on paraffin sections of normal skin tissues and keloid scar

tissues was performed as previously mentioned. The sections were probed with

antibodies against activin-A (Oxford Bio-Innovations, Oxford, UK) and follistatin (Gift

from Dr David Phillips, Monash University, Melbourne, Australia) followed by

secondary antibodies. Both activin-A and follistatin were found to be localized to the

basal layer of the epidermis in both normal and keloid skins. However, an increased

intensity of these proteins was observed in keloid tissues compared with normal skin

tissues (Fig. 10). This suggested that the activin-follistatin regulatory system might be

implicated in keloid pathogenesis.

• Keloid scars expressed higher levels of activin-A mRNA compared with normal

skin

The expression profile of activin-A was studied using a RNase protection assay in keloid

scars and normal skin tissues (Fig. 11). Normal skins and treated scars expressed very

Page 65: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

54

low levels of activin-A mRNA. In contrast, keloid scar samples had abundant levels of

activin-A mRNA.

• Increased production of activin-A and follistatin by KF.

NF and KF were cultured and conditioned media from these cultures were collected.

Activin-A was measured in conditioned media samples using a specific enzyme linked

immunosorbent assay (ELISA) (Knight et al., 1996) according to the manufacturer’s

instructions (Oxford Bio-Innovations, Oxfordshire, UK) with some modifications for

media samples as described previously (Buzzard et al., 2003). The average intra-plate %

coefficient of variation (CV) was 8.1% (n = 4 plates) and the inter-plate %CV was 6.4%.

The lower limit of detection was 0.01 ng/ml. Follistatin concentrations were measured

using a discontinuous radioimmunoassay as described previously (O’Connor et al., 1999)

with modifications for media samples. Conditioned media showed a linear dose-response

curve which was parallel to the standard. The average intra-assay %CV was 7.7% (n = 3

assays), the inter-assay %CV was 6.4% and the limit of detection was less than 1.94

ng/ml. When cultured in isolation, KFs demonstrated a 29-fold higher secretion of

activin-A compared with NFs (P < 0.0001; Fig. 12a). In contrast, NK produced

significantly higher activin-A than KK (P < 0.0001; Fig. 12a). For follistatin, there were

significant differences in secreted levels between KK and NK (P=0.019; Fig. 12b); KFs

demonstrated a 5-fold increase in secreted levels of follistatin compared with NFs (P <

0.0001; Fig. 12b).

Page 66: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

55

• Over-expression of activin-A in HaCaT keratinocytes increased the proliferation

of underlying fibroblasts during co-culture

As the activin-follistatin regulatory system was shown to be upregulated in keloid tissues,

we determined if HaCaT keratinocytes overexpressing activin-A (hβAHaCaT) modulated

underlying fibroblasts in a paracrine manner. Normal human dermal fibroblasts

cocultured with these overexpressing hβAHaCaT keratinocytes (Gift from Prof Sabine

Werner, ETH, Zurich, Switzerland) demonstrated a 66% increase in proliferation

compared with non-cocultured NFs (P < 0.0001; Fig. 13). In comparison NFs cocultured

with neoHaCaT cells (cells transfected with the empty plasmid) (Gift from Prof Sabine

Werner, ETH, Zurich, Switzerland) demonstrated only a 30% increase (P < 0.0001; Fig.

13).

• Activin-A increased proliferation in NFs and KFs and stimulated expression of

ECM components

Given that keratinocytes overexpressing activin-A stimulated the proliferation of

underlying fibroblasts, the direct effect of activin-A on proliferation and ECM production

were tested. Treatment of NFs and KFs with activin-A (100 ng/ml) resulted in a 23 % (p

< 0.001) and 46% (p < 0.0001) increase, respectively, in proliferation compared with

untreated controls (Fig. 14). This was comparable to the proliferation brought about by

treatment with TGF-β1, although at a lower dose (10 ng/ml).

It was then determined if activin-A and follistatin affected the production of ECM

components like collagen, fibronectin and α-SMA in NFs and KFs. Using Western

Page 67: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

56

blotting, an increase in collagen and α-SMA was observed in both NF and KF treated

with activin-A compared with untreated cells (Fig. 15a, 15b, 15c, 15d, 15g, 15h).

Elevated collagen and fibronectin levels in the conditioned medium in response to

activin-A was confirmed in KFs (Fig. 15e, 15f). To assess the neutralizing ability of

follistatin in this system, exogenous follistatin was co-administered with activin-A which

resulted in a marked reduction in the expression levels of collagen and α-SMA.

Follistatin treatment alone, unlike activin-A, significantly reduced the secreted levels of

collagen, strongly suggesting that follistatin was blocking the stimulatory effects of

endogenously produced activin-A (Fig. 15e). Activin when combined with TGF-β did not

show significant difference in expression of both collagen and α-SMA when compared to

activin only treatment (Fig 15g, Fig 15h).

Page 68: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

57

Figure 10: Increased localization of activin-A and follistatin in the basal layer of epidermis in normal and keloid tissues. Paraffin sections of normal (a, b) and keloid (c, d) skin tissues were prepared and labeled with antibodies specific for activin-A (a, c) and follistatin (b, d) and observed with microscopy (100x). In each panel, the insert shows the same tissue labeled with a non-immune mouse antibody of the appropriate immunoglobulin isotype instead of the specific activin or follistatin antibody. Keloid tissues had more intense labeling in the basal layer of the epithelium compared with normal tissues. The localizations of activin-A and follistatin (or its lack in the negative control inserts) is shown by arrows. The dermis and epidermis are represented by ‘D’ and ‘E’, respectively

(a

(c

(b

(d

E

E

E

E

D

D D

D

E

E

E

D

D D

E

D

20µm 20µm

20µm 20µm

Page 69: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

58

Figure 11: Expression of endogenous activin-βA mRNA derived from normal and keloid tissues. RNA was isolated from normal and keloid human tissues and analyzed by RNase protection assay for the presence of activin-A mRNA. Lanes 1, 2, 12: NS1, NS2, NS8 (normal skin). Lanes 3, 4, 5, 6, 7: KS4, KS5, KS6, KS7, KS11 (keloid scars from burn patients). Lane 8, 9: Keloid treated-skin before surgical excision. Lane 10, 11: KS5, KS19 (keloid scars from earlobe).

Page 70: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

59

Figure 12: Elevated levels of activin-A obtained from keloid fibroblast cultures Conditioned media obtained from NF (NF1,NF3,NF4,NF5,NF9), KF (KF16,KF31,KF32,KF43,KF48), NK (NK1,NK3,NK4,NK5,NK9) normal and KK (KK16,KK31,KK32,KK43,KK48) were assayed using immunoassays specific for activin-A (a) or follistatin (b). Data are presented as mean ± SD.

Page 71: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

60

Figure 13: Increased proliferation of NF when cocultured with activin-A overexpressing hβAHaCaT cells Immortalized hβAHaCaT cells overexpressing the βA subunit of activin (striped bar) and neoHaCaT cells with only the resistance plasmid (solid bar) were co-cultured with normal NF. The control cultures (open bar) comprised of non-cocultured KF. Data are means ± SD.

Page 72: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

61

Figure 14: Increased proliferation of NF and KF treated with rhActivin-A Cultures of NF (a, b) or KF (c, d) were grown until 50% confluence and then serum starved for 48 hrs. The fibroblasts were then treated with activin-A (100 ng/ml, solid bars) TGF-β1 (1 ng/ml dotted bars), TGF-β1 (10 ng/ml, grey bars) then subjected to MTT proliferation assay. Untreated samples were used as control (open bars). Values are mean ± SD absorbance at 570 nm as a percentage of control.

Page 73: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

62

Page 74: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

63

Figure 15: Effect of activin-A, follistatin or TGF-β1 on collagen, fibronectin and α-SMA expression NF or KF were cultured in the absence or presence of different concentrations of activin-A and TGF-β1. Cells were harvested, lysed and both cell lysate and conditioned media collected for Western blot analysis for collagen, fibronectin or α-SMA. (a) and (b) Lane 1: Untreated NF; Lane 2: NF + activin-A; Lane 3: NF + TGF-β1; Lane 4: NF4 + follistatin. (c) and (d) Lane 1: Untreated KF; Lane 2: KF + activin-A; Lane 3, 4: KF + TGF-β1; Lane 5: KF + follistatin; Lane 6: KF + follistatin + activin-A. (e) and (f) Lane 1: Untreated KF; Lane 2: KF + activin-A; Lane 3: KF + TGF-β1; Lane 4: KF + follistatin. All of the blots were probed with a β-actin antibody to confirm equal loading. For conditioned media 4 ml of conditioned media was concentrated from each treatment group to 100 µl which was used for analysis. Histograms represent % OD volume of the bands in the corresponding blots. Solid bars represent collagen; striped bars represent α-SMA; Open bars represent fibronectin. (g) Lane 1: Untreated KF; Lane 2: KF + activin-A; Lane 3: KF + TGF-β1; Lane 4: KF + activin-A (100 ng/ml) + TGF-β1 5 ng/ml. (h) Lane 1: Untreated KF; Lane 2: KF + activin-A 100 ng/ml; Lane 3: KF + activin-A 50 ng/ml; Lane 4: KF + activin-A (100 ng/ml) + TGF-β1 5 ng/ml; Lane 5: KF + TGF-β1 10 ng/ml.

Page 75: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

64

Page 76: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

65

Page 77: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

66

Page 78: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

67

4.3 DISCUSSION Activin-A, a member of the TGF-β superfamily, has been detected in high levels in

various inflammatory (Jones et al., 2004) and fibrotic disorders (Hübner et al., 1996;

Sugiyama et al., 1998; Matsuse et al., 1996, Gaedeke et al., 2005). To confirm its role in

keloid formation, the present study showed that activin-A was indeed overexpressed in

keloid tissues. For example, immunolocalization of activin-A was found in the basal

layers of the epithelium in human skin tissue, but interestingly, keloid tissues showed

more intense immunolocalization compared with normal tissues. Since epithelial–

mesenchymal interactions have recently been explored as a possible important factor

contributing to excessive scar formation, in our experimental set-up although there is no

staining for the fibroblasts, one cannot rule out the paracrine effect of Activin- A

expressed by KK on KF.

The activin-follistatin system is an important regulatory system controlling cellular

proliferation and differentiation in many epithelio-mesenchymal organs, including the

kidney, prostate, mammary gland, lung, pancreas, and salivary gland (Ritvos et al.,1995;

Hildén et al., 1994; Roberts et al., 1994; Tuuri et al., 1994; Cancilla et al., 2001; Liu et

al., 1996; Zhao et al., 1996; Miralles et al., 1998; Furukawa et al., 1995; Yamaoka et al.,

1998; Maldonado et al., 2000; Bläuer et al., 1996). When the localization of follistatin in

keloid and normal tissues was investigated, it was found to have similar pattern to that of

activin. The increase in follistatin in the basal layer of keloid tissue is likely due to a

short-loop negative feedback mechanism operative between activin-A and follistatin

(Phillips et al., 1998). Taking into consideration the localization and expression of both

Page 79: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

68

activin-A and follistatin in the basal layer of the epidermis, where mitotically-active

keratinocytes are present, one could hypothesize that the activin-follistatin system

modulates keratinocyte dynamics in tissue repair and keloid pathogenesis (Werner et al.,

2001).

Activin-A, in addition to being a potent activator of fibroblasts, has also been shown to

have an antiproliferative effect on various types of cells, including epithelial cells,

lymphocytes, prostate cancer cells, and others (Chen et al., 2002). However, it has been

observed that many carcinoma cells are somehow able to escape the negative regulatory

action of activin-A by activating production of follistatin (Dowling et al., 2000;

Rossmanith et al., 2002). In this study, activin-A was secreted in large amounts by KF

when compared with NF but KK secreted less. Interestingly, KK secreted more follistatin

than NK, suggesting that a mechanism similar to that seen in carcinoma cell types might

be active in KK, where these cells were secreting more follistatin to neutralize the

activin-A.

Previous work has demonstrated a large amount of ECM deposition below activin-A

over-expressing keratinocytes (Munz et al., 1999) suggesting that an autocrine, paracrine

and/or endocrine effect of activin-A might be active in the process of wound healing. In

the present study, we cocultured activin-A overexpressing keratinocytes (hβAHaCaT

cells) with NF. An increased proliferation in fibroblasts exposed to hβAHaCaT cells was

observed as compared to those exposed to both non-cocultured and neo HaCaT cells.

This could be due to the increased paracrine levels of activin-A in the hβAHaCaT /NF

coculture system as compared with NK cocultures.This data demonstrated that activin-A

Page 80: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

69

secreted from keratinocytes exerted a paracrine effect on the fibroblasts and that an

excess of activin-A might lead to over-proliferation of these cells, thus leading to fibrosis.

While analyzing this data one has to take in account that HaCaT cells in which activin-A

has not been overexpressed, has an endogenous level of this protein and several other

proliferative factors like IGF-1 (Insulin-like growth factor-1), TGF-β (transforming

growth factor-beta), interleukins and others. Hence one would expect them to also

increase the proliferation of the fibroblasts. Other groups have also observed such effects

when coculturing with neo\HaCaT cells. Skobe’s group (1998) observed that when

cocultured with HaCaT neo cells, fibroblasts show a 75% increase in cell numbers.

As further substantiation of a pro-proliferative effect of activin-A on human dermal

fibroblasts, exogenous activin-A increased proliferation of both NF and KF, consistent

with results from other groups (Ohga et al., 1996; Yamashita et al., 2004). Since TGF-β1

is known to be an important player in fibrotic disorders and has been demonstrated to be

highly expressed in KFs (Lee et al., 1999; Younai et al., 1994; Polo et al., 1999), it was

tested if activin-A and TGF-β1 might act synergistically to stimulate fibrotic processes.

However, co-treatment of cells with both activin-A and TGF-β1 showed no significant

synergistic effect.

ECM components are involved in many cellular processes and their patterns of

expression is different from that usually present in normal skin during different stages of

wound healing (Costa et al., 1999). Tissue fibrosis is thought to be partly the result of

excessive induction by TGF-β1, which signals fibroblasts to increase the synthesis of

ECM, decrease the production of matrix-degrading proteases and increase the production

Page 81: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

70

of the inhibitors of these proteases (Border et al., 1994). NFs and KFs, when exposed to

activin-A, increased expression of cytoplasmic collagen (procollagen). Procollagen

further undergoes post translational modifications (hydroxylation, glycosylation) and

subsequently forms a triple helix and is secreted out of the cell into the conditioned

medium. It was observed that there was an increased concentrations of secreted collagen

in the conditioned media from KFs when treated with activin-A, underscoring the

importance of activin-A in the regulation of collagen production by dermal fibroblasts in

keloid pathogenesis.

In addition, there was an increase in the expression of α-SMA, a phenotypic marker for

myofibroblasts (Yamashita et al., 2004), suggesting activin’s importance in regulating

expression and contractile activities of α-SMA. Not surprisingly, exogenous follistatin

had no effect on the expression of ECM components directly, but blocked the activin-

induced increases. Follistatin antagonizes the effect of activin-A by binding to activin

itself, rendering it inactive (Sugino et al., 1997; Phillips et al., 1998).

In summary, the present study demonstrated that activin-A was upregulated in keloid

tissues. Keratinocytes overexpressing activin-A stimulated proliferation of dermal

fibroblasts, suggesting a paracrine role to promote fibroblast proliferation, induce the

expression of α-SMA, and enhance collagen formation in KFs. However, follistatin, an

antagonist of activin-A, blocked the expression of ECM components. Thus, the findings

strongly suggest that activin-A is a potent inducer of fibroblast activation and involved in

the pathogenesis of keloids. They also emphasize the importance of follistatin in

Page 82: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

71

regulating activin-A bioactivity and suggest a possible therapeutic potential of follistatin

in the treatment and prevention of keloids (Fig 16).

Page 83: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

72

Figure 16: Activin-A expression increased by epithelial-mesenchymal interactions resulting in a fibrotic phenotype. KK expressed elevated levels of activin-A which has a paracrine effect on underlying fibroblasts thus increasing their proliferative potential and ability to produce ECM components which are charecteristics of a fibrotic phenotype. Follistatin can bind to activin-a and render it inactive thus having a potential as a possible therapeutic agent.

FOLLISTATIN THERAPY

KELOID SCAR

Proliferation

Collagen

Fibronectin

α-SMA

KELOID KERATINOCYTE

KELOID FIBROBLAST

+ +

Activin-A

Activin-A receptor

Page 84: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

73

5 ROLE OF PROTEOGLYCANS IN KELOID PATHOGENESIS

Page 85: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

74

5.1 INTRODUCTION

The TGF-β family of growth factors has been widely studied and is thought to play a

central role in tissue regeneration by regulating fibroblast proliferation, differentiation

and matrix production (Derynck et al., 2003; Kim et al., 2000; Niessen et al., 1999; Tuan

et al., 1998). However various other soluble and insoluble effectors are equally important

in wound repair and may thus play a part in keloid formation. One such effector group

comprises cell surface proteoglycans, namely the syndecans, which are a family of

transmembrane heparan sulphate proteoglycans that are differentially expressed during

development and wound repair (Carey et al., 1997; Gotte et al., Lories et al., 1992).

