101
|
Enoch S, Moseley R, Stephens P, Thomas D. The oral mucosa: a model of wound healing with reduced scarring. ACTA ACUST UNITED AC 2008. [DOI: 10.1111/j.1752-248x.2007.00005.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
102
|
Alonso PE, Rioja LF, Pera C. Keloids: A viral hypothesis. Med Hypotheses 2008; 70:156-66. [PMID: 17509771 DOI: 10.1016/j.mehy.2007.03.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Accepted: 03/20/2007] [Indexed: 11/30/2022]
Abstract
The triggering cause of keloid formation on a healing wound remains an enigma. In fact, the hypotheses put forward so far to explain this phenomenon seem inconsistent with some clinical features of the disease. The recently established bonds between infectious agents and some pathologies of unknown origin such as peptic ulcer disease, Kaposi's sarcoma or cervical cancer among others led us to consider a potential infectious origin for keloids. This paper presents an infection-based hypothesis (specifically, a viral hypothesis) intended to account for most of their clinical features. Essentially, we hypothesize that healthy individuals carrying a virus, whether known or unknown, associated to some adjuvant, and having some genetic susceptibility, may develop keloids during the scar maturation process in the following manner: the virus would make the bone marrow or lymphatic system its reservoir, residing there in a silent state, and reach the wound via two different mechanisms. The primary mechanism might use an internal circuit through which the viral genome would be transported from its myeloid reservoir to the wound via bone marrow or circulating fibrocytes chemotactically attracted to the damaged skin region. The secondary mechanism might involve an external circuit by which infecting virions via saliva would be shed in the wound directly (preferentially in the sternal or deltoid region) or indirectly (other satellite regions) via the hands or some fomites. A combination of both mechanisms might also be possible. Once in the wound, the virus would switch from a silent state to a latent state by effect of some chemical stimulus probably generated during the tissue repair process; in the new state, the transcription of some of the powerful viral proteins might cause thorough derailment of the normal repair process. As a result, keloid growth might depend both on individual susceptibility and on the viral load deposited into the wound; the greater the susceptibility and viral load were, the more markedly the keloid would develop and the more aggressive it would be.
Collapse
Affiliation(s)
- Pedro E Alonso
- Plastic Surgery Unit, Hospital USP Marbella, Avda. Severo Ochoa, 22, Marbella, Málaga, Spain.
| | | | | |
Collapse
|
103
|
Chu Y, Guo F, Li Y, Li X, Zhou T, Guo Y. A novel truncated TGF-beta receptor II downregulates collagen synthesis and TGF-beta I secretion of keloid fibroblasts. Connect Tissue Res 2008; 49:92-8. [PMID: 18382895 DOI: 10.1080/03008200801913924] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Hypertrophic scars and keloid are dermal proliferative disorders in wound healing. Transforming growth factor beta (TGF-beta) has been implicated in scar formation through the activation of fibroblasts and the acceleration of collagen deposition. Our study aimed to design a novel truncated (27-123 residues) type II TGF-beta receptor (tTGFbetaRII) and to determine its effects on the proliferation of keloid fibroblasts and the collagen synthesis as well as TGF-beta I expression of the cells. The coding sequences of TGF-beta I and tTGFbetaRII were amplified using RT-PCR and then cloned into pGBKT7 and pGADT7 vectors. A yeast two-hybrid experiment and a glutathione S-transferase (GST)-pull down assay were performed to verify the affinity of tTGFbetaRII to TGF-beta I. Our results indicated that treatment with tTGFbetaRII inhibited the growth of keloid fibroblasts and suppressed the synthesis of type I collagen in keloid fibroblasts in a concentration-dependent manner. Moreover, northern and western blot analysis revealed a decline of the TGF-beta I expression at both mRNA and protein levels after exposure to 5, 10 or 20 mug/ml of tTGFbetaRII. Together, our data suggested that the exogenous tTGFbetaRII can efficiently trap TGF-beta I from access to wild-type receptors and can suppress TGF-beta I triggered signals. Thus it may potentially be clinically applied to scar therapy.
Collapse
Affiliation(s)
- Yanhui Chu
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
104
|
Wang Z, Gao Z, Shi Y, Sun Y, Lin Z, Jiang H, Hou T, Wang Q, Yuan X, Zhu X, Wu H, Jin Y. Inhibition of Smad3 expression decreases collagen synthesis in keloid disease fibroblasts. J Plast Reconstr Aesthet Surg 2007; 60:1193-9. [PMID: 17889631 DOI: 10.1016/j.bjps.2006.05.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2005] [Revised: 04/30/2006] [Accepted: 05/09/2006] [Indexed: 12/22/2022]
Abstract
BACKGROUND Keloids represent a dysregulated response to cutaneous wounding that result in an excessive deposition of extracellular matrix (ECM), especially types I and III collagen. In keloid scars, the ratio of 'type I to type III collagen' varies compared to normal skin. Transforming growth factor beta (TGF-beta) plays a central role in the pathogenesis of fibrosis by inducing and sustaining activation of keloid fibroblasts. However, the underlying mechanisms are poorly understood. RNA interference (RNAi) is an evolutionally conserved mechanism for repressing targeted gene expression. In mammalian cells, RNAi is mediated by small interfering RNA (siRNA). In this paper, we examined the function of Sma and Drosophila mothers against decapentaplegic homolog 3 (Smad3), recently characterized as intracellular effector of TGF-beta signalling, in keloid fibroblasts using siRNA. METHODS Dermal fibroblasts obtained from one female patient aged 21 years were maintained in Dulbecco's modified Eagle's medium. Cells (<6 passages) were treated with or without Smad3 siRNA and the expression levels of related genes were examined by Reverse Transcription Polymerase Chain Reaction (RT-PCR) and Western Blot. Statistical analysis was performed using one-way ANOVA (Dunnett correction) and the Excel 7.0 software, with significance set at p<0.05. RESULTS The knockdown of Smad3 expression in mRNA and protein levels was confirmed using RT-PCR and Western Blot. Compared to blank, the mRNA levels of types I and III procollagen were also significantly and uniquely decreased following the reduction of Smad3 by siRNA (p<0.05). The results indicate that Smad3 plays an important role in the TGF-beta induced fibrosis in keloid. Downregulation of Smad3 expression in keloid fibroblasts can significantly decrease procollagen gene expression. SiRNA targeting Smad3 was an efficient reagent to reduce ECM deposition and attenuate process of fibrosis. It could be a new promising therapeutic approach to improve skin wound healing and inhibit progression of fibrotic conditions by interrupting the TGF-beta signal pathway.
Collapse
Affiliation(s)
- Zimin Wang
- Department of Orthopaedics, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Increased CCN2 Transcription in Keloid Fibroblasts Requires Cooperativity Between AP-1 and SMAD Binding Sites. Ann Surg 2007; 246:886-95. [DOI: 10.1097/sla.0b013e318070d54f] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
106
|
Satterwhite TS, Chong AK, Luo J, Pham H, Costa M, Longaker MT, Wyss-Coray T, Chang J. In Vitro Analysis of Transforming Growth Factor-β1 Inhibition in Novel Transgenic SBE-Luciferase Mice. Ann Plast Surg 2007; 59:207-13. [PMID: 17667417 DOI: 10.1097/01.sap.0000252732.25168.34] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Transforming growth factor beta1 (TGF-beta1) expression correlates with scarring. A novel transgenic mouse model with a Smad2/3-responsive luciferase reporter construct (SBE-luc) has been developed. We hypothesized that bioluminescence in SBE-luc dermal fibroblasts could be measured to assess TGF-beta1 inhibition. MATERIALS AND METHODS Cultured dermal fibroblasts from SBE-luc mice were treated simultaneously with TGF-beta1 and increasing doses of either neutralizing antibody to TGF-beta (NA-TGFbeta) or SB-431542, a novel TGF-beta receptor kinase inhibitor. Fibroblasts were measured for luciferase activity. SBE-luc fibroblasts underwent Western blot analysis for collagen type I production. RESULTS TGF-beta1 produced maximal luciferase activity in SBE-luc fibroblasts at 0.1 ng/mL (P < 0.05). NA-TGFbeta and SB-431542 inhibited luciferase activity in a dose-dependent fashion, with complete inhibition achieved by 0.1 microg/mL and 1 microM, respectively (P < 0.05). NA-TGFbeta and SB-431542 inhibited collagen type I production. CONCLUSIONS Our in vitro results provide validation for further in vivo real-time imaging studies using the SBE-luc mouse as a novel wound-healing model.
Collapse
Affiliation(s)
- Thomas S Satterwhite
- Section of Plastic Surgery, Department of Veterans Affairs and Division of Plastic Surgery, Stanford University Medical Center, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
107
|
Abstract
Transforming growth factor-β (TGF-β), a prototype of multifunctional cytokine, is a key regulator of extracellular matrix (ECM) assembly and remodeling. Specifically, TGF-β isoforms have the ability to induce the expression of ECM proteins in mesenchymal cells, and to stimulate the production of protease inhibitors that prevent enzymatic breakdown of the ECM. Elevated TGF-β expression in affected organs, and subsequent deregulation of TGF-β functions, correlates with the abnormal connective tissue deposition observed during the onset of fibrotic diseases. During the last few years, tremendous progress has been made in the understanding of the molecular aspects of intracellular signaling downstream of the TGF-β receptors. In particular, Smad proteins, TGF-β receptor kinase substrates that translocate into the cell nucleus to act as transcription factors, have been studied extensively. The role of Smad3 in the transcriptional regulation of typeIcollagen gene expression and in the development of fibrosis, demonstrated both in vitro and in animal models with a targeted deletion of Smad3, is of critical importance because it may lead to novel therapeutic strategies against these diseases. This review focuses on the mechanisms underlying Smad modulation of fibrillar collagen expression and how it relates to fibrotic processes.
Collapse
Affiliation(s)
- Franck Verrecchia
- INSERM U697, Hopital Saint-Louis, Pavillon Bazin, 1 avenue Claude Vellefaux, Paris 75010, France.
| | | |
Collapse
|
108
|
Hsu YC, Wang LF, Chien YW. Nitric oxide in the pathogenesis of diffuse pulmonary fibrosis. Free Radic Biol Med 2007; 42:599-607. [PMID: 17291983 DOI: 10.1016/j.freeradbiomed.2006.11.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 10/26/2006] [Accepted: 11/24/2006] [Indexed: 11/26/2022]
Abstract
By studying the responses of nitric oxide in pulmonary fibrosis, the role of inducible nitric oxide synthase in diffuse pulmonary fibrosis as caused by lipopolysaccharide (LPS) treatment was investigated. When compared to rats treated with LPS only, the rats pretreated with 1400W (an iNOS-specific inhibitor) were found to exhibit a reduced level in: (i) NOx (nitrate/nitrite) production, (ii) collagen type I protein expression, (iv) soluble collagen production, and (iv) the loss of body weight and carotid artery PO2. In the pulmonary fibroblast culture, exogenous NO from LPS-stimulated secretion by macrophages or from a NO donor, such as DETA NONOate, was observed to induce the expression of TIMP-1, HSP47, TGF-beta1, and collagen type I as well as the phosphorylation of SMAD-2. After inhalation of NO for 24 h, an up-regulation of collagen type I protein was also noted to occur in rat pulmonary tissue. The results suggest that the NO signal pathway enhanced the expression of TGF-beta1, TIMP-1, and HSP47 in pulmonary fibroblasts, which collectively demonstrate that the NO signal pathway could activate the SMAD-signal cascade, by initiating a rapid increase in TGF-beta1, thereby increasing the expression of TIMP-1 and HSP47 in pulmonary fibroblasts, and play an important role in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yi-Chiang Hsu
- InnovaTherapeutics Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | |
Collapse
|
109
|
Stoff A, Rivera AA, Mathis JM, Moore ST, Banerjee NS, Everts M, Espinosa-de-los-Monteros A, Novak Z, Vasconez LO, Broker TR, Richter DF, Feldman D, Siegal GP, Stoff-Khalili MA, Curiel DT. Effect of adenoviral mediated overexpression of fibromodulin on human dermal fibroblasts and scar formation in full-thickness incisional wounds. J Mol Med (Berl) 2007; 85:481-96. [PMID: 17219096 DOI: 10.1007/s00109-006-0148-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 06/17/2006] [Accepted: 09/28/2006] [Indexed: 12/19/2022]
Abstract
Fibromodulin, a member of the small leucine-rich proteoglycan family, has been recently suggested as a biologically significant mediator of fetal scarless repair. To assess the role of fibromodulin in the tissue remodeling, we constructed an adenoviral vector expressing human fibromodulin cDNA. We evaluated the effect of adenovirus-mediated overexpression of fibromodulin in vitro on transforming growth factors and metalloproteinases in fibroblasts and in vivo on full-thickness incisional wounds in a rabbit model. In vitro, we found that Ad-Fibromodulin induced a decrease of expression of TGF-beta(1) and TGF-beta(2) precursor proteins, but an increase in expression of TGF-beta(3) precursor protein and TGF-beta type II receptor. In addition, fibromodulin overexpression resulted in decreased MMP-1 and MMP-3 protein secretion but increased MMP-2, TIMP-1, and TIMP-2 secretion, whereas MMP-9 and MMP-13 were not influenced by fibromodulin overexpression. In vivo evaluation by histopathology and tensile strength demonstrated that Ad-Fibromodulin administration could ameliorate wound healing in incisional wounds. In conclusion, although the mechanism of scar formation in adult wounds remains incompletely understood, we found that fibromodulin overexpression improves wound healing in vivo, suggesting that fibromodulin may be a key mediator in reduced scarring.
Collapse
Affiliation(s)
- Alexander Stoff
- Division of Human Gene Therapy, Gene Therapy Center, University of Alabama at Birmingham, 901 19th Street South, BMR2 502, Birmingham, AL, 35294-2172, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Gao Z, Wang Z, Shi Y, Lin Z, Jiang H, Hou T, Wang Q, Yuan X, Zhao Y, Wu H, Jin Y. Modulation of Collagen Synthesis in Keloid Fibroblasts by Silencing Smad2 with siRNA. Plast Reconstr Surg 2006; 118:1328-1337. [PMID: 17051103 DOI: 10.1097/01.prs.0000239537.77870.2c] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Keloids represent a dysregulated response to cutaneous wounding that results in an excessive deposition of extracellular matrix, especially types I and III collagen. Transforming growth factor (TGF)-beta plays a central role in the pathogenesis of fibrosis by inducing and sustaining activation of keloid fibroblast. However, the underlying mechanisms are poorly understood. In this study, the authors examined the function of Smad2, a recently characterized intracellular effector of TGF-beta signaling, in keloid fibroblasts using small interfering RNA (siRNA). METHODS Three pairs of siRNA duplexes targeting human Smad2 were designed; the most efficient one was selected and used for further research. Keloid fibroblasts were treated with or without Smad2 siRNA, and the expression levels of related genes were examined by reverse-transcriptase polymerase chain reaction and immunofluorescence. RESULTS The down-regulation of Smad2 by siRNA led to a significant decrease in mRNA levels of Smad2 in both a dose-dependent and time-dependent manner. The knockdown of Smad2 expression in protein level was confirmed using immunofluorescence. The mRNA levels of types I and III procollagen were also significantly and uniquely decreased following the reduction of Smad2 by siRNA. CONCLUSIONS The results indicate that Smad2 plays an important role in TGF-beta-induced fibrosis in keloids. Down-regulation of Smad2 expression in keloid fibroblasts can significantly decrease procollagen gene expression. Also, siRNA targeting Smad2 was an efficient reagent with which to reduce extracellular matrix deposition and attenuate process of fibrosis. It could be a new, promising therapeutic approach for improving skin wound healing and inhibiting progression of fibrotic conditions by interrupting the TGF-beta signaling pathway.