Owing to their unique function of integrating signaling from circulating growth factors

and ECM proteins with other cellular receptors like the integrins, they have often been

referred to as “tuners of transmembrane signaling” (Zimmermann et al., 1999). They

exist as four isoforms: syndecan-1, syndecan-2, syndecan-3 and syndecan-4, each having

characteristic cell- and tissue-specific distributions throughout development (Bernfield et

al., 1993; Kim et al., 1994).

Syndecan-2, also called fibroglycan, is especially abundant in mesenchymal cells (Gallo

et al., 2000) and has been shown to play an important role in diverse biological processes.

Mouse cells of mesenchymal origin in kidney, lung, and stomach and also in cells that

form cartilage and bone (David et al., 1993) are known to express high levels of

syndecan-2. Syndecan-2 modulates cell behavior by affecting cell adhesion and signaling

(Essner et al., 2006). Chen and his co-workers (2004) demonstrated that syndecan-2

regulates TGF-β signaling by binding to it through the core protein of its ectodomain. It

Page 86: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

75

has been shown to interact with various other cytokines and growth factors, like FGF-2

and VEGF, and mediate their effects on promoting mesenchymal tissue growth and

angiogenesis (Burgess et al., 1989; Leung et al., 1989; Besner et al., 1990; Clasper et al.,

1999).

Syndecan-2 has also been implicated in various types of cancers, with increased

expression demonstrated in human ovarian carcinoma biopsies (Davies et al., 2004) and

other cancer cell lines (Gulyas et al., 2004). It has also been shown to modulate colon

cancer cell adhesion, motility and proliferation (Park et al., 2002; Han et al., 2004).

Owing to these properties, syndecan-2 has recently become a subject of widespread

research. Its role in carcinogenesis, embryogenesis, wound healing and other clinical and

physiological conditions is being thoroughly explored.

In addition to heparan sulphate proteoglycans, soluble chondroitin and dermatan sulphate

proteglycans like decorin have also been shown to play a role in cell-matrix interactions.

Decorin is an abundant component of bone and skin ECM and associates with type I, II

and VI collagen fibrils as well as fibronectin in vivo (Laine et al., 2000; Lozzo et al.,

1996; 1998). Decorin is expressed abundantly in NF in which its expression has been

shown to be enhanced by IL-1 and dexamethasone and inhibited by TGF-β (Laine et al.,

2000; Heino et al., 1988; Kähäri et al., 1995; 1991). Unlike the syndecans, decorin

inhibits the biological activity of TGF-β by binding with the core protein in various cell

types (Hilderbrand et al., 1994) and thus has been explored as a potential antifibrotic

agent. Its antifibrotic effect has been demonstrated in experimental kidney, lung, cerebral

Page 87: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

76

and muscular fibrosis (Border et al., 1992; Giri et al., 1997; Logan et al., 1999;

Fukushima et al., 2001)

In addition to modulating cell-matrix interaction, decorin has also been shown to have an

antiproliferative effect on various carcinoma cells. The antiproliferative effect of decorin

was demonstrated to be mediated by either the upregulation of cyclin-dependent kinase

inhibitors like p21 and p27 or by interacting with various growth factors (De Luca et al.,

1996; Fischer et al., 2001). It was seen that malignant transformation of cells resulted in

loss of decorin expression. However when decorin expression was restored, it caused

malignant cell growth arrest, highlighting its protective effect on cellular dynamics

(Lozzo et al., 1999; Santra et al., 1995).

In the present study, intrinsic syndecan-2, FGF-2 and decorin expressions were studied in

keloid tissues by performing immunohistochemical analysis and western blot assays. The

role of serum on the expression profile of syndecan-2 and decorin was further

investigated by stimulating the NF and KF with 10% fetal calf serum. The role of FGF-2

in shedding of the syndecan-2 ectodomain and its implications in modulating cell

behavior was studied by treating KF cultures with recombinant FGF-2 and

immunoblotting the conditioned media for the expression of syndecan-2. The role of

epithelial-mesenchymal interactions in modulating syndecan-2, FGF-2 and decorin

expression dynamics was investigated using an established two-chamber serum-free

coculture model. In addition, the antifibrotic effect of decorin was studied by

investigating its effect on the expression of ECM components such as collagen,

fibronectin and α-SMA.

Page 88: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

77

5.2 RESULTS • Keloid scar intrinsically expressed higher levels of syndecan-2 and FGF-2 and

lower levels of decorin

The expression profiles of syndecan-2, FGF-2 and decorin in keloids and normal skin

tissues were studied using a western blot assay. Normal skin expressed low levels of

syndecan-2 (Fig. 17) and FGF-2 (Fig. 22) however decorin was abundantly expressed

(Fig. 25). In contrast, keloid scar samples had abundant levels of syndecan-2 (Fig. 17),

FGF-2 (Fig. 22) and low levels of decorin (Fig. 25). Although decorin was

downregulated in keloid tissues a 32kda band was observed in this sample which was

completely missing in normal tissue.

• Increased localization of FGF-2 seen in the basal epidermis and dermis of

keloids compared to normal skin

Paraffin sections of normal skin and keloid scar were subjected to immunohistochemical

analysis for the expression of FGF-2. Increased levels of FGF-2 were observed in the

dermis and basal epidermis of keloid scar tissues compared to normal skin (Fig. 21ab).

FGF-2 also stained strongly in keloid dermis whereas the dermis of normal skin

demonstrated significantly diminished or negative staining (Fig. 21c d).

Page 89: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

78

• Serum growth factors upregulated syndecan-2 and downregulated decorin

expression in KF

Stimulation with 10% FCS is interpreted by fibroblasts as a wound healing signal, thus

NF and KF were treated with 10% FCS to investigate if syndecan-2 and decorin were

involved in the initial phases of wound healing. Both NFs and KFs showed increased

levels of syndecan-2 (Fig. 18) on stimulation with serum growth factors, suggesting a

possible role of syndecan-2 in the initial wound healing mechanism. However, decorin

expression was downregulated on stimulation with serum (Fig. 26).

• Coculture of KK with KF increased levels of syndecan-2 in the conditioned

media

To investigate if epithelial-mesenchymal interactions modulate syndecan-2 expression, a

coculture model was set up wherein NK and KK were cocultured with NF and KF

respectively. Fibroblasts were subsequently lysed and total cell extracts were subjected to

western blot. Although no difference was observed in cellular levels of syndecan-2 in

both NFs AND KFs cocultured with their respective keratinocytes (Fig. 19a), an

increased level of syndecan-2 was observed in the conditioned media of KK/KF

cocultures compared to KFs in monoculture (Fig. 19b) suggesting the possible cleavage

of syndecan-2 in KFs upon coculture with KKs.

Page 90: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

79

• Cocultured keratinocytes and fibroblasts demonstrated increased expression of

FGF-2 compared to cells in monoculture

In order to investigate the effect of epithelial-mesenchymal interactions on the dynamics

of FGF-2 expression, NK and KK were cocultured with NF and KF respectively, with

cell lysates collected from both groups and blotted for FGF-2 expression. An increase in

FGF-2 expression was observed in both cocultured keratinocytes (Fig. 23a) and

fibroblasts (Fig. 24) as compared to those in monoculture, highlighting a possible

autocrine effect of FGF-2 on keratinocytes as well as a paracrine effect on the underlying

fibroblasts. In addition, cocultured KKs expressed higher levels of FGF-2 as compared to

cocultured NKs (Fig. 23b) suggesting a more active mechanism within KKs compared to

NK.

• Increased levels of decorin found only in the conditioned media from NK/NF

cocultures as compared to monocultures

Although conditioned media obtained from the coculture of NF with normal NK

demonstrated higher levels of decorin compared to normal fibroblast monocultures (Fig

27a), no significant difference in decorin levels was observed in the conditioned media of

cocultured KFs as compared to KFs in monoculture (Fig 27b) suggesting a loss KK

control of underlying KF decorin expression.

Page 91: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

80

• rhFGF-2 increased syndecan-2 cleavage and thus its detectable levels in KF

conditioned media

The role of FGF-2 in the cleavage of fibroblast syndecan-2 resulting in its increased

levels in conditioned media was investigated by treating KFs with exogenous FGF-2 and

analyzing the conditioned media for expression of syndecan-2 by western blot. A

significant increase in the levels of syndecan-2 was observed compared to untreated KFs

(Fig. 20).

• Decorin decreased the expression of ECM components in both NF and KF

To investigate the effect of decorin on the expression of ECM components decorin at

various concentrations (250 ng/ml, 500 ng/ml, 1000 ng/ml and 2000 ng/ml) was added to

both NF and KF after which cell lysates and conditioned media were analyzed for

collagen, fibronectin and α-smooth muscle actin expressions. A decrease in the

expressions of the ECM components was observed both in the cell lysate (Fig. 28a b) and

conditioned media (Fig. 28c).

Page 92: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

81

Figure 17: Elevated levels of syndecan-2 in tissue extracts obtained from keloid tissue. Expression of syndecan-2 in normal and keloid skin tissues were analyzed by western blot assay. Study groups comprised of normal skin and keloids. Tissue specimens were sonicated in lysis buffer and 100 µg of total protein extracts were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-syndecan-2 polyclonal antibody Lane 1-5: Keloid scar (K50, K51, K52, K53, and K54). Lane 6-8: Normal skin (N31, N32, N33). Equal loading was confirmed by blotting with β-actin.

Page 93: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

82

Figure 18: Serum growth factors upregulated syndecan-2 expressions in NF and KF. Expression of syndecan-2 by fibroblasts after exposure to 10% FCS was detected by western blot assay. Fibroblasts were seeded in 6-well plates at a density of 1 × 104 cells/ml in DMEM/10% FCS for 24 hrs and subsequently in serum-free medium for another 48 hr. The fibroblasts were subject to either DMEM/10% FCS or serum-free DMEM for 24 hr. Fibroblasts were lysed and 100µg total protein cell lysates were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-syndecan-2 polyclonal antibody. Lane 1: NF4+DMEM, Lane 2: NF4+DMEM/10% FCS, Lane 3: NF14+DMEM, Lane 4: NF14+DMEM/10%FCS, Lane 5: KF45+DMEM, Lane 6: KF45+DMEM/10%FCS, Lane 7: KF49+DMEM, Lane 8: KF49+DMEM/10% FCS. Equal loading was confirmed by blotting with β-actin for cell extracts

Page 94: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

83

Figure 19: Increased ectodomain shedding of syndecan-2 is observed in keloid cocultures as compared to monocultures. a) Cell lysate obtained from NFs and KFs cocultured with NKs and KKs was assayed by western blot for syndecan-2 expression. Cell lysates from NF and KF in monoculture were used as controls. No difference in the expression of syndecan-2 was observed in cocultured fibroblast cell extracts compared to fibroblasts in monoculture. Lane 1: NF1 non cc, Lane 2: NK1/NF1, Lane 3: NF2 non cc, Lane 4: NK3/NF2, Lane 5: KF 45 non cc, Lane 6: KK45/KF45, Lane 7: KF 48 non cc, Lane 8: KK48/KF48. b) Conditioned media from KF cocultures were assayed by western blot for syndecan-2 expression. Conditioned media from KFs in monoculture were used as controls. Equal volumes of conditioned media were subjected to SDS-PAGE. Conditioned media from KF cocultures shed more syndecan-2 into the conditioned media as compared to the KF monocultures. Lane 1: KF48 non cc, Lane 2: KK48/KF48, Lane 3: KF45 non cc, Lane 4: KK45/KF45, Lane 5: KF24, Lane 6: KK24/KF24.

Page 95: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

84

Figure 20: Exogenous rhFGF-2 stimulated shedding of syndecan-2 from fibroblast cell surface into the conditioned media. rhFGF-2 (10 ng/ml) was added to the fibroblast cultures and conditioned media assayed for the expression of syndecan-2. Conditioned media from untreated KFs were used as controls. Equal volumes of conditioned media were subjected to SDS-PAGE. KFs treated with FGF-2 shed more syndecan-2 as compared to untreated fibroblasts. Lane 1: KF43 non cc, Lane 2: KF43+FGF-2, Lane 3: KF29 non cc, Lane 4: KF29+FGF-2.

Page 96: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

85

Figure 21: Increased localization of FGF-2 in the basal layer of epidermis and dermis in keloid tissues. Paraffin sections of normal (a, c) and keloid (b, d) tissues were prepared and labeled with antibodies specific for FGF-2 and observed with microscopy (40x). In each panel, the insert shows the same tissue labeled with a non-immune rabbit antibody of the appropriate immunoglobulin isotype instead of the specific FGF-2 antibody. Keloid tissues had more intense labeling in the basal layer of the epithelium and the dermis compared with normal skin. The localization of FGF-2 (or its lack in the negative control inserts) is shown by an arrow. The dermis and epidermis are represented by ‘D’ and ‘E’, respectively.

10 µm 10 µm

10 µm 10 µm

Page 97: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

86

Figure 22: Elevated levels of FGF-2 in tissue extracts obtained from keloid tissue. Expression of FGF-2 in normal and keloid skin tissues were analyzed by western blot assay. Study groups comprised of normal skin and keloids. Tissue specimens were sonicated in lysis buffer and 100 µg of total protein extracts were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-FGF-2 polyclonal antibody Lane 1-8: Keloids (K50, K51, K52, K53, K54, K55, K56 and K57). Lane 9-13: Normal skin (N31, N32, N33, N34, N35). Equal loading was confirmed by blotting with β-actin.

Page 98: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

87

Figure 23: Cocultured NK and KK expressed increased levels of FGF-2 as compared to monocultured keratinocytes. Cell lysates from monocultured NK and KK were extracted and analyzed by western blot for the expression of FGF-2. Similarly, NKs and KKs which had been cocultured with NFs and KFs respectively were lysed and cell extracts were analyzed by western blot for the expression of FGF-2. a) Lane 1: NK1, Lane 2: NK1/NF1, Lane 3: KK48, Lane 4: KK48/KF48, b) Lane 1: NK1/NF1, Lane 2: NK2/NF2, Lane 3: NK3/NF3, Lane 4: KK48/KF48, Lane 5: KK45/KF45, Lane 6: KK49/KF49. Equal loading was confirmed by blotting with β-actin for cell extracts.

Page 99: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

88

Figure 24: Cocultured NF and KF expressed increased levels of FGF-2 as compared to cells in monoculture. Cell lysates from monocultured NFs and KFs were extracted and analyzed by western blot for the expression of FGF-2. Similarly, NFs and KFs which had been cocultured with NKs and KKs respectively were lysed and cell extracts were analyzed by western blot for the expression of FGF-2. Cocultured fibroblasts expressed increased levels of FGF-2 compared to moncoultures. Equal loading was confirmed by blotting with β-actin for cell extracts Lane 1: NF1, Lane 2: NK1/NF1, Lane 3: NF2, Lane 4: NK2/NF2, Lane 5: KF48, Lane 6: KK48/KF48, Lane 7: KF49, Lane 6: KK49/KF49

Page 100: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

89

Figure 25: Downregulation of decorin in tissue extracts obtained from keloid tissue. Expression of decorin in normal and keloid skin tissues were analyzed by western blot assay. Study groups comprised of normal skin and keloids. Tissue specimens were sonicated in lysis buffer and 100µg of total protein extracts were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-decorin monoclonal antibody Lane 1-3: Keloids (K51, K52 and K53). Lane 4-6: Normal skin (N31, N32, N33). Equal loading was confirmed by blotting with β-actin.

Page 101: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

90

Figure 26: Serum growth factors downregulated decorin expressions in NF and KF. Expressions of decorin by fibroblasts after exposure to 10% FCS was detected by western blot assay. Fibroblasts were seeded in 6-well plates at a density of 1 × 104 cells/ml in DMEM/10% FCS for 24 hrs and subsequently in serum-free medium for another 48 hrs. The fibroblasts were subject to either DMEM/10% FCS or serum-free DMEM for 24 hr. Fibroblasts were lysed and 100 µg total protein cell lysates were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-decorin monoclonal antibody. Lane 1: NF4+DMEM, Lane 2: NF4+DMEM/10% FCS, Lane 3: NF14+DMEM, Lane 4: NF14+DMEM/10%FCS, Lane 5: KF45+DMEM, Lane 6: KF45+DMEM/10%FCS, Lane 7: KF49+DMEM, Lane 8: KF49+DMEM/10% FCS. Equal loading was confirmed by blotting with β-actin for cell extracts.