Collapse
Affiliation(s)
- Zhongyu Gao
- Shanghai, People's Republic of China From the Department of Plastic Surgery, Changzheng Hospital, Second Military Medical University; Department of Orthopedics, Changhai Hospital, Second Military Medical University; and State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Verrecchia F, Mauviel A, Farge D. Transforming growth factor-beta signaling through the Smad proteins: role in systemic sclerosis. Autoimmun Rev 2006; 5:563-9. [PMID: 17027893 DOI: 10.1016/j.autrev.2006.06.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Accepted: 06/08/2006] [Indexed: 12/12/2022]
Abstract
Transforming growth factor-beta (TGF-beta) plays a critical role in the development of tissue fibrosis. Its expression is consistently elevated in affected organs and correlates with increased extracellular matrix deposition. During the last few years, tremendous progress has been made in understanding the molecular aspects of intracellular signaling downstream of the TGF-beta receptors. In particular, Smad proteins, TGF-beta receptor kinase substrates that translocate into the cell nucleus to act as transcription factors, have been studied extensively. Their role in the transcriptional regulation of type I collagen and other extracellular matrix (ECM) genes expression, and in the development of fibrosis is of critical importance because it may lead to novel therapeutic strategies for the treatment of these multi-organ tissue reactions to injury. Systemic sclerosis (SSc) is a complex autoimmune disease characterized by pathological remodelling of connective tissues correlated to the activation of TGF-beta/Smad signaling pathway. This review focuses on the mechanisms underlying Smad modulation of gene expression and how they relate to fibrotic process. Potential implications for the development of therapeutic approaches against tissue fibrosis during SSc are discussed.
Collapse
Affiliation(s)
- Franck Verrecchia
- INSERM U697, Hôpital Saint-Louis, Pavillon Bazin, 1 Avenue Claude Vellefaux, Paris, France.
| | | | | |
Collapse
|
112
|
Moioli EK, Hong L, Guardado J, Clark PA, Mao JJ. Sustained release of TGFbeta3 from PLGA microspheres and its effect on early osteogenic differentiation of human mesenchymal stem cells. ACTA ACUST UNITED AC 2006; 12:537-46. [PMID: 16579687 PMCID: PMC4035024 DOI: 10.1089/ten.2006.12.537] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Despite the widespread role of transforming growth factor-beta3 (TGFbeta3) in wound healing and tissue regeneration, its long-term controlled release has not been demonstrated. Here, we report microencapsulation of TGFbeta3 in poly-d-l-lactic-co-glycolic acid (PLGA) microspheres and determine its bioactivity. The release profiles of PLGA-encapsulated TGFbeta3 with 50:50 and 75:25 PLA:PGA ratios differed throughout the experimental period. To compare sterilization modalities of microspheres, bFGF was encapsulated in 50:50 PLGA microspheres and subjected to ethylene oxide (EO) gas, radio-frequency glow discharge (RFGD), or ultraviolet (UV) light. The release of bFGF was significantly attenuated by UV light, but not significantly altered by either EO or RFGD. To verify its bioactivity, TGFbeta3 (1.35 ng/mL) was control-released to the culture of human mesenchymal stem cells (hMSC) under induced osteogenic differentiation. Alkaline phosphatase staining intensity was markedly reduced 1 week after exposing hMSC-derived osteogenic cells to TGFbeta3. This was confirmed by lower alkaline phosphatase activity (2.25 +/- 0.57 mU/mL/ng DNA) than controls (TGFbeta3- free) at 5.8 +/- 0.9 mU/mL/ng DNA (p < 0.05). Control-released TGFbeta3 bioactivity was further confirmed by lack of significant differences in alkaline phosphatase upon direct addition of 1.35 ng/mL TGFbeta3 to cell culture (p > 0.05). These findings provide baseline data for potential uses of microencapsulated TGFbeta3 in wound healing and tissue-engineering applications.
Collapse
Affiliation(s)
- Eduardo K Moioli
- Tissue Engineering Laboratory, University of Illinois at Chicago, Illinois, USA
| | | | | | | | | |
Collapse
|
113
|
Jagadeesan J, Bayat A. Transforming growth factor beta (TGFbeta) and keloid disease. Int J Surg 2006; 5:278-85. [PMID: 17660136 DOI: 10.1016/j.ijsu.2006.04.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Revised: 04/09/2006] [Accepted: 04/12/2006] [Indexed: 12/11/2022]
Abstract
Keloids are benign fibroproliferative diseases of unknown aetiology. They occur as a result of derangement of the normal wound healing process in susceptible individuals. Although several factors have been postulated in the aetiopathogenesis of this condition, there has been growing evidence to suggest a role for Transforming Growth Factor beta (TGFbeta) family members in its pathogenesis. TGFbeta has also been found to be associated with fibrotic diseases affecting different organs of the body including liver, kidney, lung as well as skin. In this review article, we will discuss the morphology and mechanism of action of TGFbeta and its isoforms and present the most up to date literature discussing the role of TGFbeta isoforms, their receptors, and intracellular signalling pathways (the SMAD pathway) in the pathogenesis of keloid disease. Understanding the role of TGFbeta in keloid disease could lead to the development of clinically useful therapeutic modalities for treatment of this condition.
Collapse
Affiliation(s)
- Jagajeevan Jagadeesan
- Department of Plastic and Reconstructive Surgery, Royal Preston Hospital, Sharoe Green Lane, Fulwood, Preston PR2 9HT, UK
| | | |
Collapse
|
114
|
Hayashi T, Nishihira J, Koyama Y, Sasaki S, Yamamoto Y. Decreased prostaglandin E2 production by inflammatory cytokine and lower expression of EP2 receptor result in increased collagen synthesis in keloid fibroblasts. J Invest Dermatol 2006; 126:990-7. [PMID: 16528361 DOI: 10.1038/sj.jid.5700227] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We investigated the metabolism of arachidonic acid in normal skin-derived fibroblasts (NF) as well as in keloid-derived fibroblasts (KF) in response to macrophage migration inhibitory factor (MIF), a pluripotent cytokine. We found that MIF enhanced cyclooxygenase-2 activity in NF more than in KF. Consistent with this finding, prostaglandin E(2) (PGE(2)), an antifibrogenic molecule, was more significantly increased in NF than in KF by MIF treatment. As regarding E prostanoid receptor 2, the level of expression was significantly lower in KF than in NF. On the other hand, Forskolin, a direct activator of adenylcyclase, decreased collagen synthesis in both NF and KF, which indicates that cAMP plays an important role in regulating collagen synthesis. As PGE(2) induces cAMP production, it is conceivable that increased collagen synthesis in KF might be owing to decreased PGE(2) and cAMP production. These findings may aid in the development of a therapeutic strategy for the regulation of collagen synthesis in keloid fibroblasts.
Collapse
Affiliation(s)
- Toshihiko Hayashi
- Department of Plastic and Reconstructive Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan.
| | | | | | | | | |
Collapse
|
115
|
Xia W, Longaker MT, Yang GP. P38 MAP kinase mediates transforming growth factor-beta2 transcription in human keloid fibroblasts. Am J Physiol Regul Integr Comp Physiol 2006; 290:R501-8. [PMID: 16467496 DOI: 10.1152/ajpregu.00472.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Keloids are abnormal fibrous growths of the dermis that develop only in response to wounding and represent a form of benign skin tumor. Previous studies have shown increased protein levels of TGF-beta in keloid tissue, suggesting a strong association with keloid formation leading us to examine mechanisms for why it is more highly expressed in keloids. Here, we use serum stimulation as an in vitro model to mimic a component of the wound microenvironment and examine differential gene expression in keloid human fibroblasts (KFs) vs. normal human fibroblasts (NFs). Transcription of TGF-beta2 was rapid and peaked between 1 and 6 h after serum stimulation in KFs vs. NFs. We confirmed increased TGF-beta activity in the conditioned medium from KFs, but not NFs. Inhibition of second messenger signaling pathways demonstrated that only the p38 MAPK inhibitor SB-203580 could block upregulation of TGF-beta2 following serum stimulation in KFs. Immunoblotting demonstrated that p38 MAPK was phosphorylated within 15 min and was maintained at a high level in KFs but not in NFs. The transcription factors activating transcription factor-2 and Elk-1 are activated by p38 MAPK, and also showed rapid and prolonged phosphorylation kinetics in KFs but not in NFs. In conclusion, increased TGF-beta2 transcription in response to serum stimulation in KFs appears to be mediated by the p38 MAPK pathway. This suggests the mechanism of keloid pathogenesis may be due in part to an inherent difference in how the fibroblasts respond to wounding.