Page 102: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

91

Figure 27: Increased secretory decorin in cocultured NFs and not KFs. NF and KF were cocultured with NK and KK, respectively and conditioned media were collected and analyzed for the expressions of decorin. a) Lane 1: NF4+DMEM, Lane 2: NF1+DMEM, Lane 3: NF14+DMEM, Lane 4: NF3+DMEM, Lane 5: NK1/NF1, Lane 6: NK2/NF2, Lane 7: NK14/NF14, Lane 8: NK5/NF5, b) Lane 1: KF29+DMEM, Lane 2: KF48+DMEM, Lane 3: KF49+DMEM, Lane 4: KF25+DMEM, Lane 5: KK48/KF48, Lane 6: KK45/KF45, Lane 7: KK25/KF25, Lane 8: KK49/KF49

Page 103: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

92

Figure 28: Effect of decorin on collagen, fibronectin and α-SMA expressions. NF or keloid fibroblasts KF were cultured in the absence or presence of different concentrations decorin. Cells were harvested, lysed and both cell lysates and conditioned media collected for western blot analysis for collagen, fibronectin or α-SMA. a) Lane 1: NF1+DMEM, Lane 2: NF1+Decorin (250 ng/ml), Lane 3: NF1+Decorin (500 ng/ml), Lane 4: NF1+Decorin (1000 ng/ml), Lane 5: NF1+Decorin (2000 ng/ml), b) Lane 1: KF48+DMEM, Lane 2: KF48+Decorin (250 ng/ml), Lane 3: KF48+Decorin (500 ng/ml), Lane 4: KF48+Decorin (1000 ng/ml), c) Lane 1: KF48+DMEM, Lane 2: KF48+Decorin (250 ng/ml), Lane 3: KF48+Decorin (500 ng/ml), Lane 4: KF48+Decorin (1000 ng/ml).

Page 104: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

93

Page 105: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

94

5.3 DISCUSSION

The healing of any skin injury involves a spontaneous cascade of events comprising

communication between cells and the release of mediators as growth factors, and

cytokines, leading to inflammation, cell proliferation, collagen-ECM deposition, scar

formation, maturation and ultimately remodeling. Any abnormality or disruption of this

sequence of events may lead to formation of abnormal scarring which may include keloid

scars, characterized by excessive collagen-ECM deposition and often, the formation of

contractures.

Recent advances in this field have seen the role of growth factors being extensively

investigated. Interestingly, researchers have found most of the growth factors and

cytokines involved in the wound healing process to be immobilized at the cell surface and

ECM through binding with proteoglycans (Rouslahti et al., 1991). These proteoglycans in

turn are known to play important roles in various physiological processes. Besides

providing mechanical strength by filling in the space between the collagen and elastin

fibres by absorbing water, they also influence collagen formation, cell proliferation, cell

migration and cell adhesion during wound healing (Pratibha et al., 2000).

Syndecan-2, a cell surface proteoglycan, plays an important role in the transmodulation

of the ECM assembly. Since keloid scars are characterized by an aberrant organization of

the ECM assembly, we investigated if syndecan-2 was being abnormally expressed in

keloid tissues as compared to normal skin tissues. Western blot analysis of the tissue

extracts demonstrated that syndecan-2 was indeed upregulated in keloid tissues and that it

could be one of the factors responsible for the excess deposition of ECM components.

Page 106: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

95

In order to understand the underlying biology of syndecan-2 in wound repair and scar

formations, the expression profiles of syndecan-2 at different stages of the wound healing

process was investigated using in-vitro models. Upon cutaneous injury, the coagulation

cascade is activated and an inflammatory response sets in as the first stage of wound

healing. The active platelet plug sequesters serum in the immediate vicinity providing a

rich source of cytokines/growth factors with paracrine effects on neighbouring cells. Such

exposure of fibroblasts to serum is interpreted to be a physiological wounding signal

(Iyer et al., 1999). It was observed that when NF and KF when exposed to serum growth

factors expressed increased amounts of syndecan-2 suggesting its possible role in the

early stages of the healing process and in the modulation of the ensuing events by

interaction with some of the macrophage-derived growth factors like FGF-2, VEGF, EGF

(Gotte et al., 2003). The hypothesis that syndecan-2 might be an important player in the

early phase of the healing process is further strengthened by numerous studies reporting

syndecan-2 to be induced by proinflamatory cytokines like IL-1α and IL-1β and TGF- β

which are abundant in the early wound environment (Clasper et al., 1999; Worapamorn

et al., 2001; Sebestyen et al., 2000).

Strikingly enough, it has been observed that during development, syndecan-2 is localized

at sites where intensive epithelial-mesenchymal interactions shape and transform the

epithelia and the mesenchyme into morphologically and functionally differentiated

tissues suggesting a possible syndecan-2’s role in modulating epithelial-mesenchymal

interactions (David et al., 1993). Previous findings have shown that epithelial-

mesenchymal interactions play an important role in keloid pathogenesis with KK

Page 107: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

96

modulating NF and KF growth and proliferation (Lim et al., 2001), influencing collagen

expression by both NF and KF, inducing NF to secrete collagen in a keloid-like manner

(Lim et al., 2002), and utilizing the IGF system of mitogens as inductive ligands for this

process. Thus to investigate if these interactions would in anyway affect the expressions

of syndecan-2, NF and KF were cocultured with NK and KK, respectively, and the cell

lysates and conditioned media blotted. Surprisingly, although no difference in the

expressions of syndecan-2 from cell lysateswas observed, a definite increase in syndecan-

2 levels was seen in the conditioned media obtained from KFs cocultured with KK. This

strongly suggests that syndecan-2 was being shed from the KF cell surface into the

conditioned media under the influence of the overlying keratinocytes. Syndecan-1 and -4

have been previously shown to be shed from the surface of mouse epithelia and SVEC4-

10 cells in vitro, and factors released during stress, injury and cancer have been

demonstrated to upregulate this process (Fitzgerald et al., 2000; Elenius et al., 2004;

Bayer et al., 2001; Seidel et al., 2000; Subramanian et al., 1997). Previous studies by

Fears’s group (Fears et al., 2006) highlighted the role of syndecan-2 shedding induced by

various factors, to promote angiogenesis. Of the various factors studied, FGF-2 was

observed to stimulate syndecan-2 shedding from microvascular endothelial cells. When

exogenous rhFGF-2 was added to the KF cultures, syndecan-2 shedding was also

observed, with increased amounts of secreted syndecan-2 found in the conditioned media.

The production and secretion of heparin binding growth factors such as FGF have been

implicated in various macrophage-mediated processes, including tumor angiogenesis,

arteriogenesis and capillary sprouting, wound healing, and inflammation (Clasper et al.,

1999). It would appear that the syndecan-FGF system could also be an important player

Page 108: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

97

in initiating aberrant wound healing processes leading to fibrosis. As rhFGF-2 increased

syndecan-2 shedding in KFs, it was investigated if FGF-2 was intrinsically upregulated in

keloid tissues compared to normal skin and, more importantly, in KK/KF cocultures

where increased levels of syndecan-2 were observed in conditioned media. Increased

expressions and localizations of FGF-2 in keloid epidermis and dermis was found as

compared to normal skin sections. In addition, fibroblasts cocultured with NK and KK

expressed increased levels of FGF-2 as compared to fibroblasts in single cell culture.

Interestingly KK/KF cocultures expressed increased levels of FGF-2 as compared

NK/NF. One arm of keloidogenesis resulting from epithelial-mesenchymal interactions

would thus appear to involve increased FGF-2 levels which in turn stimulate the shedding

of syndecan-2 from fibroblasts. This shed syndecan-2 would not only modulate the

activity of FGF-2 but also other profibrotic growth factors (Han et al., 2004).

FGF-2 has also been demonstrated to decrease decorin mRNA expression in

osteosarcoma cells. The reduced expression of decorin corresponded with increased

collagen production by these cell types (Bodo et al., 2002; Wegrowski et al., 2000).

Since keloids are marked by increased production of type 1 collagen the expression of

decorin in keloid tissues was investigated. As expected a decreased expression of decorin

was observed in keloid tissues as compared to normal skin. Interestingly a 32 kda band

was observed in keloid tissues probably due to the degradation of decorin to an inactive

fragment by various MMP’S known to be present in keloids (Fujiwara et al., 2005).

Previous studies have shown MMP’s to degrade decorin to an inactive fragment which is

then unable to further bind to TGF-β, releasing it from the ECM with resultant increased

activity in vivo (Imai et al., 1997). Further studies will be required to assess if this

Page 109: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

98

mechanism plays a role in keloid pathogenesis. When stimulated with 10% serum, a

reduction in decorin was observed in both NF and KF. This may be due to negative

feedback from increased levels of TGF-β and other growth factors in the serum. Previous

results by Zandvoort’s group (Zandvoort et al., 2006) demonstrated TGF-ß1 stimulation

to decrease decorin production in isolated pulmonary parenchymal fibroblasts of COPD

patients.

The effect of epithelial-mesenchymal interactions in modulating the expression of

decorin in a coculture model was investigated and it was observed that although NK/NF

cocultures expressed increased amounts of decorin compared to monocultures, KK/KF

cocultures did not show any significant difference in decorin levels as compared to

monocultures. This would suggest that the overlying keratinocytes might have directly or

indirectly blocked the mechanisms responsible for modulating fibroblast decorin

expression. FGF-2 demonstrated above to be upregulated in cocultures might well be one

of the factors responsible, and further studies will be required to confirm this hypothesis.

Finally, it was investigated if exogenous administration of decorin could be used as an

anti- fibrotic agent. NF and KF were treated with decorin at different concentrations and

expressions of different ECM components were investigated. It was observed that

collagen was downregulated on treatment in a dose-dependent manner both in the cell

lysates and the conditioned media. In addition α-SMA, a phenotypic marker for

myofibroblasts, and fibronectin were also downregulated on treatment with decorin,

which would suggest the potential utility of decorin against excess fibrosis.

Page 110: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

99

Overall, it was shown that syndecan-2 and FGF-2 were not only overexpressed in keloid

tissues but could also interact with each other resulting in the shedding of syndecan-2

which could in turn activate a whole cascade of events resulting in keloidogenesis (Fig

29). In addition, decorin appeared to be downregulated in keloid tissues, and this protein

with strong antifibrotic effects could have the potential to be used as a therapeutic agent

for keloids.

Page 111: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

100

Figure 29: Ectodomain shedding of syndecan-2 by FGF-2 stimulated by epithelial-mesenchymal interactions resulting in a fibrotic phenotype One arm of keloidogenesis results from epithelial-mesenchymal interactions which appear to involve increased FGF-2 levels which in turn stimulate the shedding of syndecan-2 from fibroblasts. This shed syndecan-2 not only modulates the activity of FGF-2 but also other profibrotic growth factors leading to keloid scars.

Keloid Fibroblasts

Stimulates Release of FGF-2

FGF-2

FGF-2 receptor

Syndecan -2

Keloid Keratinocytes

+ +

ECM

Cleaved syndecan -2 binds to profibrotic factors

KELOID SCAR

Page 112: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

101

6 ROLE OF STEM CELL FACTOR and c-KIT SYSTEM IN KELOID PATHOGENESIS

Page 113: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

102

6.1 INTRODUCTION Stem cell factor (SCF), also known as mast cell growth factor, steel factor or KIT ligand

is a hematopoietic and tissue growth factor that serves as a ligand for the c-KIT oncogene

(Gabrilove et al., 1994). It is widely expressed throughout the body by stromal cells,

fibroblasts and endothelial cells (Ashman et al., 1999) and is detectable at a low level in

the circulation (Broudy et al., 1997; Lyman et al., 1998). KIT (CD117) on the other

hand, coded by proto-oncogene c-KIT, is a 145–160 kDa transmembrane tyrosine kinase

belonging to the type III family of receptor kinases (Yarden et al., 1987). KIT is

expressed in a variety of normal cells and tissues (Natalie et al., 1992; Lammie et al.,

1994) often with its ligand (Williams et al., 1990). Previous studies have demonstrated

expression of c-KIT mRNA or protein by mast cells (Mayrhofer et al., 1987; Majumdar

et al., 1988; Nocka et al., 1989), melanocytes (Nocka et al., 1989), testis (Majumdar et

al., 1988), and in bone marrow (Wang et al., 1989). In addition c-KIT protein expression

has been found in non-haemopoietic cell types, including vascular endothelial cells

(Broudy et al., 1994), astrocytes, renal tubules, breast glandular epithelial cells and sweat

glands (Natalie et al., 1992; Lammie et al., 1994). Binding of SCF to KIT results in

dimerisation of the receptors followed by activation of its intrinsic tyrosine kinase

activity (Blume-Jensen et al., 1991). The interaction between c-KIT and SCF ligand

plays an important role in hematopoiesis, in mast cell development and function,

embryogenesis, proliferation and migration of primordial germ cells and melanoblasts

(Huang et al., 1990; Zsebo et al., 1990).

Page 114: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

103

SCF and its receptor c-KIT also play important roles in the process of wound healing.

SCF-KIT interactions in mast cells trigger degranulation at the site of wounding. The

released mediators increased fibroblast proliferation and collagen synthesis, contributing

to abnormal tissue growth leading to fibrosis in various organs (Garbuzenko et al., 2002;

Fireman et al., 1999; El Kossi et al., 2003). In addition to their role in fibrosis, autocrine

or juxtacrine cycles involving c-KIT and SCF have been demonstrated to be important in

some solid tumors, for example, small cell lung cancer (Krystal et al., 1996; Hibi et al.,

1991).

In the present study, owing to its function in wound healing, we hypothesized a role for

SCF/c-KIT system in keloid scar pathogenesis. Moreover, its upregulation in other

fibrotic disorders compels its investigation as a possible factor responsible for a keloidic

phenotype.The results in this study demonstrated that both SCF and c-KIT were

upregulated in keloid scar tissues in vivo thus corroborating our hypothesis. They were

also upregulated in cultured fibroblasts on stimulation with serum highlighting their

importance in the initial phase of wound healing. In addition it was demonstrated that

epithelial-mesenchymal interactions, mimicked by coculture of keratinocytes and

fibroblasts in vitro, not only stimulated secretion of soluble form of SCF in keloid

cocultures but also brought about shedding of the extracellular domain of c-KIT perhaps

by upregulation of TACE. TACE was also shown to be upregulated in keloid scars in

vivo and keloid cocultures in vitro. In addition the KIT receptor was activated, as

demonstrated by increase in phospho c-KIT, in keloid cocultures as compared to normal

cocultures.

Page 115: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

104

6.2 RESULTS • Elevated expression of SCF, c-KIT, phosphorylated c-KIT (Tyr 703, Tyr 721)

and TACE in keloid scar tissues as compared to normal skin tissues

Stem cell factor (SCF) (Fig. 30) and both mature (145kDa) and soluble (95kDa) forms of

its receptor c-KIT (Fig. 33a) were found to be overexpressed in keloid scar tissue extracts

as compared to normal. In addition phosphorylated forms of c-KIT (tyr 703, tyr 721)

were also over expressed in keloid scar tissues though the difference was significant only

in tyr 721 (Fig. 33b). TACE or ADAM17, a known sheddase of many membrane proteins

was also shown to be expressed at higher levels in keloid scar tissues compared to normal

skin tissues (Fig. 37).

• Increased localization of c-KIT in the keloid epidermis compared to normal skin

Immunohistochemical studies were carried out to investigate the expression of c-KIT and

TACE in normal skin and keloid scar tissues. An increased level of c-KIT expression was

observed in the basal epidermis (rete-ridge) of keloid scar. The normal skin epidermis

demonstrated significantly less or negative staining (Fig. 34a b).

• Serum growth factors upregulated SCF levels in KF

Stimulation with 10% FCS is interpreted by fibroblasts as a wound healing signal thus

NF and KF were treated with 10% FCS to investigate if SCF was involved in the initial

phases of wound healing. Both NF (Fig 31a) and KF (Fig 31b) demonstrated an increase

Page 116: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

105

in levels of SCF on stimulation with serum growth factors suggesting a possible role of

SCF in the initial wound healing mechanism.

• Coculture of KK with KF increased levels of SCF in the conditioned media

To investigate if epithelial-mesenchymal interactions modulate SCF expression, a

coculture model was set up wherein KK were cocultured with KF. Fibroblasts were

subsequently lysed and total cell extracts were subjected to western blot. Although no

difference was observed in cellular levels of SCF in KF cocultured with KK (Fig. 32a),

an elevated level of SCF was observed in the conditioned media of KK/KF cocultures

compared to fibroblasts in monoculture (Fig. 32b).

• Upregulation of c-KIT in KKs as compared to Nks

Normal and keloid cells were cultured for 24 hrs followed by serum starvation for 48 hrs.

The cells were subsequently treated with growth medium for 24 hrs and subjected to flow

cytometry. KKs expressed higher levels of c-KIT as compared to NKs (Fig. 35ab).

• Increased levels of c-KIT in cocultured keratinocyte/fibroblast conditioned

media as compared to monocultures

NKs and KKs were cocultured with NFs and KFs respectively and the cell lysates from

both the cell types were collected and blotted for c-KIT. A downregulation of c-KIT

expression was observed in both cocultured keratinocyte (Fig 36a) and fibroblast cell

lysates (Fig 36b) as compared to monocultures. Phospho c-KIT (tyr 721) was however

upregulated in keloid cocultured cell lysates as compared to normal coculture lysates

Page 117: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

106

indicating activation of c-KIT in coculture conditions (Fig 36e). The conditioned media

obtained from the cocultured fibroblasts demonstrated elevated levels of c-KIT

suggesting a possible cleavage of c-KIT into the conditioned media upon coculture (Fig

36c). In addition higher cleaved c-KIT levels were observed in conditioned media

obtained from keloid cocultures as compared to normal cocultures suggesting an

enhanced shedding mechanism prevalent in keloid cocultures (Fig 36d).

• Elevated expression of TACE in cocultured KFs as compared to cocultured NFs

An increased expression of mature TACE was observed in total cell lysates obtained

from KFs coculutred with KKs as compared to NFs cocultured with NKs suggesting an

activation of this enzyme system in keloids (Fig. 38).

Page 118: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

107

Figure 30: Elevated levels of SCF in tissue extracts obtained from keloid tissues Expression of SCF in normal and keloid skin tissues were analyzed by western blot assay. Study groups comprised of normal skin and keloids. Tissue specimens were sonicated in lysis buffer and 100 µg of total protein extracts were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti SCF polyclonal antibody Lane 1-5: Keloid scar (K50, K51, K52, K53). Lane 6-8: Normal skin (N31, N32, N33). Equal loading was confirmed by blotting with β-actin

Page 119: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

108

Figure 31: Serum growth factors upregulated SCF expression in normal and keloid fibroblasts. Expression of SCF by fibroblast after exposure to 10% FCS was detected by Western blot assay. Fibroblasts were seeded in 6-well plates at a density of 1 × 104 cells/ml in DMEM/10% FCS for 24 hrs and subsequently in serum-free medium for another 48 hrs. The fibroblasts were subject to either DMEM/10% FCS or serum-free DMEM for 24 hrs. Fibroblasts were lysed and 100µg total protein cell lysates were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-SCF polyclonal antibody. a) Lane 1: NF4+DMEM, Lane 2: NF4+DMEM/10% FCS, Lane 3: NF14+DMEM, Lane 4: NF14+DMEM/10%FCS. b) Lane 1: KF45+DMEM, Lane 2: KF45+DMEM/10%FCS, Lane 3: KF49+DMEM, Lane 4: KF49+DMEM/10% FCS. Equal loading was confirmed by blotting with β-actin for cell extracts.

Page 120: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

109

Figure 32: Increased levels of SCF in conditioned media obtained from keloid cocultures as compared to monocultures a) Cell lysates obtained from KFs cocultured with KKs was assayed by western blot for SCF expression. Cell lysate from keloid fibroblasts in monoculture were used as controls. No difference in the expression of SCF was observed in cocultured fibroblast cell extracts compared to fibroblasts in monoculture. Lane 1: KF 49 non cc, Lane 2: KK49/KF49, Lane 3: KF 48 non cc, Lane 4: KK48/KF48. b) Conditioned media from KF cocultures were assayed by western blot for SCF expression. Conditioned media from KF in monoculture were used as controls. Equal volumes of conditioned media were subjected to SDS-PAGE. Conditioned media from KF cocultures demonstrated higher levels of SCF as compared to the KF monocultures. Lane 1: KF48 non cc, Lane 2: KK48/KF48, Lane 3: KF49 non cc, Lane 4: KK49/KF49

Page 121: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

110

Figure 33: Elevated levels of c-KIT and Phospho c-KIT (tyr 703, tyr 721) in tissue extracts obtained from keloid tissues. Expression of c-KIT and Phospho c-KIT at sites tyr 703 and tyr 721 in normal and keloid skin tissues were analyzed by western blot assay. Study groups comprised of normal skin and keloid scars. Tissue specimens were sonicated in lysis buffer and 100 µg of total protein extracts were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-c-KIT polyclonal, Phospho c-KIT tyr 703, Phospho c-KIT tyr 721 monoclonal antibodies. a) Lane 1-5: Normal skin (N31, N32, N33, N34, N35) Lane 6-13: Keloids (K50, K51, K52, K53, K54, K55, K56, K57). b) Lane 1-2: Normal skin (N31, N32) Lane 3-5: (K50, K51, K52)

Page 122: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

111

Figure 34: Increased localization of c-KIT in the basal layer of epidermis of keloid tissues. Paraffin sections of normal (a) and keloid (b) tissues were prepared and labeled with antibodies specific for c-KIT and observed with microscopy (40x). In each panel, the insert shows the same tissue labeled with a non-immune mouse antibody of the appropriate immunoglobulin isotype instead of the specific c-KIT antibody. Keloid tissues had more intense labeling in the basal layer of the epithelium compared with normal skin. The localization of c-KIT (or its lack in the negative control inserts) is shown by an arrow. The dermis and epidermis are represented by ‘D’ and ‘E’, respectively.

Page 123: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

112

Figure 35: c-KIT upregulated in KK as compared to NK. NK and KK were cultured, trypsinised and then probed with c-KIT monoclonal antibody followed by anti FITC conjugated secondary antibody. c-KIT was upregulated in keloid keratinocytes as compared to NK. a) NK 103, b) KK48

Page 124: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

113

Figure 36: Increased ectodomain shedding of c-KIT in KK/KF cocultures. Cell lysates from monocultured NK and KK were extracted and analyzed by western blot for the expression of c-KIT. Similarly, NK and KK which had been cocultured with NF and KF respectively were lysed and cell extracts were analyzed by western blot for the expression of c-KIT. Similarly NF and KF in monoculture and coculture were probed with anti-c-KIT and anti phospho c-KIT (tyr 721) antibody and subjected to western blot. Conditioned media from normal and keloid cocultures were collected and analyzed for the expression of c-KIT. Conditioned media from monocultures was used as control. a) Lane 1: NK1 Day 1, Lane 2: NK1/NF1 Day 1, Lane 3: NK1 Day 2, Lane 4: NK1/NF1 Day 2, Lane 5: NK1 Day 3, Lane 6: NK1/NF1 Day3, Lane 7: NK1 Day 4, Lane 8: NK1/NF1 Day 4, Lane 9: NK1 Day 5, Lane 10: NK1/NF1 Day 5, Lane 11: KK 48 Day 1, Lane 12: KK48/KF48 Day 1, Lane 13: KK48 Day 2, Lane 14: KK48/KF48 Day 2, Lane 15: KK48 Day 3, Lane 16: KK48/KF48 Day 3, Lane 17: KK48 Day 4, Lane 18: KK48/KF48 Day 4, Lane 19: KK48 Day 5, Lane 20: KK48/KF48 Day 5. b) Lane 1: NF1, Lane 2: NK1/NF1, Lane 3: NF2, Lane 4: NK2/NF2, Lane 5: KF48, Lane 6: KK48/KF48, Lane 7: KF49, Lane 8: KK49/KF49. Equal loading was confirmed by blotting with β-actin for cell extracts. c) Lane 1: KF 48, Lane 2: KK48/KF48, Lane 3: KF 49, Lane 4: KK49/KF49. d) Lane 1: NK1/NF1, Lane 2: NK2/NF2, Lane 3: NK3/NF3, Lane 4: NK8/NF8, Lane 5: KK48/KF48, Lane 6: KK49/KF49, Lane 7: KK45/KF45, Lane 8: KK 44/KF44. e) Lane 1: NK3/NF3, Lane 2: NK8/NF8, Lane 3: KK48/KF48, Lane 4: KK49/KF49

Page 125: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

114

Page 126: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

115

Figure 37: TACE overexpressed in keloid scar compared to normal skin Expressions of TACE in normal and keloid skin tissues were analyzed by western blot assay. Study groups comprised of normal skin and keloid scars. Tissue specimens were sonicated in lysis buffer and 100 µg of total protein extracts were subjected to SDS-PAGE and western blotting. Blots were hybridized with anti-TACE polyclonal antibody. Lane 1-6: Keloids (K50, K51, K52, K53, K54, K55, K56, K57), Lane 7-11: Normal skin (N31, N32, N33, N34)

Page 127: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

116

Figure 38: Upregulation of TACE in KF cocultures as compared to NF cocultures Expression of TACE in cocultured NF and KF were analyzed by western blot assay. Lane 1: NK1/NF1, Lane 2: NK2/NF2, Lane 3: NK3/NF3, Lane 4: NK8/NF8, Lane 5: KK48/KF48, Lane 6: KK49/KF49, Lane 7: KK45/KF45, Lane 8: KK 44/KF44.

Page 128: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

117

6.3 DISCUSSION The keloid is a dermal form of FPD characterized by increased proliferation of

fibroblasts, as well as, increased production of collagen in vitro (Cohen et al., 1990;

Ladin et al., 1995). Apoptosis-related genes and proteins are believed to be

downregulated or mutated in keloid tissues and fibroblasts (Sayah et al., 1999; Chodon et

al., 2000). In this study, the role of SCF and its receptor c-KIT was investigated in keloid

pathogenesis both in normal skin and keloid tissue sections, as well as in keratinocyte and

fibroblast cultures. A tyrosine kinsase inhibitor, gleevec was explored as a possible

therapeutic agent for keloids.

In order to determine the overall expressions of SCF in keloid and normal skin tissues, an

immunoblot analysis was performed on the tissue extract obtained from keloid and

normal skin. It was observed that SCF (50kda and 25kda forms) were highly expressed in

keloids compared to normal tissues, supporting the hypothesis that SCF might play a

crucial role in keloid pathogenesis. In addition, both mature (145kDa) and soluble

(95kDa) forms of KIT were found to be upregulated in keloid tissues suggesting a

possible aberration in receptor-ligand interaction. The tyr703 and tyr721 phosphorylation

sites have been previously demonstrated to be responsible for the activation of Ras/Erk

and PI3-Kinase pathways, respectively. A number of studies have implicated the critical

importance of the Ras/Erk pathway in cell division and survival (Lewis et al., 1998).

Adaptor proteins like Grb2 can directly associate with phosphorylated tyr703 in KIT

which may lead to activation of subsequent downstream pathways (Thommes et al.,

Page 129: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

118

1999). Activation of PI3-Kinase via KIT has been linked to mitogenesis, differentiation,

survival, adhesion, secretion and actin cytoskeleton reorganization (Yee et al., 1993;

Timokhina et al., 1998; Serve et al., 1995; Kubota et al., 1998; Vosseller et al., 1997). It

was found that tyr721 when phosphorylated, interacts directly with PI3-Kinase (Serve et

al., 1995). In this study, both tyr 703 and tyr 721 phosphorylation sites of c-KIT were

found to be upregulated in keloid scar tissues compared to normal skin tissues, although

the difference was significant only in case of tyr 721.

In order to study the localization of the KIT protein in the skin, immunohistochemistry

was performed to investigate the relative expressions of KIT protein in both keloid and

normal skin. An increased staining at the basal layer of keloid epidermis was observed

compared to normal skin. Thus, as the basal layer consists of mitotically active

keratinocytes, KIT could be playing a major role in modulating the dynamics of these

active and undifferentiated keratinocytes. These results were confirmed by flow

cytometry which demonstrated that KKs expressed increased levels of c-KIT as

compared to NKs.

When fibroblasts are exposed to serum, they interpret it to be a physiological wounding

signal (Iyer et al., 1999). Hence, exposure to serum reflects how fibroblasts would react

in vivo in the early phase of wounding. The increase in SCF observed upon exposure to

serum in both NFs and KFs suggested that SCF might have a role in early wound healing

process. This is consistent with previous studies, which found that SCF peaked on the

first day of wounding. The increase in SCF production increased the activation of mast

cells and triggered their degranulation, leading to the release of factors such as IL-1α and

Page 130: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

119

TNF-α that lead to inflammation, which is an early response in wound healing (Huttunen

et al., 2002).

Previous findings have shown that epithelial-mesenchymal interactions play an important

role in keloid pathogenesis with KK modulating the growth and proliferation of NF and

KF (Lim et al., 2001), influencing collagen expression by both NF and KF, inducing NF

to secrete collagen in a keloid-like manner (Lim et al., 2002), and utilizing the IGF

system of mitogens as inductive ligands for this process. Thus to investigate if these

interactions in anyway affected expression of SCF and its receptor c-KIT, NF and KF

were cocultured with NK and KK and blotted. No significant differences in SCF

expression between the fibroblast cell lysates derived from coculture and monoculture

was observed. However an increase in SCF levels was detected in the conditioned media

derived from the cocultures. ADAM19 and ADAM33 in COS and HEK293 cells

(Chesneau et al., 2003; Zou et al., 2004), MMP- 9 in hematopoetic stem cells (Heissig et

al., 2002), IL1-β produced by keratinocytes (Da silva et al., 2002) have all been shown to

promote cleavage and secretion of SCF. The soluble SCF can then act as a proliferative

agent (Ren et al., 2003). The mechanism by which epithelial-mesenchymal interactions

brought about an increase in secreted levels of SCF is not know and further studies are

needed to be unravel to delineate the mechanism. The role of the interactions between the

mesenchyme and the epithelium on the expression of c-KIT was further investigated.

Although the phosphorylated levels of c-KIT (tyr 721) were elevated in keloid cocultures

as compared to normal cocultures, both the keratinocytes and fibroblasts which had been

cocultured expressed decreased amounts of c-KIT compared to that from monoculture.

However the conditioned media obtained from these cocultures demonstrated elevated

Page 131: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

120

levels of the receptor suggesting a shedding of c-KIT. Keloid cocultures demonstrated

increased levels of soluble c-KIT as compared to normal cocultures. To investigate the

mechanism by which c-KIT was cleaved, a sheddase TACE, which was previously

demonstrated to be critical for shedding of c-KIT (Cruz et al., 2004) was explored. Not

only an increase of mature active TACE in keloid tissue extracts compared to normal

tissue extracts was observed but also found it to be upregulated in cocultured KF cell

lysates compared to cocultured NF confirming the prevalence of a TACE mediated

shedding mechanism. Although the presence of an ectodomain shedding exists in keloid

cocultures, the role of soluble c-KIT in keloid pathogenesis is still unclear. It has been

previously reported that binding of SCF results in rapid kinase activity-dependent

receptor endocytosis, ubiquitination and degradation (Yee et al., 1993; 1994; Miyazawa

et al., 1994). Activation of the receptor also results in cleavage and release of the soluble

extracellular domain (Yee et al., 1993; 1994; Miyazawa et al., 1994; Brizzi et al., 1994)

which then competes for the ligand with the membrane bound form (Broudy et al., 1997).

These mechanisms are activated as a negative feedback response to regulate ligand

receptor induced signal transduction. A similar mechanism may exist in keloid cocultures

where increased activation of c-KIT, as seen by increased level of phospho c-KIT, results

in ectodomain shedding by proteolytic cleavage. There have been numerous studies

demonstrating presence of soluble c-KIT, as a negative feedback response, in serum in

various disorders (Tajima et al., 1998; Kawakita et al., 1995). Alternative to the negative

feedback hypothesis, the soluble c-KIT bound to the free SCF ligands in the ECM, may

also help to increase the retention of the ligand in the extracellular environment, resulting

in a prolongation of the half-life due to the increased size and stability of the complex

Page 132: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

121

(Broudy et al., 1997; Peters et al., 1996). By prolonging the overall half-life of the SCF

ligand, the soluble KIT receptors may then serve to stabilize free SCF levels between

peaks and valleys of SCF secretion, leading to the sustained activation of the SCF-KIT

pathways (Heaney et al., 1995). Thus to confirm the exact role of ectodomain shedding

observed, more mechanistic studies are warranted to ascertain the behavior and effect of

the soluble KIT receptor-SCF complexes in vivo.

In summary, it has been shown for the first time that SCF and its receptor c-KIT were

both upregulated and activated in keloid tissues. In addition, epithelial–mesenchymal

interactions seemed to modulate both the dynamics of SCF and c-KIT resulting in their

shedding into the conditioned media, a process mediated probably by TACE (Fig 39a,

39b). Given these results the SCF/c-KIT system might be a target for therapeutic

intervention for keloid scars.

Page 133: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

122

Figure 39: c-KIT system in keloid pathogenesis a) SCF loop: Under the influence of the overlying keratinocytes, fibroblasts produce soluble SCF which not only has an autocrine effect but also a paracrine effect on the keratinocytes thus forming a short positive loop system between the two.. b) c-KIT shedding: Epithelial-meenchymal interactions result in mature KIT (145kDa) to be cleaved by proteolytic enzymes (TACE) thus releasing soluble KIT (95kDa) into the ECM. The soluble c-KIT then binds to SCF. The soluble KIT can be either a result of a negative feedback mechanism to regulate the overactivated scf/c-KIT system in keloidic cells or can be a mechanism in itself to activate the KIT system by increasing the half life and stability of SCF by binding to it and resulting in its sustained effect.

SCF C-KIT TACE

a)

KELOID KERATINOCYTES

KELOID FIBROBLAST

Page 134: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

123

b)

Keloid Fibroblast

c-Kit ectodomain

Shed c-Kit either binds to SCF

Shed kit as bio marker

Keloid Keratinocytes

Page 135: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

124

7 INVESTIGATION OF GLEEVEC AS A THERAPEUTIC AGENT FOR KELOID SCARS

Page 136: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

125

7.1 INTRODUCTION In the past, a wide variety of treatments has been used for keloids, including intralesional

corticosteroids, surgery, cryosurgery, silicone gel sheeting, radiotherapy, pressure

therapy, and laser therapy like flashlamp pulsed-dye laser treatment. Newer therapies like

interferon α2b, bleomycin injections and intralesional 5-fluorouracil (5FU) have also

been explored. Unfortunately, various side-effects like severe pain, ulceration, necrosis

and hyperpigmentation limit the utility of all these treatments (Nanda et al., 2004, Mustoe

et al., 2002). Neither of the mentioned therapies proved to be effective for patients,

underscoring the need to discover alternatives.