Collapse
Affiliation(s)
- Wei Xia
- Department of Surgery, Stanford University Medical School, California, USA
| | | | | |
Collapse
|
116
|
Xia W, Phan TT, Lim IJ, Longaker MT, Yang GP. Differential transcriptional responses of keloid and normal keratinocytes to serum stimulation. J Surg Res 2006; 135:156-63. [PMID: 16564547 DOI: 10.1016/j.jss.2006.01.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2005] [Revised: 01/25/2006] [Accepted: 01/31/2006] [Indexed: 11/18/2022]
Abstract
BACKGROUND Keloids are benign tumors that occur only in response to injury, for which there is no effective treatment. We demonstrated previously that keloid keratinocytes (KKs) promote fibroblast proliferation more than normal keratinocytes (NKs) and that transforming growth factor (TGF)-beta is a component of that signal. We used the transcriptional response to serum stimulation to examine how TGF-beta expression is stimulated in KKs. MATERIALS AND METHODS Quiescent KKs and NKs were stimulated using serum; harvested using RNA at 0, 1, 6, 12, and 24 h; and analyzed using quantitative real-time polymerase chain reaction. TGF-beta activity in the conditioned medium was measured with an MLEC/PAI-luciferase assay. Inhibition of ERK1/2, p38 kinase, and JNK pathways was performed with PD98059, SB203580, and SP600125, respectively. RESULTS Increased transcription of TGF-beta2 occurs within 1 h of serum stimulation in KKs but not in NKs. In contrast, TGF-beta3 transcription was suppressed in KKs compared with NKs. No significant differences were observed in the transcriptional response of TGF-beta1. Increased TGF-beta2 mRNA correlated with increased TGF-beta biological activity in the conditioned medium. Inhibition of the ERK, p38 kinase or JNK signal transduction pathways blocked the transcriptional up-regulation of TGF-beta2, TbetaR1, and TbetaR2 in KKs. CONCLUSIONS KKs produce more TGF-beta2 mRNA than NKs in response to serum stimulation, resulting in increased TGF-beta activity in conditioned medium. Combining these results with our previous data lead us to propose a model of keloid formation characterized by an exaggerated response to cellular stress and abnormal epithelial-mesenchymal signaling promoting keloid formation.
Collapse
Affiliation(s)
- Wei Xia
- Department of Surgery, Stanford University Medical School, Stanford, California, USA
| | | | | | | | | |
Collapse
|
117
|
Abstract
For centuries, keloids have been an enigma and despite considerable research to unravel this phenomenon no universally accepted treatment protocol currently exists. Historically, the etiology of keloids has been hypothesized by multiple different theories; however, a more contemporary view postulates a multifactoral basis for this disorder involving nutritional, biochemical, immunological, and genetic factors that play a role in this abnormal wound healing. Critical to the process of preventing or managing keloids is the need to locally control fibroblasts and their activities at the wound site. In recent years, considerable evidence has accumulated demonstrating the importance of fatty acids and bioactive lipids in health and disease, especially those involving inflammatory disorders or immune dysfunction. If hypertrophic scarring and keloid formation can be argued to have significant inflammatory histories, then it is possible to postulate a role for lipids in their etiology and potentially in their treatment. This report briefly visits past views and theories on keloid formation and treatment, and offers a theoretical rationale for considering adjuvant fatty acid therapy for keloid management. Sufficient scientific evidence in support of fatty acid strategies for the prevention and treatment of keloids currently exists, which offer opportunities to bridge the gap between the laboratory and the clinic. The intent of this paper is to serve as a basic guideline for researchers, nutritionists, and clinicians interested in keloids and to propose new directions for keloid management.
Collapse
Affiliation(s)
- Louise Louw
- Department of Otorhinolaryngology, Division of Research, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa.
| |
Collapse
|
118
|
Abstract
LEARNING OBJECTIVES After studying this article, the participant should be able to: 1. Have a greater appreciation of the extent of differences and similarities between keloid and hypertrophic scarring. 2. Have a greater appreciation of the significance of the stage of maturation of a keloid or hypertrophic scar with regard to its morphologic, biochemical, and molecular profile. 3. More critically review basic science research that is based on poorly characterized scar tissue. 4. More critically review clinical studies that are based on poorly characterized scar tissue. BACKGROUND Hypertrophic and keloid scars remain extremely challenging, particularly in their variable response to treatment. The understanding of hypertrophic and keloid scarring is evolving from a position where they were regarded as different stages of the same process to the contemporary perspective of two separate entities. This article reviews the differences in the two forms of scarring and discusses the implications for future research. METHODS The authors conducted a MEDLINE search of all English language reviews linking key words "hypertrophic," "keloid," and "scarring." RESULTS Over the past four decades, there has been considerable clinical and experimental research looking at the biological nature and therapeutic response of keloid and hypertrophic scarring. As more differences are emerging regarding the fundamental biology of the scars, investigators are giving more detailed characterization of their source material. It is evident that even within the broad categories of hypertrophic and keloid scarring there is a heterogenous distribution of pathologic connective tissue matrix biology. CONCLUSION Considerable advances have been made in our understanding of the fundamental biology of scarring. As research methodology becomes even more sophisticated, it will be even more crucial to extensively characterize source material, recognizing major differences not only between keloid and hypertrophic scar but also between scars of varying stages of maturation and histomorphological, biochemical, and molecular variations within individual scars.
Collapse
Affiliation(s)
- Andrew Burd
- Division of Plastic and Reconstructive Surgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.
| | | |
Collapse
|
119
|
Phan TT, Lim IJ, Aalami O, Lorget F, Khoo A, Tan EK, Mukhopadhyay A, Longaker MT. Smad3 signalling plays an important role in keloid pathogenesis via epithelial-mesenchymal interactions. J Pathol 2005; 207:232-42. [PMID: 16052471 DOI: 10.1002/path.1826] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Smad signalling plays important roles in developmental and cancer biology as well as in fibropathogenesis. Its role in keloid biology is not known. Epithelial-mesenchymal interactions, originally described in normal skin, have recently been established to play a significant role in keloid pathogenesis, and demonstrate the important influence of keratinocyte paracrine factor signalling on fibroblast behaviour. The present study investigated the role of downstream Smad cascade induction in this interaction. Normal fibroblasts (NF) and keloid fibroblasts (KF) were co-cultured in serum-free medium with normal keratinocytes (NK) or keloid keratinocytes (KK) for 5 days, after which fibroblast cell lysates were subjected to western blot and immunoprecipitation analysis to quantify the levels of Smad and Smad2/3/4 binding complex. In another set of experiments, wild-type (wt), Smad2-null (Smad2-/-) and Smad3-null (Smad3-/-) mouse embryonic fibroblasts (MEF) were assayed for cell proliferation and collagen production after serum-free co-culture with KK or exposure to conditioned media collected from serum-free KK/KF co-culture. Compared to normal skin, keloids expressed high basal levels of TGFbetaR1 and TGFbetaR2, Smad2, 3 and 4 and phospho-Smad2. Upregulation of TGFbetaR1 and TGFbetaR2, Smad3 and p-Smad2 was observed in KF co-cultured with KK, together with enhanced Smad3 phosphorylation and Smad2/3/4 binding complex production. When MEF-wt, MEF-Smad2-/- or MEF-Smad3-/- were co-cultured with KK or exposed to KK/KF co-culture conditioned media, enhanced proliferation and collagen production were seen in MEF-wt and MEF-Smad2-/- but not in MEF-Smad3-/- cells. The activation of Smad signalling, importantly that of Smad3, appears to be one facet of the complex epithelial-mesenchymal interactions in keloid pathogenesis, resulting in active KF proliferation and collagen-ECM production in co-culture with KK. This finding suggests the suppression of Smad signalling as a novel approach in keloid therapy.