Having identified SCF and its tyrosine kinase receptor c-KIT to be upregulated and

activated in keloid pathogenesis, it was explored if a tyrosine kinase inhibitor could be

used as a possible therapeutic agent for keloid scars. In recent years, biopharmaceutical

research has actively targeted protein kinases for the treatment of several neoplastic

diseases. Two FDA approved examples are Gleevec™ (Imitanib Mesynate – Novartis

AG) for the treatment of chronic myeloid leukemia and gastrointestinal stromal tumors

and Iressa™ (Gefitinib - AstraZeneca) for the treatment of metastatic non-small cell lung

cancer by inhibiting intracellular tyrosine kinase phosphrylation.

Gleevec essentially inhibits four kinases: Bcr-abl, c-abl, c-KIT and PDGF (Capdeville et

al., 2002). As imatinib is an inhibitor of the receptor tyrosine kinases for PDGF and SCF,

it is able to block PDGF and SCF-mediated cellular events (Lefevre et al., 2004).

Gleevec in particular has been previously demonstrated to be effective for the treatment

of chronic myeloid leukemia (CML) by blocking cell proliferation and inducing

Page 137: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

126

apoptosis in Bcr-abl- expressing CML. It has been effective against gastrointestinal

stroma tumor (GIST) owing to its inhibitory action on KIT (Sattler et al., 2004). It has

also been demonstrated to have antifibrotic effects in organ fibrosis like lung fibrosis

(Aono et al., 2005) and renal fibrosis (Wang et al., 2005).

In the present study, the effects of various concentrations of gleevec on the proliferation

of KK and KF was explored by MTT assay. Studies were performed to further investigate

if gleevec was able to inhibit the c-KIT pathway by blotting with antibodies against

phospho c-KIT and the ensuing molecules in the PI-3 kinase pathway. The expression of

the ECM components like collagen, fibronectin and α-smooth muscle actin and the

expressions of profibrotic and pro-angiogenic growth factors like VEGF, TGF-β, HDGF,

FGF-2, SCF on treatment with gleevec were also monitored. To investigate if gleevec

could reduce scar contracture, an in-vitro model which consisted of a FPCL was used and

its contraction on treatment with gleevec studied. Finally the effect of gleevec on the

bioenergetics of the cell was investigated by studying the intracellular ATP and the rate

of intracellular ATP synthesis in KF on treatment with gleevec.

Page 138: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

127

7.2 RESULTS • No significant difference in the proliferative potential of KK and KF on

treatment with gleevec

Gleevec was added in varying concentrations (0.5, 1, 2.5, 5, 10, and 20 µg/ml) in

presence of 5% v/v serum to KK and KF and MTT assay was performed. No significant

difference was observed in the proliferative potential of both keratinocytes and fibroblast

on treatment with gleevec (Fig. 40).

• Gleevec inhibited the SCF/c-KIT pathway by dowregulating the

phosphorylation of c-KIT at tyr 721, Akt and mTOR

KFs were treated with 5 µg/ml of gleevec in DMEM containing 5% v/v serum for 15

min, 1 hr and 24 hrs.Total extracts from cells were probed with antibodies against

phosphorylated forms of c-KIT (Tyr 721), Akt and mTOR. A decrease in the expression

of phospho c-KIT, phospho Akt and phospho mTOR was observed within 15 minutes of

treatment with gleevec (Fig. 41).

• Gleevec downregulated the expressions of ECM and profibrotic factors in KFs

in a dose-dependent manner

Gleevec in presence of 5% v/v serum was added in varying concentrations (2.5, 5, and

10µg/ml) to KFs and the total cell extracts subjected to western blot. Significant decrease

in levels of collagen, fibronectin and α-SMA were observed on treatment with gleevec in

a dose-dependent manner (Fig. 42a). A significant decrease in expression levels of

Page 139: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

128

profibrotic factors like VEGF, HDGF, TGF-β1, SCF, and FGF-2 were also observed

(Fig. 42b).

• Gleevec decreased the contraction of collagen lattice populated by KF in a dose -

dependent manner.

FPCL were prepared as previously described and treated with various conentrations of

gleevec in a medium containing 5% v/v serum for 48 hrs. The medium was subsequently

removed and the lattices stained by rhodamine solution to give it a pink colour. The

lattices were then photographed and the area calculated as previously mentioned for side-

by-side analysis. It was observed that collagen lattices populated with Keloid-derived

fibroblasts demonstrated significantly lesser contraction when treated with gleevec as

compared to collagen lattices treated with medium containing 5% v/v serum which was

used a control (Fig 43). Lattices in serum-free media were used a negative controls.

• Gleevec reduced the rate of ATP synthesis and intracellular ATP levels in KFs.

KFs were treated with gleevec (5 µg/ml) for 24 hrs and subjected to luciferin-luciferase

assay. The rate of ATP synthesis in KF was decreased on treatment with gleevec at a

concentration of 5 µg/ml. In addition the intracellular ATP levels in KFs were also

significantly reduced at similar concentrations of gleevec (Fig 44).

Page 140: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

129

Figure 40: No significant difference in the proliferative potential of KK and KF. KKs and KFs were treated with different concentrations of gleevec in 5% serum (0.5, 1, 5, 10, 20 µg/ml). Cells were subjected to MTT assay after 24hrs of treatment. Untreated fibroblasts in 5% serum were used as control. Fibroblasts in DMEM only were used as a negative control. No significant difference in proliferation was observed.a) KK48. b) KF48.

Page 141: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

130

Figure 41: Gleevec downregulated expression of Phospho Akt, Phospho mTOR, Phospho c-KIT (tyr 721). KFs were treated with gleevec (5 µg/ml) and the cell lysates were collected after 15 min, 1 hr, and 24 hrs of treatment. The lysates were blotted with antibodies against Phospho Akt, Phospho mTOR, Phospho c-KIT (tyr 721). Gleevec downregulated the expression of phosphor Akt, Phospho mTOR and Phospho c-KIT (tyr 721) after 15 min. KFs treated with 5% serum were used as control. Lane 1: KF48 +DMEM with 5% serum after 15 min, Lane 2: KF48 + gleevec (5 µg/ml) in 5% serum after 15 min, Lane 3: KF48 +DMEM with 5% serum after 1 hr, Lane 4: KF48 + gleevec (5 µg/ml) in 5% serum after 1 hr, Lane 5: KF48 +DMEM with 5% serum after 24 hrs, Lane 6: KF48 + gleevec (5 µg/ml) in 5% serum after 24 hrs. Equal loading was confirmed by blotting with β-actin.

Page 142: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

131

Figure 42: Gleevec downregulated expression of collagen, fibronectin, α-SMA, VEGF, HDGF, TGF-β1, SCF, FGF-2. Keloid fibroblasts were treated with different concentrations of gleevec in 5% serum (2.5µg/ml, 5µg/ml). After treatment for 24hrs the cells were lysed and blotted with antibodies against extracellular matrix components like a) collagen, fibronectin, α-SMA and b) profibrotic factors like TGF-β1, SCF, VEGF, HDGF, FGF-2. Untreated fibroblasts in 5% serum were used as controls. Lane 1: KF48 +DMEM with 5% serum, Lane 2: KF48+ 2.5µg/ml gleevec in 5% serum, Lane 3: KF48+ 5µg/ml gleevec in 5% serum. Equal loading was confirmed by blotting with β-actin.

Page 143: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

132

Figure 43: Gleevec significantly reduced the contraction of collagen gel lattice incorporated with keloid fibroblasts. The fibroblast populated collagen lattices were treated with various concentrations of gleevec (0.5µg/ml, 2.5µg/ml, 5µg/ml, 10µg/ml) for 48hr. The lattices were then collected and stained with Rhodamin B and photographed. a) KF 48+DMEM, b) KF48 +DMEM with 5% serum, c) KF48+ 0.5µg/ml gleevec in 5% serum, d) KF48+ 2.5µg/ml gleevec in 5% serum, e) KF48+ 5µg/ml gleevec in 5% serum, f) KF48+ 10µg/ml gleevec in 5% serum.

Page 144: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

133

Figure 44: Gleevec significantly reduced the intracellular ATP and the rate of ATP synthesis in KF. KFs were treated with gleevec (5 µg/ml) for 24 hrs and analysed for the levels of intracellular ATP. The rate of ATP synthesis was also calculated in the treated samples with untreated samples as control. Open bar represents untreated KFs and solid bars represent treated KFs.

Page 145: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

134

7.3 DISCUSSION In this study, no significant difference in the proliferative potential of KK and KF on

treatment with various concentrations of gleevec was observed. This was in accordance

with previous studies by Distler and his co-workers (2006) in which they reported that

gleevec showed no effect on the proliferative potential of dermal fibroblasts. However

gleevec was able to reduce the expression of ECM components, like collagen,

fibronectin, α-SMA significantly in KF in a dose-dependent manner highlighting its

importance as a potential antifibrotic agent. Gleevec also downregulated TGF-β and

VEGF which have been previously shown to be upregulated in KF (Kim et al., 2000; Xia

et al., 2004; Ong et al., 2007). It was dowregulated expression of HDGF which was

previously reported to be involved in organ development and modeling (Mori et al.,

2004) and recently demonstrated to be upregulated in keloid tissues (unpublished). FGF-2

and SCF, growth factors implicated in various fibrotic disorders (Yu et al., 2003;

Garbuzenko et al., 2002), were also downregulated upon treatment with gleevec in a

concentration-dependent manner. the effects of gleevec on FPCL, an established 3-d

wound contraction and scar contracture model (Bell et al., 1979) was further tested.

Gleevec was significantly reduced the contraction of the lattices thus highlighting its role

in modulating scar contracture.

Having demonstrated an antifibrotic effect of gleevec, it was explored if gleevec owed its

inhibitory activity to c-KIT receptor inactivation. Activation of PI-3 kinase via KIT has

been linked to mitogenesis, differentiation, survival, adhesion, secretion and actin

cytoskeleton reorganization (Yee et al., 1993; Timokhina et al., 1998; Serve et al., 1995;

Page 146: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

135

Kubota et al., 1998; Vosseller et al., 1997). It was found that tyr721 when

phosphorylated, interacts directly with PI-3 kinase (Serve et al., 1995). Within 15 min of

treatment with gleevec, the expression of phospho c-KIT (tyr 721) in KF was reduced. In

addition, downstream signaling molecules like phospho Akt and phospho mTOR were

also downregulated in these KF suggesting an inhibition of the P13 kinase pathway in

these fibroblasts on treatment with gleevec.

Ueda’s team (1999) previously demonstrated a high level of ATP for a longer time in KF.

Thus we investigated if gleevec could reduce the intracellular ATP levels as well as the

rate of ATP production in KF. It was observed that gleevec could significantly decrease

both the rate of ATP production, as well as total intracellular ATP levels in KFs.

However the mechanism by which it has such an effect needs to be explored.

To summarize, this study demonstrated that a selective tyrosine kinase inhibitor, gleevec,

was able to downregulate the expression of ECM components, as well as profibrotic

factors in KFs via inhibition of the c-KIT mediated PI3 kinase signaling pathway. It was

also able to make the KFs less bioenergetic by reducing the ATP levels. Thus given its

effects, it could well be used as a potential therapeutic agent for keloids.

Page 147: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

136

GENERAL CONCLUSION

The achievement of this project is the identification and elucidation of novel molecular

pathways in the pathogenesis of keloid scars. Using a large database of clinical tissue

samples from keloid and normal patients and various in vitro models of wound repair, it

was demonstrated for the first time that activin-A, stem cell factor (SCF) and its tyrosine

kinase receptor c-KIT, fibroblast growth factor-2 (FGF-2) and proteoglycans like

syndecan-2 and decorin play important roles in keloid pathogenesis and could be possible

targets for therapeutic intervention. Development of specific antibodies that antagonize

these growth factors or block its receptor could result in reduced collagen accumulation,

predicated on the results of this study. Similarly, based on these results, there is a scope

for development of peptide antagonists or receptor decoy molecules based on molecular

modeling of the receptor–ligand binding characteristics. Another approach would be to

screen for small molecules, potentially identifying compounds that antagonize the

receptors and its downstream signaling.

In addition to identifying these new targets, this study also underscores the importance of

epithelial-mesenchymal interactions in keloid pathogenesis. The interaction between the

dermis and the overlying epidermis was demonstrated to be critical in scar contracture

and in modulating the biology of activin-A, SCF/c-KIT, FGF-2, syndecan-2 and decorin

in keloid scars. This observation may have immense clinical relevance. Currently

available therapy involves the intralesional corticosteroid injection which is very painful

for the patients. Having demonstrated the importance of the epithelium, topical

application of chemical agents and antagonists of profibrotic growth factors could be

Page 148: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

137

used as novel delivery systems thus making the therapy more patient compliant and

friendly.

Another interesting data which sprouted from this study was the shedding of the c-KIT

receptor in keloidic conditions which had been previously shown to occur in malignant

disorders. Keloids, although ‘benign’, have always been acknowledged to possess growth

autonomy to a certain degree resembling malignant tissues, resulting in the invasion and

involvement of adjacent skin in the course of its growth, and thus the clinical description

of the keloid as a scar that exceeds the boundaries for the original wound or incision. The

finding of c-KIT receptor shedding might well reflect growth autonomy in keloid

fibroblasts resembling transformed cells.

This study went on to explore new pharmacological interventions. This resulted in the

identification of growth factors, like follistatin and decorin, and chemical agents like

gleevec, as potential therapeutic agents for keloid scars. Follistatin is a natural antagonist

of activin-A and having identified activin-A as a player in keloids, it could be used as a

potential therapy. In this study follistatin was shown to downregulate the expression of

ECM components like collagen, fibronectin and α-SMA. Another growth factor which

was shown to be effective in downregulating ECM significantly was decorin. The

observation that decorin is downregulated in keloid scars and exuberantly expressed in

normal skin underscores its importance in normal wound repair. Finally having identified

a tyrosine kinase receptor namely c-KIT to be important in keloid pathogenesis, a well

known FDA-approved tyrosine kinase inhibitor, gleevec, was explored. Using in vitro

models for anti-scar research experimentation, it was found that gleevec could reduce the

Page 149: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

138

expression of ECM components, as well as key profibrotic and pro-angiogenic growth

factors like VEGF, TGF-β, and HDGF. In addition it was also able to downregulate SCF

and FGF-2, which were also identified in this study, as key regulators of keloidogenesis.

Gleevec reduced contractile s of KF and also rendered the fibroblast less bioenergetic. At

the molecular levels, gleevec was shown to effectively block the PI-3 kinase mediated

SCF/c-KIT pathway. Figure 45 summarizes the growth factors involved in keloid

pathogenesis and the various possible targets for therapeutic intervention elucidated by

our research.

Page 150: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

139

Figure 45: Summary of growth factors involved in keloid pathogenesis and possible therapeutic targets.

Page 151: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

I

REFERENCES

• Ala-Kokko L, Rintala A, Savolainen ER. Collagen gene expression in keloids:

analysis of collagen metabolism and type I, III, IV, and V procollagen mRNAs in keloid tissue and keloid fibroblast cultures. J Invest Dermatol 1987, 89:238-244.

• Al-Attar A, Mess S, Thomassen JM, Kauffman CL, Davison SP. Keloid pathogenesis and treatment. Plast Reconstr Surg 2006, 117:286-300.

• Aono Y, Nishioka Y, Inayama M, Ugai M, Kishi J, Uehara H, Izumi K, Sone S. Imatinib as a novel antifibrotic agent in bleomycin-induced pulmonary fibrosis in mice. Am J Respir Crit Care Med 2005, 171:1279-1285.

• Ashman LK. The biology of stem cell factor and its receptor c-KIT. Int J Biochem Cell Biol 1999, 31:1037-1051.

• Bayat A, Bock 0, Mrowietz U. Genetic susceptibility to Keloid disease and transforming growth factor beta 2 polymorphism. Br J Plast Surg 2002 , 55:283-86.

• Bayat A, Bock O, Morwietz U. William O, Ferguson M. Genetic Susceptibility to Keloid Disease and Hypertrophic Scarring: Transforming growth factor beta 1 common polymorphisms and Plasma level. Plast Reconstr Surg 2003, 111:535-43.

• Bayat A, Arscott G, Ollier WE, Ferguson MW, Mc Grouther DA. Description of site-specific morphology of keloid phenotypes in an afrocaribbean population.Br J Plast Surg 2004, 57:122-133.

• Bayat A, Walter J, Bock O, Mrowietz U, Ollier W, Ferguson M. Genetic susceptibility to Keloid disease: mutation screening of the TGFβ3 gene. Br J Plast Surg 2005, 58:914-21.

• Bayer-Garner IB, Sanderson RD, Dhodapkar MV, Owens RB, Wilson CS. Syndecan-1/CD138 immunoreactivity in bone marrow biopsies of multiple myeloma: Shed syndecan-1 accumulates in fibrotic regions. Mod Path 2001, 14:1052-1058.

• Bell E, Ivarsson B, Merril C. Production of a tissue-like structure by contraction of collagen lattices by human fibroblasts of different proliferative potentials in vitro. Proc Natl Acad Sci 1979, 76: 1274-1278.

• Berman B, Bieley HC. Adjunct therapies to surgical management of keloids. J Am Acad Dermatol 1995, 33:117-123.

• Berman B, Kaufman J. Pilot study of the effect of postoperative imiquimod 5% cream on the recurrence rate of excised keloids. J Am Acad Dermatol 2002, 47:S209-211

• Besner, GE, Higashiyama, S, Klagsbrun, M. Isolation and characterization of a macrophage-derived heparin-binding growth factor. Cell Regul 1990, 1: 811–819.

• Bitterman PB, Henke CA. Fibroproliferative disorders. Chest 1991, 99:81s-84s. • Bläuer M, Wichmann L, Punnonen R, Tuohimaa P. Measurement of activin B in

human salivary and localization of activin subunits in rat salivary glands. Biochem Biophys Res Commun 1996, 222:230-235.

• Bloom D. Heredity of keloids. N Y State Med J 1956, 56:511-519.

Page 152: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

II

• Blume-Jensen P, Claesson-Welsh L, Siegbahn A, Zsebo KM, Westermark B, Heldin CH. EMBO J 1991, 10:4121-4128.

• Bodo M, Lilli C, Bellucci C, Carinci P et al. Basic fibroblast growth factor autocrine loop controls human osteosarcoma phenotyping and differentiation. Mol Med 2002, 8: 393-404.

• Bodokh, I., Burn, P. Traitment des cheloides par infiltrations de Bleomycine. Ann Dermatol Venereol. 1996, 123:791.

• Border WA, Noble NA, Yamamoto T, Harper J. Natural inhibitor of TGF-β protects against scarring in experimental kidney disease. Nature 1992, 360:361-364.

• Border WA, Nobel NA. Transforming growth factor-beta in tissue fibrosis. N Engl J Med 1994, 331:1286–1292.

• Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976, 72:248-254.

• Brent B. The role of pressure therapy in management of ear lobe keloids: Preliminary report of a controlled study. Ann Plast Surg 1978, 1:579.

• Brizzi MF, Blechman JM, Cavalloni G, Givol D, Yarden Y, Pegoraro L.Protein kinase C-dependent release of a functional whole extracellular domain of the mast cell growth factor (MGF) receptor by MGF-depdendent human myeloid cells. Oncogene 1994, 9:1583-1589.

• Broudy VC, Kovach NL, Bennett LG, Lin N, Jacobsen FW, Kidd PG. Blood 1994, 83:2145-2152.

• Broudy VC. Stem cell factor and Hematopoiesis. Blood 1997, 90:1345-1364. • Burgess WH, Maciag T. The heparin-binding (fibroblast) growth factor family of

proteins. Ann Rev Biochem 1989, 58: 575-606. • Buzzard JJ, Farnworth PG, de Krester DM, O’Connor AE, Wreford NG, Morrison

JR. Proliferative phase Sertoli cells display a developmentally regulated response to activin in vitro. Endocrinology 2003, 144:474-483.

• Calderon M, Lawrence WT, Banes AJ. Increased proliferation in keloid fibroblasts wounded in vitro. J Surg Res 1996, 61:343-347.

• Cancilla B, Jarred RA, Wang H, Mellor SL, Cunha GR, Risbridger GP. Regulation of prostate branching morphogenesis by activin A and follistatin. Dev Biol 2001, 237:145-158.

• Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (ST1571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov 2002, 1:493-503.

• Carell A, Hartmann A. Cicatrisation of wounds. I. The relation between the size of a wound and the rate of its cicatrisation. J Exp Med 1916, 24:429-450.

• Carey DJ. Syndecans: multifunctional cell –surface co-receptors. Biochem J 1997, 327: 1-16.

• Chen L, Klass C, Woods A. Syndecan-2 regulates transforming growth factor-beta signaling. J Biol Chem 2004, 279(16):15715-15718.

• Chen YG, Lui HM, Lin SL, Lee JM, Ying SY. Regulation of cell proliferation, apoptosis, and carcinogenesis by activin. Adv Exp Med Biol 2002, 227:75-87.

Page 153: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

III

• Chesneau V, Becherer JD, Zheng Y, Erdjument-Bromage H, Tempst P, Blobel CP. Catalytic properties of ADAM19. J Biol Chem 2003, 278: 22331-22340.

• Chin BY, Petrache I, Choi AM, Choi ME. Transforming growth factor beta 1 rescues serum deprivation-induced apoptosis via the mitogen-activated protein kinase (MAPK) pathway in macrophages. J Biol Chem 1999, 274:11362-11368.

• Chodon T, Sugihara T, Igawa H, Funayama E, Furukawa H. Keloid-derived fibroblasts are refractory to Fas-mediated apoptosis and neutralization of autocrine transforming growth factor-β1 can abrogate this resistance. Am J Pathol 2000, 157:1661-1669.

• Clark RA. In: The Molecular and Cellular Biology of Wound Repair (2nd edition), edited by Clark RA. New York 1996: Plenum:3-50.

• Clasper S, Vekemans S, Fiore M, Plebanski M, Wordsworth P, David G, Jackson DG. Inducible expression of the cell surface heparan sultphate proteoglycan syndecan-2 (Fibroglycan) on human activated macrophages can regulate fibroblast growth factor action. J Biol Chem 1999, 274: 24113-24123.

• Cohen IK, Peacock EE. Keloids and Hypertrophic Scars in Plastic Surgery, edited by McCarthy JG, Philadelphia: Saunders 1990:732-747.

• Cosman B, Crikelair GF, Ju DMC, Gaulin JC, Lattes R. The surgical treatment of keloids. Plast Reconstr Surg 1961, 27:335-356.

• Costa AM, Peyrol S, Porto LC, Comparin JP, Foyatier JL, Desmouliere A. Mechanical forces induce scar remodelling. Study in non-pressure-treated versus pressure-treated hypertrophic scars. Am J Pathol 1999, 155:1671-1679.

• Crockett DJ. Regional keloid susceptibility. Br J Plast Surg 1964, 17:245-253. • Cruz AC, Frank BT, Edwards ST, Dazin PF, Peschon JJ, Fang KC. Tumor

necrosis factor-alpha converting enzyme controls surface expression of c-KIT and survival of embryonic stem cell-derived mast cells. J Biol Cell 2004, 279:5612-5620.

• Da Silva CA, Kassel O, Mathieu E, Massard G, Gasser B, Frossard N. Inhibition by glucocorticoids of the interleulin-1beta-enhanced expression of the mast cell growth factor SCF. Br J Pharmacol 2002, 135:1634-1640.

• David G, Bai XM, Van Der Schueren B, Marynen P, Cassimann JJ, Van den Berghe H. Spatial and temporal changes in expression of fibroglycan (syndecan-2) during mouse embryonic development. Development 1993, 119: 841-854.

• Davies EJ, Blackhall FH, Shanks JH, David G, McGown AT, Swindell R, Slade RJ, Martin-Hirsch P, Gallagher JT, Jayson GC. Distribution and Clinical Significance of Heparan Sulfate Proteoglycans in Ovarian Cancer. Clin Cancer Res 2004, 10: 5178-5186.

• De Luca A, Santra M, Baldi A, Giordano A, Iozzo RV. Decorin induced growth suppression is associated with up-regulation of p21, an inhibitor of cyclin-dependent kinases. J Biol Chem 1996, 271:18961-18965.

• Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-β family signaling. Nature 2003, 425:577-584.

• Di Cesare PE, Cheung DT, Perelman N, Libaw E, Peng L, Nimni ME. Alteration of collagen composition and cross-linking in keloid tissues. Matrix 1990, 10:172-178.

Page 154: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

IV

• Diegelmann RF, Bryant CP, Cohen IK. Tissue alpha globulins in keloid formation. Plast Reconstr Surg 1977, 59:418.

• Dowling CR, Risbridger GP. The role of inhibins and activins in prostate cancer pathogenesis. Endocr Relat Cancer 2000, 7(4):243-256.

• Elenius V, Gotte M, Reizes O, Elenius K, Bernfield M. Inhibition by the soluble syndecan-1 ectodomains delays wound repair in mice overexpressing syndecan-1. J Biol Chem 2004, 279: 41928–41935.

• El Kossi MM, El Nahas AM. Stem cell factor and crescentic glomerulonephritis. Am J Kidney Dis 2003, 41:785-795.

• Engrav LH, Covey MH, Dutcher KD, Heimbach DM, Walkinshaw MD, Arvin JA.

Impairment, time out of school, and time off from work after burns. Plast Reconstr Surg 1987, 79:927-932.

• Essner JJ, Chen E, Ekker SC. Syndecan-2. Int J Biochem Cell Biol 2006, 38(2): 152-156.

• Fears CY, Gladson CL, Woods A. Syndecan-2 are expressed in the microvasculature of gliomas and regulate angiogenic processes in microvascular endothelial cells. J Biol Chem 2006, 281: 14533-14536.

• Finesmith TH, Broadley KN, Davidson JM. Fibroblasts from wounds of different stages of repair vary in their ability to contract a collagen gel in response to growth factors. J Cell Physiol 1990; 144: 99–107.

• Fireman E, Kivity S, Shahar I, Reshef T, Mekori YA. Secretion of stem cell factor by alveolar fibroblasts in interstitial lung diseases. Immunol Lett 1999, 67:229-236.

• Fischer JW Kinsella MG, Levkau B, Clowes AW, Wight TN. Retroviral overexpression of decorin differentially affects the response of arterial smooth muscles cells to growth factors. Arterioscler Thromb Vasc Biol 2001, 21:777-784.

• Fitzgerald ML, Park PW, Wang Z, Park PW, Murphy G, Bernfield M. Shedding of syndecan-1 and -4 ectodomains is regulated by multiple signaling pathways and mediated by a TIMP-3 sensitive cell surface metalloproteinase. J Cell Biol 2000, 148:811-824.

• Fitzpatrick RE. Treatment of inflamed hypertrophic scars using intralesional 5-FU. Dermatol Surg 1999, 25:224:232.

• Fujiwara M, Muragaki Y, Ooshima A. Keloid-derived fibroblasts show increased secretion of factors involved in collagen turnover and depend on matrix metalloproteinase for migration. Br J Dermatol 2005, 153: 295.

• Fukushima K, Badlani N, Usas A, Riano F, Fu F, Huard J. The use of an antifibrosis agent to improve muscle recovery after laceration. Am J Sports Med 2001, 29: 394-402.

• Funayama E, Chodon T, Oyama A, Sugihara T. Keratinocytes promote proliferation and inhibit apoptosis of the underlying fibroblasts: An important role in the pathogenesis of keloid. J Invest Dermatol 2003, 121:1326-1331.

• Furukawa M, Eto Y, Kojima I. Expression of immunoreactive activin A in fetal rat pancreas. Endocr J 1995, 42:63-68.

Page 155: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

V

• Fusenig NE. Epithelial-mesenchymal interactions regulate keratinocyte growth and differentiation in vitro. In The Keratinocyte Handbook, Leigh I, Lane B, Watt F (eds). Cambridge University Press: Cambridge, UK, 1994

• Gabrilove JL, White K, Rahman Z, Wilson EL. Stem cell factor and basic fibroblast growth factor are synergistic in augmenting committed myeloid progenitor cell growth. Blood 1994, 83:907-910.

• Gaedeke J, Boehler T, Budde K, Neumayer HH, Peters H. Glomerular activin A overexpression is linked to fibrosis in anti-Thy1 glomerulonephritis. Nephrol Dial Transplant 2005, 20:319-328.

• Gallo RL, Bernfield M, Kainulainen V. Syndecan biology in wound repair, In: Scarless Wound Healing edited by Garg HG, Longaker MT. US: Marcell Dekker Inc., 2000:37-61.

• Garbuzenko E, Nagler A, Pickholtz D, Gillery P, Reich R, Maquart FX, Levi-Schaffer F. Human mast cells stimulate fibroblast proliferation, collagen synthesis and lattice contraction: a direct role for mast cells in skin fibrosis. Clin Exp Allergy 2002, 32:237.

• Genestier L, Kasibhatla S, Brunner T, Green DR. Transforming growth factor-ß inhibits Fas ligand expression and subsequent activation-induced cell death in T cells via downregulation of c-Myc. J Exp Med 1999, 189:231-239.

• Giri S, Hyde D, Braun R, Gaaede W. Antifibrotic effect of decorin in a bleomycin hamster model of lung fibrosis. Biochem Pharmacol 1997, 54:1205-1217.

• Gotte M. Syndecans in inflammation. FASEB J 2003, 17: 575-91. • Granstein RD, Rook A, Flotte TJ, Haas A, Gallo RL, Jaffe HS, Amento EP. A

controlled trial of intralesional recombinant interferon-gamma in the treatment of keloid scarring. Clinical and histological findings. Arch Dermatol 1990, 126:1295-1302.

• Grinell F. Fibroblasts. Myofibroblasts and wound contraction. Mini review on the cellular mechanism of diseases. J Cell Biol 1994, 124:401-404.

• Gulyas M, Hjerpe A. Proteoglycans and WT1 as markers for distinguishing adenocarcinoma, epithelioid mesothelioma, and benign mesothelium. J Pathol 2004, 199: 479–487.

• Haisa M, Okochi H, Grtendorst GR. Elevated levels of PDGF alpha receptors in keloid fibroblasts contribute to an enhanced response to PDGF. J Invest Dermatol 1994, 103:560-563.

• Han I, Park H and Oh ES. New Insights into Syndecan-2 Expression and Tumorigenic Activity in Colon Carcinoma cells. J Mol Histol 2004, 35: 319-326.

• Hansen MB, Nielsen SE, Berg K. Re-examination and further development of a precise and rapid dye method for measuring cell growth/cell kill. J Immunol Meth 1989, 119:203-210.

• Harris AK, Stopak D, Wild P. Fibroblast traction as a mechanism for collagen morphogenesis. Nature 1981, 290:249-251.

• Heaney ML, Golde DW. Soluble cytokine receptors. Blood 1996, 87:847-857. • Heino J, Kähäri VM, Mauviel A, Krusius T. Human recombinant interleukin 1

regulates cellular mRNA levels of dermatan sulphate proteoglycan core protein. Biochem J 1988, 252: 309-312.

Page 156: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

VI

• Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell 2002, 109: 625-637.

• Hibi K, Takahashi T, Sekido Y, Ueda R, Hida T, Ariyoshi Y, Takagi H, Takahashi T. Coexpression of the stem cell factor and c-KIT genes in small-cell lung cancer. Oncogene 1991, 6:2291-2296.

• Hildén K, Tuuri T, Eramaa M, Ritvos O. Expression of type II activin receptor genes during differentiation of human K562 cells and cDNA cloning of the human type IIB activin receptor. Blood 1994, 83:2163-2170.

• Hilderbrand A, Romanis M, Rasmussen L, Heinegard D, Twardzik DR, Border WA, Rouslahti E. Interaction of the small interstitial proteoglycans biglycan, decorin and fibromodulin with transforming growth factor beta. Biochem J 1994, 302:527-534.

• Huang E, Nocka K, Beier DR, Chu TY, Buck J, Lahm HW, Wellner D, Leder P, Besmer P. The hematopoietic growth factor KL is encoded by the SI locus and is the ligand of the c-KIT receptor, the gene product of the W locus. Cell 1990, 63:225-233.

• Hübner G, Hu G, Smola H, Werner S. Strong induction of activin expression after injury suggests an important role of activin in wound repair. Dev Biol 1996, 173: 490-498.

• Hunt TK, Dunphy JE. Fundamentals of Wound Management. Appleton-Century-Crofts, Newyork 1979.

• Hurle JM, Ganan Y. Interdigital tissue chondrogenesis induced surgical removal of the ectoderm in the embryonic chick leg bud. J Embryol Exp Morph 1986, 94:231-244.

• Huttunen M, Naukkarinen A, Horsmanheimo N, Harvima IT. Transient production of stem cell factor in dermal cells but increasing expression of Kit receptor in mast cells during normal wound healing. Arch Dermatol Res 2002, 294:324-330.

• Imai K, Hiramatsu A, Fukushima D, Pierschbacher M D, Okada Y. Degradation of decorin by matrix metalloproteinases: identification of the cleavage sites, kinetic analyses and transforming growth factor-β1 release. Biochem J 1997, 322: 809-814.

• Iozzo RV, Chakrani F, Perrotti D, McQuillan DJ, Skorski T, Calabretta B, Eichstetter I. Cooperative action of germ line mutations in decorin and p53 accelarates lymphoma tumorigenesis. Proc Natl Acad Sci 1999, 96: 3092-3097.

• Jones KL, de Kretser DM, Patella S, Phillips DJ. Activin A and follistatin in systemic inflammation. Mol Cell Endocrinol 2004, 225:119-125.

• Kähäri V M, Häkkinen L, Westermarck J, Larjava H. Differential regulation of decorin and biglycan gene expression by dexamethasone and retinoic acid in cultured human skin fibroblasts. J Invest Dermatol 1995, 104:503-508.

• Kähäri VM, Larjava H, Uitto J. Differential regulation of extracellular matrix proteoglycan (PG) gene expression. Transforming growth factor-β1 up-regulates biglycan (PG1), and versican (Large fibroblast PG) but down regulates decorin (PGII) mRNA levels in human fibroblasts in culture. J Biol Chem 1991, 266: 10608-10615.

Page 157: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

VII

• Kamamoto F, Paggiaro AO, Rodas A, Herson MR, Mathor MB, Ferreira MC. A wound contraction experimental model for studying keloids and wound healing modulators. Artif Organs 2003, 27:701-705.