Collapse
Affiliation(s)
- T T Phan
- Department of Surgery, National University of Singapore, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
120
|
García-Ulloa AC, Arrieta O. Tubal occlusion causing infertility due to an excessive inflammatory response in patients with predisposition for keloid formation. Med Hypotheses 2005; 65:908-14. [PMID: 16005574 DOI: 10.1016/j.mehy.2005.03.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Accepted: 03/23/2005] [Indexed: 12/20/2022]
Abstract
Infertility is a condition that affects approximately 15-25% of couples with the desire to procreate. The integrity of the feminine reproductive tract is essential for this purpose, but the occlusion of the Fallopian tubes occurs in 12-33% of infertile women. The infection by Chlamydia trachomatis is one of the principle causes of tubal injury, which could finally lead to tubal occlusion. The tract infection has also been related to the use of intrauterine device, basically due to the fact that the insertion of the device could carry bacteria to the endometrial cavity. Keloid scars result from alterations in the normal process of wound healing, and it affects principally the population in reproductive age, maybe due to specific hormonal influence. These fibroproliferative alterations may produce significant deformations and alter organ function. The genetic factors have been studied in order to have a better understanding of the pathophysiology of keloid scarring. With these assessments, many other factors have been known to have a relationship with this abnormal healing process. This keloid scarring involves an excess in extracellular matrix production and inhibition of apoptosis, for which a several growth factors and interleukins are needed. One of the most important growth factors is IGF-1, which increases the expression of type I and III procollagen (found in the uterus); the IGF-1 receptor is overexpressed in the fibroblasts of keloids. Based on those previous observations a hypothesis that the chronic and repeated infection, and the use of IUD, generate an exaggerated inflammatory response in patients with a predisposition for keloid formation (which frequently form in childbearing age), in comparison to the patients that do not form this type of scarring, has been proposed. This makes a major frequency of adherences and finally tubal occlusion and infertility. The tendency of excessive scarring could not be exclusive of skin and generate abnormal scarring responses in feminine reproductive tract, leading to a major frequency of infertility. Thus, it could be suggested the use of other contraceptive methods and a more aggressive treatment against infections of the reproductive tract, taking in consideration the pathophysiology of keloid scar formation and its relationship with tubal occlusion.
Collapse
Affiliation(s)
- Ana Cristina García-Ulloa
- Department of Medical Oncology, Instituto National de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, 14080 Tlalpan, Mexico City, Mexico
| | | |
Collapse
|
121
|
Bock O, Yu H, Zitron S, Bayat A, Ferguson MWJ, Mrowietz U. Studies of transforming growth factors beta 1-3 and their receptors I and II in fibroblast of keloids and hypertrophic scars. Acta Derm Venereol 2005; 85:216-20. [PMID: 16040405 DOI: 10.1080/00015550410025453] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Keloids are benign skin tumours occurring during wound healing in genetically predisposed patients. They are characterized by an abnormal deposition of extracellular matrix components, particularly collagen. There is uncertain evidence that transforming growth factor-beta (TGFss) is involved in keloid formation. Therefore we investigated the expression of TGFss1, 2 and 3 and their receptors in keloids, hypertrophic scars and normal skin. Dermal fibroblasts were obtained from punch biopsies of patients with keloids and hypertrophic scars and from normal skin of healthy individuals. Total RNA was isolated and the expression of TGFss1, 2 and 3 and of TGFss receptors I and II (TGFssRI and II) was analysed by real-time PCR using the Lightcycler technique. Our data demonstrate significantly lower TGFss2 mRNA expression in hypertrophic scar fibroblasts as compared with fibroblasts derived from keloids and normal skin (p<0.05). In contrast, TGFss3 mRNA expression was significantly lower in keloid fibroblasts in comparison with fibroblasts derived from hypertrophic scar and normal skin (p<0.01). TGFssRI mRNA expression was significantly decreased in hypertrophic scar fibroblasts (p<0.01) and TGFssRII mRNA expression was decreased in keloids compared with hypertrophic scar fibroblasts (p<0.001). The ratio of TGFssRI/TGFssRII expression was increased in keloids compared with hypertrophic scar and normal skin fibroblasts. As recently supposed, an increased TGFssRI/TGFssRII ratio could promote fibrosis. Therefore our data support a possible role of TGFssRI and TGFssRII in combination with a certain TGFss expression pattern as fibrosis-inducing factors in keloids.
Collapse
Affiliation(s)
- Oliver Bock
- Primary Health Care Center, Fabriksgatan 17, SE-552 85 Jönköping, Sweden.
| | | | | | | | | | | |
Collapse
|
122
|
Luzina IG, Tsymbalyuk N, Choi J, Hasday JD, Atamas SP. CCL18-stimulated upregulation of collagen production in lung fibroblasts requires Sp1 signaling and basal Smad3 activity. J Cell Physiol 2005; 206:221-8. [PMID: 16021625 DOI: 10.1002/jcp.20452] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A CC chemokine CCL18 stimulates collagen production in pulmonary fibroblasts through an unknown signaling mechanism. In this study, involvement of Sp1 and Smad3 in CCL18 signaling in primary human pulmonary fibroblast cultures was investigated. Phosphorylation of Sp1, DNA-binding by Sp1, and the activity of an Sp1-dependent reporter were all increased in response to CCL18 stimulation. CCL18 did not stimulate a detectable increase in Smad3 phosphorylation or Smad3/4 DNA-binding activity, although some basal phosphorylation and DNA binding by Smad3/4 were noted. Transient overexpression of dominant negative mutants of Sp1 and Smad3 abrogated CCL18-dependent upregulation as well as basal production of collagen. These observations suggested that CCL18 activates collagen production in pulmonary fibroblasts through an Sp1-dependent pathway that also requires basal Smad3 activity. Possible involvement of autocrine TGF-beta in CCL18 signaling was considered. CCL18 stimulated increases in collagen mRNA and protein production without detectable changes in TGF-beta1, -beta2, and -beta3 mRNA or protein levels. Neutralizing anti-TGF-beta antibodies, latency-associated peptide, ALK5-specific inhibitor SD431542, and an inhibitor of the protease-dependent TGF-beta activation aprotinin, each failed to block CCL18-stimulated collagen production. These observations suggest that both CCL18 signaling in pulmonary fibroblasts and basal Smad3 activity are independent of autocrine TGF-beta.
Collapse
Affiliation(s)
- Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore VA Medical Center, Baltimore, Maryland 21201, USA.
| | | | | | | | | |
Collapse
|
123
|
Affiliation(s)
- Abelardo Medina
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, 8440-112 Street, Edmonton, Alberta, Canada T6G 2B7
| | | | | | | |
Collapse
|
124
|
Abstract
Cutaneous wounds inevitably heal with scars, which can be disfiguring and compromise function. In general, the greater the insult, the worse the scarring, although genetic make up, regional variations and age can influence the final result. Excessive scarring manifests as hypertrophic and keloid scars. At the other end of the spectrum are poorly healing chronic wounds, such as foot ulcers in diabetic patients and pressure sores. Current therapies to minimize scarring and accelerate wound healing rely on the optimization of systemic conditions, early wound coverage and closure of lacerations, and surgical incisions with minimal trauma to the surrounding skin. The possible benefits of topical therapies have also been assessed. Further major improvements in wound healing and scarring require an understanding of the molecular basis of this process. Promising strategies for modulating healing include the local administration of platelet derived growth factor (PDGF)-BB to accelerate the healing of chronic ulcers, and increasing the relative ratio of transforming growth factor (TGF)beta-3 to TGFbeta-1 and TGFbeta-2 in order to minimize scarring.