• Kawakita M, Yonemura Y, Miyake H, Ohkubo T, Asou N, Hayakawa K, Nakamura M, Kitoh T, Osawa H, Takatsuki K. Br J Haematol 1995, 91:23-29.

• Kelly AP. Keloids. Dermatol Clin 1988, 6:413-424. • Khoo YT, Ong CT, Mukhopadhyay A, Han HG, Do DV, Lim IJ, Phan TT.

Upregulation of secretory connective tissue growth factor in keratinocyte-fibroblast coculture contributes to keloid pathogenesis. J Cell Physiol 2006, 208:336-343.

• Kikuchi K, Kadono T, Takehara K. Effects of various growth factors and histamine on cultured keloid fibroblasts. Dermatology 1995, 190:4-8.

• Kim CW, Goldberger OA, Gallo RL, Bernfield M. Members of the syndecan family of heparan sulphate proteoglycans are expressed in distinct cell-, tissue- and development –specific patterns. Mol Bio Cell 1994, 5: 797-805.

• Kim ES, Kim RS, Ren RF, Hawver DB, Flanders KC. Transforming growth factor- ß inhibits apoptosis induced by ß-amyloid peptide fragment 25–35 in cultured neuronal cells. Mol Brain Res 1998, 62:122-130

• Kim WJH, Levinson H, Gittes GK, Longaker MT. Molecular Mechanisms in Keloid Biology, In: Scarless Wound Healing edited by Garg HG, Longaker MT. US: Marcell Dekker Inc., 2000:161-171

• Kischer CW, Theis AC, Chvapil M. Pervascular myofibroblasts and microvascular occlusion in hypertrophic scars and keloids. Hum Pathol 1982, 13:819.

• Kischer CW. The microvessels in hypertrophic scars, keloids and related lesions: a review. J Submicrosc Cytol Pathol 1992, 24:281-296.

• Knight PG, Muttukrishna S, Groome NP. Development and application of a two-site enzyme immunoassay for the determination of ‘total’ Activin-A concentration in serum and follicular fluid. J Endocrinol 1996, 148:267-279.

• Knighton DR, Hunt TK, Sceuensthuhl H, Halliday BJ, Werb Z, Banda MJ. Oxygen tension regulates the expression of angiogenesis factor by macrophages. Science 1983, 221:1283.

• Kozak LJ, Bacon WD, Krzyzanowski M, Wojtyniak B. Hospital use in Poland and the Unites States: In: U.S. Department of Health and Human Services Publication No. (PHS) 88-1478. Vital and Health Statistics, Series 5, No 2. Washington, DC:Goverment Printing Office 1988

• Kozian DH, Ziche M, Augustin HG. The activin-binding protein follistatin regulates autocrine endothelial cell activity and induces angiogenesis. Lab Invest 1997, 76:267–276.

• Krystal GW, Hines SJ, Organ CP. Autocrine growth of small cell lung cancer mediated by coexpression of c-KIT and stem cell factor. Cancer Res 1996, 56:370-376.

• Kubota Y, Angelotti T, Niederfellner G, Herbst R, Ullrich A. Activation of phosphotidylinositol 3-kinase is necessary for differentiation of FDC-P1 cells

Page 158: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

VIII

following stimulation of type III receptor tyrosine kinases. Cell Growth Differ 1998, 9:247–256.

• Ladin D, Garner WL, Smith DS Jr. Excessive scarring as a consequence of healing. Wound Repair Regen 1995, 3:6-14.

• Ladin D. The art and science of wound care. Plast Reconstr Surg 1995, 96:748. • Lahav M, Cadet JC, Chirambo M, Rehani U. Ishii Y. Corneal Keloids- a

histopathological study. Graefes Arch Exp Opthalmol 1982, 218:256-261. • Laine P, Reunanen N, Ravanti L, Foschi M, Santra M, Iozzo RV, Kahari VM.

Activation of extracellular signal-regulated protein kinase1,2 results in down regulation of decorin expression in fibroblasts. J Biol Chem 2000, 349: 19-25.

• Lammie A, Drobnjak M, Gerald W, Saad A, Cote R, Cordon-Cardo C. Histochem Cytochem 1994, 42:1417-1425.

• Larabee WF, East CA, Jaffe HS, Stephenson C, Peterson KE. Intralesional interferon gamma treatment for keloids and hypertrophic scars. Arch Otolaryngol Head Neck Surg 1990, 116:1159-1162.

• Layton, AM, Yip, J, Cunliffe, WJ. A comparison on intralesional triamcinolone and cryosurgery in the treatment of acne keloids. Br J Dermatol 1994, 130: 498.

• Le AD, Zhang Q, Wu Y, Messadi DV, Akhondzadeh A, Nguyen AL, Aqhaloo TL, Kelly AP, Bertolami CN. Elevated vascular enothelial growth factor in keloids: relevance to tissue fibrosis. Cell Tissues Organs 2004, 176:87-94.

• Lee JY, Yang CC, Chao SC, Wong TW. Histopathological differential diagnosis of keloid and hypertrophic scar. Am J Dermatopathol 2004, 26:379-384.

• Lee TY, Chin GS, Kim WJ, Chau D, Gittes GK, Longaker MT. Expression of transforming growth factor beta 1,2,3 proteins in keloids. Ann Plast Surg 1999, 43:179-184.

• Lefevre G, Gliotin AL, Calipel A, Mouriaux F, Tran T, Spinelli M, Rosolen A, Ricotti E, Basso G. Roles of stem cell factor/c-KIT and effects of Glivec/ST1571 in human uveal melanoma cell tumorigenesis. J Biol Chem 2004, 279:31769-31779.

• Leung DW, Cachianes G, Kuang WJ, Goeddel, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 1989, 246: 1306.

• Lewis TS, Shapiro PS, Ahn NG. Vascular endothelial growth factor is a secreted angiogenic mitogen. Adv Cancer Res 1998, 74:49-139.

• Lim CP, Phan TT, Lim IJ, Cao X. Stat3 contributes to keloid pathogenesis via promoting collagen production, cell proliferation and migration. Oncogene 2006, 25:5416-5425.

• Lim IJ, Phan TT, Bay BH, QI R, Huynh H, Tan WTL, Lee ST, Longaker MT. Fibroblasts co-cultured with keloid keratinocytes: Normal fibroblasts secrete collagen in keloid like manner. Am J Physiol Cell Physiol 2002, 283:C212-C222.

• Lim IJ, Phan TT, Song C, Tan WTL, Longaker MT. Investigation of the influence of keloid-derived keratinocytes on fibroblast growth and proliferation in vitro. Plast Reconstr Surg 2001, 107:797-808.

• Lim IJ, Phan TT, Tan EK, Nguyen TT, Tran E, Longaker MT, Song C, Lee ST, Hunyh H. Synchronous activation of ERK and phosphotidylinositol3-kinase pathways is required for collagen and extracellular matrix production in keloids. J Biol Chem 2003, 278:40851-40858.

Page 159: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

IX

• Liu QY, Niranjan B, Gomes P, Gomm JJ, Davies D, Coombes RC, Buluwela L. Inhibitory effects of activin on the growth and morphogenesis of primary and transformed mammary epithelial cells. Cancer Res 199656:1155-1163.

• Liu W, Cai Z, Wang D, Wu X, Cui L, Shang Q, Qian Y, Cao Y. Blocking transforming growth factor- beta receptor signaling down regulates transforming growth factor beta 1 autoproduction in keloid fibroblasts. Chin J Traumatol 2002, 5:77-81.

• Logan A, Baird A, Berry M. Decorin attenuates gliotic scar formation in the rat cerebral hemisphere. Exp Neurol 1999, 159: 504-510.

• Lories V, Cassiman JJ, Berghe HVV, David G. Differential expression of cell surface heparan sulphate proteoglycans in human mammary epithelial cells and lung fibroblasts. J Biol Chem 1992, 267:1116-1122.

• Lozzo RV, Murdoch A. Proteoglycans of the extracellular environment: clues from the gene and protein side offer novel perspectives in molecular diversity and function. FASEB J 1996, 10: 598-614.

• Lozzo RV. Matrix proteoglycans: from molecular design to cellular function. Annu Rev Biochem 1998, 67: 609-652.

• Lyman SD, Jacobsen SE. c-KIT ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities. Blood 1998, 91:1101-1134.

• Maas-Szabowski N, Stark HJ, Fusenig NE. Keratinocyte growth regulation in defined organotypic cultures through IL-1 induced keratinocyte growth factor expression in resting fibroblasts. J Invest Dermatol 2000, 114:1075-1084.

• Mackenzie IC. Epithelial-mesenchymal interactions in the development and maintenance of epithelial tissues. In The Keratinocyte Handbook, Leigh I, Lane B, Watt F (eds). Cambridge University Press: Cambridge, UK, 1994.

• Majumder S, Brown K, Qiu FH, Besmer P. c-KIT protein, a transmembrane kinase: identification in tissues and characterization. Mol Cell Biol 1988, 8:4896-4903.

• Maldonado TS, Kadison AS, Crisera CA, Grau JB, Alkasab SL, Longaker MT, Gittes GK. Ontogeny of activin B and follistatin in developing embryonic mouse pancreas: implications for lineage selection. J Gastrointest Surg 2000, 4:269-275.

• Manuskiatti W, Fitzpatrick RE. Treatment response of keloidal and hypertrophic sternotomy scars: comparison among intralesional corticosteroid, 5 Flourouracil and 585- nm flash lamp pumped pulsed dye laser treatments. Arch Dermatol 2002, 138:1149-1155.

• Marneros AG, Krieg T. Keloids - clinical diagnosis, pathogenesis, and treatment options. J Dtsch Dermatol Ges 2004, 2:905-913.

• Mayrhofer G, Gadd SJ, Spargo LD, Ashman LK. Specificity of a mouse monoclonal raised acute myeloid leukemia cells for mast cells in human mucosal and connective tissues. Immunol Cell Biol 1987, 65:241-250.

• Massague J. The transforming growth factor –β family. Annu Rev Cell Biol 1990, 6:597-641.

• Mathews LS, Vale WW. Molecular and functional characterization of activin receptors. Receptors 1993, 3:173-181.

Page 160: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

X

• Mathews LS. Activin receptors and cellular signalling by the receptor serine kinase family. Endocr Rev 1994, 15:310–325.

• Matsuse T, Ikegami A, Ohga E, Hosoi T, Oka T, Kida K, Fukayama M, Inoue S, Nagase T, Ouchi Y, Fukuchi Y. Expression of immunoreactive activin A protein in remodeling lesions associated with interstitial pulmonary fibrosis. Am J Pathol 1996, 148:707-713.

• McCarthy SA, Bicknell R. Inhibition of vascular endothelial cell growth by activin-A. J Biol Chem 1993, 268:23066–23071.

• Milsom JP, Craig RD. Collagen degradation in cultured keloid and hypertrophic scar tissue. Br J Dermatol 1973, 89:635.

• Miralles F, Czernichow P, Scharfmann R. Follistatin regulates the relative proportions of endocrine versus exocrine tissue during pancreatic development. Development 1998, 125:1017-1024.

• Miyazawa K, Toyama K, Gotoh A, Hendrie PC, Mantel C, Broxmeyer HE. Ligand-dependent polyubiquitination of c-kit gene product: a possible mechanism of receptor down modulationin M07e cells. Blood 1994, 83:137-145.

• Montesano R, Orci L. Transforming growth factor-β stimulates collagen-matrix contraction by fibroblasts: implications for wound healing. Proc Natl Acad Sci 1998, 85:4894–7.

• Munz B, Smola H, Engelhardt F, Bleuel K, Brauchle M, Lein I, Evans LW, Huylebroeck D, Balling R, Werner S. Overexpression of activin A in the skin of transgenic mice reveals new activities of activin in epidermal morphogenesis, dermal fibrosis and wound repair. EMBO J 1999, 18:5205-5215.

• Murray JC, Pollack SV, Pinnell SR. Keloids: A review. J Am Acad Dermatol 1981, 4:461-470.

• Mustoe T A, Cooter R D, Gold M H, Hobbs R, Ramelet A, Shakespeare P G, Stella M, Teot L, Wood F M, Ziegler U E. International clinical recommendations on scar management. Plastic and Reconstructive Surgery 2002, 110:560-571.

• Mutalik S. Treatment of keloids and hypertrophic scars. Indian J Dermatol Venereol Leprol 2005, 71:3-8.

• Nanda S, Reddy BSN. Intralesional 5-flurouracil as a treatment modality of keloids. Dermatol Surg 2004, 30:54-57.

• Natali PG, Nicotra MR, Sures I, Santoro E, Bigotti A, Ullrich A. Cancer Res 1992, 52:6139-6143.

• Nedelec B, Tredget EE, Ghahary A. The molecular biology of wound healing following thermal injury: the role of fibrogenic growth factors. In: Critical Care of Burn Patient. Barcelona: Springer-Verlag, 1996.

• Ng LE, Vincent AS, Halliwell B, Wong KP. Action of diclofenac on kidney mitochondria and cells. Biochem Biophys Res Commun 2006, 348, 494-500.

• Nocka K, Majumder S, Chabot B, Ray P, Cervone M, Bernstein A, Besmer P. Expression of c-KIT gene products in known cellular targets of W mutations in normal and W mutant mice-evidence for an impaired c-KIT kinase in mutant mice. Genes Dev 1989, 3:816-826.

• Norris JEC. Superficial X-ray Therapy in keloid management: A retrospective study of 24 cases and literature review. Plast Reconstr Surg 1995; 95:1051.

Page 161: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XI

• Ohga E, Matsuse T, Teramoto S, Katayama H, Nagase T, Fukuchi Y, Ouchi Y. Effects of activin A on proliferation and differentiation of human lung fibroblasts. Biochem Biophys Res Commun 1996, 12:391-396.

• Oluwasanmi JO. Keloids in the African. Clin Plast Surg 1974, 1:179-195. • Omo-Dare P. Genetic studies on keloid. J Natl Med Assoc 1975, 67:428-432. • Ong C, Khoo Y, Tan E, Mukhopadhyay A, Do D, Han H, Lim I, Phan T.

Epithelial-mesenchymal interactions in keloid pathogenesis modulate vascular growth factor expression and secretion. J Pathol 2007, 211:95-108.

• Park H, Kim Y, Lim Y, Han I and Oh E S. Syndecan-2 mediates adhesion and proliferation of colon carcinoma cells. J Biol Chem 2002, 277(33): 29730-29736.

• Peltonen J, Hsiao LL, Jaakkola S, Sollberg S, Aumailley M, Timpl R, Chu ML, Uitto J. Activation of collagen gene expression in keloids: co-localization of type I and VI collagen and transforming growth factor-beta 1 mRNA. J Invest Dermatol 1991, 97:240-248.

• Peters M, Jacobs S, Ehlers M, Vollmer P, Mullberg J, Wolf E, Brem G, Meyer zum buschenfelde KH, Rose-John S. The function of the soluble interleukin 6 receptor in vivo: Sensitization of human soluble IL-8 receptor transgenic mice towards IL-6 and prolongation of the plasma half-life of IL-6. J Exp Med 1996, 183:1399-13406.

• Phan TT, Lim IJ, Aalami O, Lorget F, Khoo A, Tan EK, Mukhopadhyay A, Longaker MT. Smad 3 signaling plays an important role in keloid pathogenesis via epithelial-mesenchymal interactions. J Pathol 2005, 207:232-242.

• Phillips DJ, de Kretser DM. Follistatin. A multifunctional regulatory protein. Front Neuroendocrinol 1998, 19:287-332.

• Phillips DJ. The activin/inhibin family, In: The Cytokine Handbook (4th ed), (Thomson AW and Lotze MT eds) Academic Press, Amsterdam, Boston, 2003: 1153-1177.

• Polo M, Smith PD, Kim YJ, Wang X, Ko F, Robson MC. Effect of TGF-beta2 on proliferative scar fibroblast cell kinetics. Ann Plast Surg 1999, 43:185-190.

• Poochareon VN, Berman B. New therapies for the management of keloids. J Craniofac Surg 2003, 14:654-657.

• Pratibha V, Gupta V D. Cutaneous wound healing. Significance of proteoglycans in scar formation. Curr Sci 2000, 78:1-5.

• Raghow R. The role of extracellular matrix in post inflammatory wound healing and fibrosis. FASEB J 1994, 8:823-831.

• Ren X, Hogaboam C, Carpenter A, Colletti L. Stem cell factor restores hepatocyte proliferation in IL-6 knockout mice following 70% hepatectomy. J Clin Invest 2003, 112:1407-1418.

• Ritvos O, Tuuri T, Eramaa M, Sainio K, Hilden K, Saxen L, Gilbert SF. Activin disrupts epithelial branching morphogenesis in developing glandular organs of the mouse. Mech Dev 1995, 50:229-245.

• Roberts VJ, Barth SL. Expression of messenger ribonucleic acids encoding the inhibin/activin system during mid- and late-gestation rat embryogenesis. Endocrinol 1994, 134:914-923.

• Robles Dt, Berg D. Abnormal wound healing: keloids. Dermatol 2007, 25:26-32.