Collapse
|
125
|
Enhanced Dupuytren's disease fibroblast populated collagen lattice contraction is independent of endogenous active TGF-beta2. BMC Musculoskelet Disord 2004; 5:41. [PMID: 15541177 PMCID: PMC534110 DOI: 10.1186/1471-2474-5-41] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Accepted: 11/12/2004] [Indexed: 11/17/2022] Open
Abstract
Background Dupuytren's disease (DD) is a debilitating fibro-proliferative disorder of the hand characterized by the appearance of fibrotic lesions (nodules and cords) leading to flexion contractures of the fingers and loss of hand function. Although the molecular mechanism of DD is unknown, it has been suggested that transforming growth factor-β2 (TGF-β2) may play an important role in the underlying patho-physiology of the disease. The purpose of this study was to further explore this hypothesis by examining the effects of TGF-β2 on primary cell cultures derived from patient-matched disease and normal palmar fascia tissue using a three-dimensional collagen contraction assay. Methods Fibroblast-populated collagen lattice (FPCL) contraction assays using primary cell cultures derived from diseased and control fascia of the same DD patients were studied in response to exogenous TGF-β2 and neutralizing anti-TGF-β2 antibodies. Results Contraction of the FPCLs occurred significantly faster and to a greater extent in disease cells compared to control cells. The addition of TGF-β2 enhanced the rate and degree of collagen contraction in a dose-dependent fashion for both control and diseased cells. Neutralizing anti-TGF-β2 antibodies abolished exogenous TGF-β2 stimulated collagen contraction, but did not inhibit the enhanced basal collagen contraction activity of disease FPCL cultures. Conclusions Although exogenous TGF-β2 stimulated both disease and control FPCL contraction, neutralizing anti-TGF-β2 antibodies did not affect the elevated basal collagen contraction activity of disease FPCLs, suggesting that the differences in the collagen contraction activity of control and disease FPCL cultures are not due to differences in the levels of endogenous TGF-β2 activity.
Collapse
|
126
|
Phan TT, Lim IJ, Chan SY, Tan EK, Lee ST, Longaker MT. Suppression of Transforming Growth Factor Beta/Smad Signaling in Keloid-Derived Fibroblasts by Quercetin: Implications for the Treatment of Excessive Scars. ACTA ACUST UNITED AC 2004; 57:1032-7. [PMID: 15580028 DOI: 10.1097/01.ta.0000114087.46566.eb] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Keloids are characterized by abnormal proliferation and overproduction of extracellular matrix. Quercetin, a dietary compound, has strong antioxidant and anticancer properties. Previous studies by the authors have shown that quercetin inhibits fibroblast proliferation, collagen production, and contraction of keloid and hypertrophic scar-derived fibroblasts. Quercetin also blocks the signal transduction of insulin-like growth factor-1 in keloid fibroblasts. This study assessed the effects of quercetin on the transforming growth factor (TGF)-beta/Smad-signaling pathway in keloid-derived fibroblasts, which may be an important biologic mechanism of this proliferative scarring. METHODS Keloid fibroblasts were isolated from keloid tissue specimens. Cells were treated with quercetin at different concentrations, then harvested, and subjected to immunoblotting analysis. RESULTS Quercetin significantly inhibited the expression of TGF-beta receptors 1 and 2 in keloid fibroblasts at three concentrations (low, medium, and high). Quercetin also strongly suppressed the basal expression of Smad2, Smad3, and Smad4 as well as the phosphorylation of Smad2 and Smad3 and the formation of the Smad2-Smad3-Smad4 complex. CONCLUSIONS Taken together, these data suggest that quercetin effectively blocks the TGF-beta/Smad-signaling pathway in keloid fibroblasts. These data indicate that quercetin-based therapies for keloids should be investigated further.
Collapse
|
127
|
Xia W, Phan TT, Lim IJ, Longaker MT, Yang GP. Complex epithelial-mesenchymal interactions modulate transforming growth factor-beta expression in keloid-derived cells. Wound Repair Regen 2004; 12:546-56. [PMID: 15453837 DOI: 10.1111/j.1067-1927.2004.012507.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Keloids are proliferative dermal growths representing a pathologic wound healing response. We have previously demonstrated that coculture of fibroblasts derived from either keloid or normal skin have an elevated proliferation rate when cocultured with keloid-derived keratinocytes vs. normal keratinocytes. In these studies, we examined the contribution of transforming growth factor-beta (TGF-beta) to this phenomenon using a two-chamber coculture system. Fibroblast proliferation in coculture was slower with the addition of a pan-TGF-beta neutralizing antibody. Keloid keratinocytes in coculture expressed more TGF-beta1, -beta3, and TGF-beta receptor 1 than normal keratinocytes. Keloid fibroblasts cocultured with keloid keratinocytes expressed more mRNA for TGF-beta1, -beta2, TGF-beta receptor 1, and Smad2. Keloid fibroblasts also produced more type I collagen, connective tissue growth factor, and insulin-like growth factor-II/mannose-6-phosphate receptor when cocultured with keloid keratinocytes vs. normal keratinocytes. Levels of total and activated TGF-beta activity increased when fibroblasts were cocultured with keratinocytes, correlating with the changes in transcriptional activity of TGF-beta. In conclusion, we find a complex paracrine interaction regulates TGF-beta mRNA expression and activation between keratinocytes and fibroblasts. These data suggest that keloid pathogenesis may result from both an increased TGF-beta production and activation by the keloid keratinocyte, and elevated TGF-beta expression, utilization, and signaling in keloid fibroblasts.
Collapse
Affiliation(s)
- Wei Xia
- Department of Surgery, Stanford University Medical School, 257 Campus Drive, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
128
|
Gira AK, Brown LF, Washington CV, Cohen C, Arbiser JL. Keloids demonstrate high-level epidermal expression of vascular endothelial growth factor. J Am Acad Dermatol 2004; 50:850-3. [PMID: 15153883 DOI: 10.1016/j.jaad.2003.11.061] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Keloids are a major cause of morbidity, and arise after operation, injury, or cutaneous infection. Clinically, keloids differ from hypertrophic scars in that they grow beyond the original borders of the injury. Keloids occur most commonly for patients of African and Asian descent, and treatment options are multiple, indicating that there is no entirely satisfactory treatment for keloids. Angiogenesis inhibition has been shown to be effective in treatment of malignancy in both animal models and human beings. OBJECTIVE We sought to determine whether keloids produce the potent angiogenic factor vascular endothelial growth factor (VEGF). METHODS We performed in situ hybridization for VEGF on keloid tissue and normal skin. RESULTS Our study demonstrated abundant production of VEGF in keloids and, surprisingly, the major source of VEGF was the overlying epidermis. CONCLUSIONS Our results suggest that the overlying epidermis is the major source of keloid angiogenesis. These findings demonstrate that keloids are angiogenic lesions. Topical antiangiogenic therapy, directed at either down-regulating epidermal VEGF or inhibiting keratinocyte-derived VEGF activity on its endothelial receptors, may be useful in the treatment of keloids.
Collapse
Affiliation(s)
- Amy Kim Gira
- Department of Dermatology, Emory University School of Medicine and Atlanta Veterans Administration Medical Center, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
129
|
Abstract
The cause of fibrotic diseases, pathologies characterized by excessive production, deposition, and contraction of extracellular matrix, is unknown. To understand the molecular basis of fibrotic disease, it is essential to appreciate how matrix deposition is normally controlled and how this process is dysregulated in fibrogenesis. This review discusses the current state of knowledge concerning interactions among the profibrotic proteins transforming growth factor-beta (TGF-beta), connective tissue growth factor (CTGF, CCN2), and ED-A fibronectin (ED-A FN) and the antifibrotic proteins tumor necrosis factor-alpha (TNF-alpha) and gamma-interferon (IFN-gamma).
Collapse
Affiliation(s)
- Andrew Leask
- Centre for Rheumatology, Department of Medicine, Royal Free & University College Medical School, Rowland Hill St., London, UK NW3 2PF.
| | | |
Collapse
|
130
|
Bayat A, Bock O, Mrowietz U, Ollier WER, Ferguson MWJ. Genetic susceptibility to keloid disease: transforming growth factor beta receptor gene polymorphisms are not associated with keloid disease. Exp Dermatol 2004; 13:120-4. [PMID: 15009106 DOI: 10.1111/j.0906-6705.2004.00165.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Keloid disease (KD) is an abnormal form of scarring with a familial predisposition. Genetic studies have yet to identify the genes involved in KD. Transforming growth factor beta (TGF-beta) has multiple cellular activities including cellular proliferation, differentiation and extracellular matrix production. TGF-beta family members such as TGF-beta(1) and TGF-beta(2) are known to be involved in KD formation. However, we previously demonstrated a lack of association between common TGF-beta(1) and TGF-beta(2) polymorphisms and KD. Other studies have implicated TGF-beta receptors in KD pathogenesis. TGF-beta receptors were therefore selected as candidate-susceptibility genes for this condition. Single-nucleotide polymorphisms (SNPs) in TGF-beta receptors I, II and III (TGF-betaRI, TGF-betaRII and TGF-betaRIII) were identified and investigated for association with the risk of developing KD. A polymerase chain reaction-restriction fragment length polymorphism method was used for genotyping novel and known TGF-beta receptor polymorphisms. DNA samples from 92 KD cases and 181 controls were examined. There were no statistically significant differences in genotype or allele frequency distributions between cases and controls for the TGF-beta receptor SNPs. Therefore, these TGF-beta receptor polymorphisms are unlikely to be associated with keloid scarring. It is possible that other SNPs in other TGF-beta family members are associated with KD. To our knowledge, this is the first report of a case-control association study with KD and TGF-beta receptor gene polymorphisms.
Collapse
Affiliation(s)
- Ardeshir Bayat
- The Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, UK.
| | | | | | | | | |
Collapse
|
131
|
Abstract
Transforming growth factor-beta (TGF-beta) plays a central role in fibrosis, contributing to the influx and activation of inflammatory cells, the epithelial to mesenchymal transdifferentiation (EMT) of cells and the influx of fibroblasts and their subsequent elaboration of extracellular matrix. TGF-beta signals through transmembrane receptor serine/threonine kinases to activate novel signalling intermediates called Smad proteins, which modulate the transcription of target genes. The use of mice with a targeted deletion of Smad3, one of the two homologous proteins which signals from TGF-beta/activin, shows that most of the pro-fibrotic activities of TGF-beta are mediated by Smad3. Smad3 null inflammatory cells and fibroblasts do not respond to the chemotactic effects of TGF-beta and do not autoinduce TGF-beta. The loss of Smad3 also interferes with TGF-beta-mediated induction of EMT and genes for collagens, plasminogen activator inhibitor-1 and the tissue inhibitor of metalloprotease-1. Smad3 null mice are resistant to radiation-induced cutaneous fibrosis, bleomycin-induced pulmonary fibrosis, carbon tetrachloride-induced hepatic fibrosis as well as glomerular fibrosis induced by induction of type 1 diabetes with streptozotocin. In fibrotic conditions that are induced by EMT, such as proliferative vitreoretinopathy, ocular capsule injury and glomerulosclerosis resulting from unilateral ureteral obstruction, Smad3 null mice also show an abrogated fibrotic response. Animal models of scleroderma, cystic fibrosis and cirrhosis implicate involvement of Smad3 in the observed fibrosis. Additionally, inhibition of Smad3 by overexpression of the inhibitory Smad7 protein or by treatment with the small molecule, halofuginone, dramatically reduces responses in animal models of kidney, lung, liver and radiation-induced fibrosis. Small moleucule inhibitors of Smad3 may have tremendous clinical potential in the treatment of pathological fibrotic diseases.
Collapse
Affiliation(s)
- Kathleen C Flanders
- Laboratory of Cell Regulation and Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD 20892-5055, USA.
| |
Collapse
|
132
|
Chin D, Boyle GM, Parsons PG, Coman WB. What is transforming growth factor-beta (TGF-β)? ACTA ACUST UNITED AC 2004; 57:215-21. [PMID: 15006522 DOI: 10.1016/j.bjps.2003.12.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2002] [Accepted: 12/10/2003] [Indexed: 12/15/2022]
Abstract
The TGF-beta superfamily of proteins produces a wide range of frequently opposing effects in different cells and tissues in the body. However, its activation and mode of action are only partially understood because of its complexity in structure and functions and the variability in its downstream targets. Current work on these cytokines focuses on their receptors and the intercellular signalling pathways, comparing bioactivities between cell types and tracking their physiological and immunological effects in vivo. Future research will yield important therapeutic applications and the ability to manipulate these proteins in vivo.
Collapse
Affiliation(s)
- David Chin
- Melanoma Genomics and Head and Neck Group, Queensland Institute of Medical Research, Herston, QLD, Australia. davidCqimr.edu.au
| | | | | | | |
Collapse
|
133
|
Jinnin M, Ihn H, Asano Y, Yamane K, Trojanowska M, Tamaki K. Tenascin-C upregulation by transforming growth factor-beta in human dermal fibroblasts involves Smad3, Sp1, and Ets1. Oncogene 2004; 23:1656-67. [PMID: 15001984 DOI: 10.1038/sj.onc.1207064] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In cultured human dermal fibroblasts, transforming growth factor (TGF)-beta induced the mRNA expression of tenascin-C (TN-C). The molecular mechanism(s) underlying this process is not presently understood. In this study, we performed serial 5' deletion and a transient transfection analysis to define a region in the TN-C promoter mediating the inducible responsiveness to TGF-beta. This region contains an atypical nucleotide recognition element for the Smad family of transcriptional regulators. A DNA affinity precipitation assay revealed that Smad2/Smad3 bound to this site in a transient and specific manner. Overexpression of Smad3 or Smad4 activated the TN-C promoter activity and superinduced the TN-C promoter activity stimulated by TGF-beta. Moreover, simultaneous cotransfection of Smad3 and Smad4 activated the TN-C promoter activity in a synergistic manner. Mutation of the Smad-binding sites, the Ets-binding sites, or Sp1/3-binding sites in the TN-C promoter abrogated the TGF-beta/Smad-inducible promoter activity. Immunoprecipitation analysis revealed that Smad3, Sp1, and Ets1 form a transcriptionally active complex. Furthermore, the interaction between Smads and CBP/p300 in TGF-beta signaling was confirmed. These findings demonstrate the existence of a novel, functional binding element in the proximal region of the TN-C promoter mediating responsiveness to TGF-beta involving Smad3/4, Sp1, Ets1, and CBP/p300.
Collapse
Affiliation(s)
- Masatoshi Jinnin
- Department of Dermatology, Faculty of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | |
Collapse
|
134
|
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; 121:1326-31. [PMID: 14675177 DOI: 10.1111/j.1523-1747.2003.12572.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interactions between epidermal keratinocytes and dermal fibroblasts play an important role in regulating tissue homeostasis and repair. Nevertheless, little is known about the role of keratinocytes in the pathogenesis of keloid. In this study, we investigated the influence of normal skin- and keloid-derived keratinocytes on normal skin- and keloid-derived fibroblasts utilizing a serum-free indirect coculture system. The keloid-derived fibroblasts showed a greater proliferation and minimal apoptosis when cocultured with normal skin- or keloid-derived keratinocytes, and the results were most significant in the latter. This difference was not observed when the fibroblasts were treated with conditioned medium obtained from normal skin- and keloid-derived keratinocytes. Nevertheless, conditioned medium-treated groups showed more proliferation and less apoptosis compared to the nonconditioned medium-treated control groups. We also analyzed the profile of factors involved in cell growth and apoptosis in fibroblasts cocultured with keratinocytes. Extracellular signal-regulated kinase and c-Jun N-terminal kinase phosphorylations and expression of Bcl-2 and transforming growth factor-beta1 were all significantly upregulated in the fibroblasts cocultured with keloid-derived keratinocytes. Together, these results strongly suggest that the overlying keratinocytes of the keloid lesion play an important role in keloidogenesis by promoting more proliferation and less apoptosis in the underlying fibroblasts through paracrine and double paracrine effects.