Page 162: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XII

• Rossmanith W, Chabicovsky M, Grasl-Kraupp B, Peter B, Schausberger E, Schulte-Hermann R. Follistatin overexpression in rodent liver tumors: a possible mechanism to overcome activin growth control. Mol Carcinog 2002, 35(1):1-5.

• Roy SG, Nozaki Y, Phan SH. Regulation of α- smooth muscle actin expression in myofibroblast differentiation from rat lung fibroblast. Int J Biochem Cell Biol 2001, 33:723-734.

• Rudolph R, Berg JV, Ehrlich HP. Wound contraction and scar contracture. In: Cohen IK, Diegelmann RE, Lindblad WJ (eds) Wound Healing: Biochemical and Clinical Aspects. Saunders, Philadelphia, 1992, pp 96-114.

• Ruoslahti, E, Yamaguchi, Y. Proteoglycans as modulators of growth factor activities. Cell 1991, 64, 867-869.

• Rusel JD, Russell SB, Trupin KM. The effect of histamine on the growth of cultured fibroblasts isolated from normal and keloid tissue. J Cell Physiol 1977, 93:389-393.

• Russell R, Horlock N, Gault D. Zimmer splintage. A simple effective treatment for keloids following ear piercing. Br J Plast Surg 2001, 54:509.

• Santra M, Skorski T, Calabretta B, Lattime EC, Iozzo RV. De novo decorin gene expression suppresses the malignant phenotype in human colon cancer cells. Proc Natl Acad Sci 1995, 92:7016-7020.

• Sattler M, Salgia R. Targeting c-KIT mutations. basic science to novel therapies. Leuk Res 2004, 28:S11-S20.

• Sayah DN, Chia S, Shaw WW, Watson J, Messad D, Longaker MT. Down-regulation of apoptosis-related genes in keloid tissues. J Surg Res 1999, 87:209-216.

• Schwall RH, Robbins K, Jardieu P, Chang L, Lai C, Terrell TG. Activin induces cell death in hepatocytes in vivo and in vitro. Hepatol 1993, 18:347-356.

• Sebestyen A, Gallai M, Knittel T, Ambrust T, Ramadori G, Kovalszky I. Cytokine regulation of syndecan expression in cells of liver origin. Cytokine 2000, 12: 1557–1560.

• Seidel C, Sundan A, Hjorth M, Turesson I, Dahl IM, Abildqaard N. Serum syndecan-1: a new independent prognostic marker in multiple myeloma. Blood 2000, 95: 388-392.

• Seishima M, Nojiri M, Esaki C, Yoneda K, Eto Y, Kitajima Y. Activin A induces terminal differentiation of cultured human keratinocytes. J Invest Dermatol 1999, 112:432-436.

• Serve H, Yee NS, Stella G, Sepp-Lorenzino L, Tan JC, Besmer P. Differential roles of PI3-kinase and kit tyrosine 821 in kit receptor-mediated proliferation, survival and cell adhesion in mast cells. EMBO J 1995, 14:473–483.

• Shah M, Foreman DM, Ferguson MW. Neutralizing antibody to TGF-beta 1, 2 reduces cutaneous scarring in adult rodents. J Cell Sci 1994, 107:1137–1157.

• Shons AR, Press BH. The treatment of earlobe keloids by surgical excision and postoperative triamcinolone injection. Ann Plast Surg 1983, 10:480-482.

• Shoukrey NM, Tabbara KF, Ultrasctructural study of a corneal keloid. Eye 1993, 7:379-387.

Page 163: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XIII

• Shukla IM, Arora NP, Arora NM. Corneal Keloid. Indian J Opthalmol 1975, 23:18-19.

• Skobe M, Fusenig E. Tumurogenic conversion of immortal human keratincoytes through stromal cell invasion. Proc Natl Acad Sci 1998, 95: 1050-1055.

• Smith P, Mosiello G, Deluca L, Ko F, Maggi S, Robson MC. TGF-ß2 activates proliferative scar fibroblasts. J Surg Res 1999, 82:319- 323.

• Solberg S, Peltonen J, Uitto J. Combined use of in situ hybridization and unlabeled antibody peroxidase methods: Simultaneous detection of type I procollagen mRNAs and factor VIII-related antigen epitopes in keloid tissue. Lab Invest 1991, 64:125.

• Sproat JE, Dalcin A, Weitauer N, Roberts RS. Hypertrophic sternal scars: Silicone gel sheet versus Kenalog injection treatment. Plast Reconstr Surg 1992, 90:988.

• Spyrou GE, Naylor IL. The effect of basic fibroblast growth factor on scarring. Br J Plast Surg 2002, 55:275-282.

• Subramanian SV, Fitzgerald ML, Bernfield M. Regulated shedding of syndecan-1 and -4 ectodomains by thrombin and growth factor receptor activation. J Biol Chem 1997, 272: 14713-20.

• Sugino H, Sugino K, Hashimoto O, Shoji H, Nakamura T. Follistatin and its role as an activin-binding protein. J Med Invest 1997, 44:1-14.

• Sugiyama M, Ichida T, Sato T, Ishikawa T, Matsuda Y, Akasura H. Expression of activin A is increased in cirrhotic and fibrotic rat livers. Gastroenterology 1998, 114:550-558.

• Tajima F, Kawatani T, Ishiga K, Nanba E, Kawasaki H. Serum soluble c-KIT receptor and expression of c-KIT protein and mRNA in acute myeloid leukemia. Eur J Haematol 1998, 60:289-296.

• Tang YW. Intra and postoperative steroid injection for keloids and hypertrophic scars. Br J Plast Surg 1992, 45:371-373.

• Mustoe TA, Cooker RD, Gold MH, Hobbs FDR, Ramelet AA, Shakespeare PG, Maurizio S, Test L, Wood FM, Ulrich EZ. International clinical recommendations on scar management. Plast Reconstr Surg 2002, 110:560-571.

• Thommes K, Lennartsson J, Carlberg M, Ronnstrand L. Identification of Tyr-703 and Tyr-936 as the primary association sites for GRb2 and GRb7 in the c-KIT stem cell factor receptor. Biochem J 1999, 341: 211-216.

• Timokhina I, Kissel H, Stella G, Besmer P. Kit signaling through PI3-kinase and Src kinase pathways: an essential role for Rac1 and JNK activation in mast cell proliferation. EMBO J 1998, 17:6250–6262.

• Tomasek JJ, Gabbiani G, Hinz B, Chapponier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodeling. Nat Rev Mol Cell Biol 2002, 3:349-363.

• Tredget EE, Shankowsky HA, Pannu R, Nedelec B, Iwashina T, Ghahary A, Taerum TV, Scott PG. Transforming growth factor beta in thermally injured patients with hypertrophic scars: effects of interferon alpha-2b. Plast Reconstr Surg 1998, 102:1317-1328.

Page 164: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XIV

• Tredget EE, Shankowsky HA, Taerum TV, Movsa GL, Alton JDM. The role of inhalation injury in burn trauma: a Canadian experience. Ann Surg 1990, 212:720-727.

• Tuan T, Nichter LS. The molecular basis of keloid and hypertrophic scar formation. Mol Med Today 1998: 19-24.

• Tuan TL, Zhu JY, Sun B, Nichter LS, Nimni ME, Laung WE. Elevated levels of plasminogen activator inhibitor-1 may account for the altered fibrinolysis by keloid fibroblasts. J Invest Dermatol 1996, 106:1007.

• Tuuri T, Eramaa M, Hilden K, Ritvos O. The tissue distribution of activin ßA- and ßB-subunits and follistatin messenger ribonucleic acids suggests multiple sites of action for the activin-follistatin system during human development. J Clin Endocrinol Metab 1994, 78:1521-152.

• Uchida G, Yoshimura K, Kitano Y, Okazaki M, Harii K. Tretinoin reverses upregulation of matrix metalloproteinase-13 in human keloid-derived fibroblasts. Exp Dermatol 2003, 12:35.

• Uitto J. IL-6 Signaling Pathway in Keloids: A Target for Pharmacologiv Intervention. J Invest Dermatol 2007, 127: 6-8.

• Vale W, Hsueh A, Rivier C, Yu J. The inhibin/activin family of hormones and growth factors, In: Peptide Growth Factors and Their Receptors II (Sporn MD, Roberts AB eds) Springer-Verlag, Berlin, Germany, 211-248, 1990.

• Vale W, Rivier J, Vaughan J, McClintock R, Corrigan A, Woo W, Karr D, Speiss J. Purification and characterization of an FSH releasing protein from the porcine ovarian follicular fluid. Nature 321:776-779, 1986.

• Vincent AS, Lim BG, Tan J, Whiteman M., Cheung NS, Halliwell B, Wong KP. Sulfite-mediated oxidative stress in kidney cells. Kidney Int 2004, 65, 393-402.

• Vosseller K, Stella G, Yee NS, Besmer P. c-KIT receptor signaling through its phophotidylinsositide-3’-kinase-binding site and protein kinase C: role in mast cell enhancement of degranulation, adhesion, and membrane ruffling. Mol Biol Cell 1997, 8:909–922.

• Wang C, Curtis JE, Geissler EN, McCulloch EA, Minden MD. The expression of the proto-oncogene c-KIT in the blast cells of acute myeloblastic leukemia. Leukemia 1989, 3:816-826.

• Wang S, Wilkes MC, Leof EB, Hirschberg R. Imatinib mesylate blocks a non-Smad TGF-beta pathway and reduces renal fibrogenesis in vivo. FASEB 2005, 19:1-11.

• Wankell M, Munz B, Hubner G, Hans W, Wolf E, Goppelt A, Werner S. Impaired wound healing in transgenic mice overexpression the activin antagonist follistatin in the epidermis. EMBO J 2001, 20:5361-5372.

• Wegrowski Y, Gillery P, Kotlarz G, Perreau C, Georges N, Maquart FX. Modulation of sulfated glycosaminoglycan and small proteoglycan synthesis by the extracellular matrix. Mol Cell Biochem 2000, 205: 125–131.

• Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev 2003, 83:835-870.

• Werner S, Smola H. Paracrine regulation of keratinocyte proliferation and differentiation. Trends Cell Biol 2001, 11:143-146.

Page 165: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XV

• Werner S, Weinberg W, Liao X, Peter KG, Blessing M, Yuspa SH, Weiner RL, Williams LT. Targeted expression of a dominant-negative FGF receptor mutant in the epidermis of transgenic mice reveals a role of FGF in keratinocyte organization and differentiation. EMBO J 1993, 2:2635-2643.

• Williams DE, Eisenman J, Baird A, Rauch C, Van Ness K, March CJ, Park LS, Martin U, Mochizuki DY, Boswell HS. Identification of a ligand for the c-KIT proto-oncogene. Cell 1990, 63:167-174.

• Worapamorn W, Haase HR, Li H, Bartold PM. Growth factors and cytokines modulate gene expression of cell-surface proteoglycans in human periodontal ligament cells. J Cell Physiol 2001, 186: 448–456.

• Wu Y, Zhang Q, Ann DK, Akhondzadeh A, Duong HS, Messadi DV, Le AD. Increased vascular endothelial growth factor may account for elevated level of plasminogen inhibitor-1 via activating ERK1/2 in keloid fibroblasts. Am J Physiol Cell Physiol 2004, 286:C905-912.

• Xia W, Phan TT, Lim IJ, Longaker MT, Yang GP. Complex epithelial – mesenchymal interactions modulate transforming growth factor-β expression in keloid-derived cells. Wound Repair Regen 2004, 12:546-556.

• Xue H, McCauley RL, Zhang W. Elevated interleukin-6 expression in keloid fibroblasts. J Surg Res 2000, 89:74-77.

• Yamaoka T, Idehara C, Yano M, Matsushita T, Yamada T, Ii S, Moritani M, Hata J, Suqino H, Noji S, Itakura M. Hypoplasia of pancreatic islets in transgenic mice expressing activin receptor mutants. J Clin Invest 1998, 102:294-301.

• Yamashita S, Maeshima A, Kojima I, Nojima Y. Activin A Is a Potent Activator of Renal Interstitial Fibroblasts. J Am Soc Nephrol 2004, 15:91-101.

• Yang GP, Lim IJ, Phan TT, Lorenz HP, Longaker MT. From scarless wounds to keloids: molecular studies in wound healing. Wound Repair Regen 2003; 11: 411–18.

• Yarden Y, Kuang WJ, Yang-Feng T, Coussens L, Munemitsu S, Dull TJ Chen E, Schlessinger J, Francke U, Ullrich A. Homo proto-oncogene c-KIT: a new cell surface receptor tyrosine kinase for an unidentified ligand. EMBO J 1987, 6:3341-3351.

• Yee NS, Hsiau CW, Serve H, Vosseller K, Besmer P. Mechanism of down-regulation of c-KIT receptor tyrosine kinase, phosphotidylinositol 3’-kinase, and protein kinase C. J Biol Chem 1994, 269:31991-31998.

• Yee NS, Langen H, Besmer P. Mechanism of kit ligand, phorbol ester, and calcium induced down regulation of c-KIT receptors in mast cells. J Biol Chem 1993, 268:14189–14201.

• Yoshimoto H, Ishihara H, Ohtsuru A, Akino K, Murakami R, Kuroda H, Namba H, Ito M, Fujii T, Yamashita S. Overexpression of insulin-like growth factor-1 (IGF-1) receptor and the invasiveness of cultured keloid fibroblasts. Am J Pathol 1999, 154:883-889.

• Younai S, Nichter LS, Wellisz T, Reinisch J, Nimni ME, Tuan TL. Modulation of collagen synthesis by transforming growth factor-beta in keloid and hypertrophic scar fibroblasts. Ann Plast Surg 1994, 33:148-151.

Page 166: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XVI

• Zandvoort A, Postma DS, Jonker MR, Noordhoek JA, Vos JT, van der Geld YM, Timens W. Altered expression of the smad signaling pathway: Implications for COPD pathogenesis. Eur Respir J 2006, 28: 533-541.

• Zhang HY, Phan SH. Inhibition of myofibroblast apoptosis by transforming growth factor β1. Am J Respir Cell Mol Biol 1999, 21:658-665.

• Zhang Q, Oh CK, Messadi DV, Duong HS, Kelly AP, Soo C, Wang L, Le AD. Hypoxia-induces HIF-1 alpha accumulation is augmented in co-culture of keloid fibroblasts and human mast cells: Involvement of ERK1/2 and P1-3K/Akt. Exp Cell Res 2006, 312:145-155.

• Zhang X, Vincent AS, Halliwell B and Wong KP. A mechanism of sulphite neurotoxicity: direct inhibition of glutamate dehydrogenase. J Biol Chem 2004, 279:43035-43045.

• Zhao Y, Silbajoris R, Young SL. Identification and developmental expression of two activin receptors in baboon lung. Biochem Biophys Res Commun 229:50-57, 1996.

• Zimmermann P, David G. The syndecans, tuners of transmembrane signaling. FASEB J 1999, 13:S91-s100.

• Zou J, Zhu F, Liu F, Wang W, Garlisi CG, Liu YH, Wang S, Shah H, Wan Y, Umland SP. Catalytic activity of human ADAM33. J Biol Chem 2004, 279:9818-9830.

• Zsebo KM, Williams DA, Geissler EN, Broudy VC, Martin FH, Atkins EL, Hsu RY, Birkett NC, Okino KH, Murdock DC. Stem cell factor is encoded at the S1 locus of the mouse and is the ligand for the c-KIT tyrosine kinase receptor. Cell 1990, 63:213-224.

Page 167: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XVII

Appendix 1

List of patients

NORMAL NF/NK1 NF2 NF/NK4 NF8 NF/NK9 NF/NK5 NF/NK3 NS31 NS32 NS33 NS34 NS35 KELOID KF6 KF46 KF3 KS/KF/KK48 KF/KK31 KF/KK32 KF/KK43 KF/KK16 KS/KF/KK 25 KF/KK30 KF/KK45 KF/KK24 KF/KK29 KS50 KS51 KS52 KS53 KS54 KS55 KS56 KS57

F M M F F F F F M M F M F M F F F M M F M F F M F M F M M F M F

Chinese Indian Chinese Malay Chinese Chinese Chinese Malay Chinese Chinese Chinese Malay Chinese Chinese Malay Indian Chinese Indian Chinese Chinese Indian Chinese Malay Malay Indian Malay Chinese Indian Malay Malay Chinese Chinese

27 years 32 years 3 years 28 years 24 years 35 years 12 years 13 years 20 years 29 years 35 years 33 years 16 years 36 years 20 years 23 years 17 years 28 years 34 years 19 years 25 years 30 years 23 years 21 years 18 years 10 years 18 years 21 years 28 years 16 years 25 years 22 years

Breast Abdominal Earlobe Breast Breast Breast Groin Groin Forearm Thigh Breast Forearm Earlobe Forearm Earlobe Earlobe Earlobe Chest Elbow Earlobe Forearm Face Forearm Chest Earlobe Face Earlobe Elbow Face Forearm Earlobe Earlobe

- - - - - - - - - - - - 1 1 1.5 1.5 0.5 2 1 1.5 1 1 1.5 2 1 1 1.5 1 1.5 1 2 1

Page 168: INVESTIGATIONS ON KELOID PATHOGENESIS AND THERAPY · interactions in keloid scar pathogenesis and carcinogenesis. A review. Current Signal Transduction Therapy (invited review paper,

XVIII