Collapse
Affiliation(s)
- Emi Funayama
- Department of Plastic and Reconstructive Surgery, Hokkaido University School of Medicine, Sapporo, Japan
| | | | | | | |
Collapse
|
135
|
Li WY, Chong SSN, Huang EY, Tuan TL. Plasminogen activator/plasmin system: a major player in wound healing? Wound Repair Regen 2003; 11:239-47. [PMID: 12846910 DOI: 10.1046/j.1524-475x.2003.11402.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The role of the plasminogen activator/plasmin system in fibrinolysis has been well established. Indeed, clinicians worldwide have successfully utilized recombinant tissue-type plasminogen activator as first-line treatment of acute myocardial infarction for almost 2 decades. Outside the field of cardiology, there has been increasing excitement regarding the possible contribution of this system in many other important biological processes, including cell adhesion, cell migration, cell-cell signaling, tumor invasion and metastasis, ovulation, and wound healing. In this review, we present evidence in the current literature that the plasminogen activator/plasmin system does have a role in wound healing, looking at both normal and abnormal healing. Furthermore, the invaluable insights provided by numerous transgenic animal experiments are summarized.
Collapse
Affiliation(s)
- Wai-Yee Li
- Department of Surgery, Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California 90027, USA
| | | | | | | |
Collapse
|
136
|
Mori Y, Chen SJ, Varga J. Expression and regulation of intracellular SMAD signaling in scleroderma skin fibroblasts. ARTHRITIS AND RHEUMATISM 2003; 48:1964-78. [PMID: 12847691 DOI: 10.1002/art.11157] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Scleroderma is characterized by excessive synthesis and accumulation of matrix proteins in lesional tissues. Transforming growth factor beta (TGFbeta) plays a central role in the pathogenesis of fibrosis by inducing and sustaining activation of fibroblasts; however, the underlying mechanisms are poorly understood. We undertook this study to examine the expression and function of SMADs, recently characterized intracellular effectors of TGFbeta signaling, in scleroderma fibroblasts. METHODS Primary dermal fibroblasts obtained from 14 patients with scleroderma and from 4 healthy adult volunteers were studied. Northern analysis was used to determine the expression of endogenous SMAD messenger RNA (mRNA), and Western analysis was used to determine SMAD protein expression. Intracellular compartmentalization of cellular SMAD proteins in the presence and absence of TGFbeta was studied by antibody-mediated immunofluorescence confocal microscopy. The effect of TGFbeta blockade on SMAD subcellular distribution was determined using anti-TGFbeta antibodies as well as a dominant-negative TGFbeta receptor type II (TGFbetaRII) vector to disrupt TGFbeta responses. SMAD-regulated luciferase reporter expression was examined to investigate the potential functional significance of activation and nuclear accumulation of endogenous SMADs in scleroderma fibroblasts. RESULTS Protein and mRNA levels of SMAD3, but not of SMAD4 or SMAD7, were variably elevated in scleroderma fibroblasts compared with those from healthy controls. In sharp contrast to control fibroblasts, which displayed predominantly cytoplasmic localization of SMAD3/4 in the absence of exogenous TGFbeta, in scleroderma fibroblasts SMAD3 and SMAD4 consistently showed elevated nuclear localization. Furthermore, phosphorylated SMAD2/3 levels were elevated and nuclear localization of phosphorylated SMAD2/3 was increased, suggesting activation of the SMAD pathway in scleroderma fibroblasts. Blockade of autocrine TGFbeta signaling with antibodies or by expression of dominant-negative TGFbetaRII failed to normalize SMAD subcellular distribution, suggesting that elevated nuclear SMAD import was due to alterations downstream of the TGFbeta receptors. The activity of a SMAD-responsive minimal promoter-reporter construct was enhanced in transiently transfected scleroderma fibroblasts. CONCLUSION This study is the first to demonstrate apparently ligand-independent constitutive activation of the intracellular TGFbeta/SMAD signaling axis in scleroderma fibroblasts. SMAD signaling may be a mechanism contributing to the characteristic phenotype of scleroderma fibroblasts and playing a role in the pathogenesis of fibrosis.
Collapse
Affiliation(s)
- Yasuji Mori
- University of Illinois at Chicago College of Medicine, Chicago, IL 60607, USA
| | | | | |
Collapse
|
137
|
Schultze-Mosgau S, Wehrhan F, Rödel F, Amann K, Radespiel-Tröger M, Grabenbauer GG. Transforming growth factor-beta receptor-II up-regulation during wound healing in previously irradiated graft beds in vivo. Wound Repair Regen 2003; 11:297-305. [PMID: 12846918 DOI: 10.1046/j.1524-475x.2003.11410.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wound healing disorders may often present in patients with head and neck cancer after surgical interventions, particularly in preirradiated tissue. Inflammatory changes and the expression of cytokines can lead to induction of fibrosis. The isoforms of the transforming growth factor beta (TGFbeta1-3) play a key role for this process. It has been shown that radiation treatment associated fibrosis is induced by TGFbeta1 and TGFbeta2, although the influence of radiation on the expression of the TGFbeta receptor-II (TGFbetaR-II) involved in the signal transduction of TGFbeta remains elusive. The objective of this in vivo study was to analyze the expression profile of TGFbetaR-II in the graft bed and in the transition area between graft and graft bed after surgery with and without prior radiation treatment to compare with the expression profiles of activated TGFbeta1 and latency-associated peptide. A total of 48 Wistar rats (male, weight 300-500 g) were used in the study. Eighteen rats were irradiated in the neck region (3 x 10 Gy) without transplantation. A free myocutaneous gracilis flap was transplanted in 30 rats, of which 16 animals were preirradiated in the neck region (3 x 10 Gy) and 14 animals were not irradiated at all. Tissue samples were taken postoperatively from the transition area between the graft and the graft bed and from the graft bed itself after 3, 7, 14, and 28 days. Tissue samples were taken from the irradiated neck region and the non-irradiated groin region 0, 4, 7, 11, 14, and 28 days after the end of the exposure. The expression of TGFbetaR-II, activated TGFbeta1 and latency-associated peptide was analyzed immunohistochemically both qualitatively and quantitatively (labeling index). The success rate for graft healing was 75% in the previously irradiated group with 30 Gy, and 86% in the non-irradiated group. Following radiation alone a significantly (p = 0.04) increased TGFbetaR-II expression in the neck was revealed 2-4 weeks following irradiation compared to non-irradiated skin. Whereas only minor differences in TGFbetaR-II expression were observed following surgery between the groups with and without prior radiation in the transition area between the graft and the graft bed, the group undergoing prior radiation and subsequent grafting showed significantly increased expression in the bed compared to the non-preirradiated group with a maximum on postoperative day 7 (week 1, p = 0.003; week 2-4, p < 0.001). In irradiated tissues the up-regulation of TGFbetaR-II expression correlated with an increase of activated TGFbeta1 and latency-associated peptide expression compared to non-irradiated tissues. After irradiation, a significantly increased TGFbetaR-II expression was identified in the irradiated graft bed, which may be the reason for delayed reepithelialization and fibrosis. Exogenous blocking or TGFbetaR-II inhibitors could therefore represent a new therapeutic approach for improving wound healing after preoperative radiotherapy.
Collapse
Affiliation(s)
- Stefan Schultze-Mosgau
- Department of Oral and Maxillofacial Surgery, University of Erlangen-Nuremberg, Erlangen, Germany.
| | | | | | | | | | | |
Collapse
|
138
|
Tuamokumo NL, Haffty BG. Clinical outcome and cosmesis in African-American patients treated with conservative surgery and radiation therapy. Cancer J 2003; 9:313-20. [PMID: 12967142 DOI: 10.1097/00130404-200307000-00014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE The purpose of this study was to evaluate the prognostic significance of race in breast cancer patients treated with lumpectomy and radiation therapy. METHODS AND MATERIALS Between 1973 and 1997, 1614 patients were treated with lumpectomy and radiation therapy at our institution. All patient data, including race, age, stage, pathology, treatment, and outcome variables, were entered into a computerized database. One hundred and one women were identified as African American, and 1513 were identified as white. A small number (22) of patients of Asian, Hispanic, or other ethnic groups were eliminated from analysis. A detailed cosmetic analysis was performed on a selected subset of 20 African-American patients and 20 white patients from the database. The two groups were intentionally matched by age, follow-up, adjuvant therapy, and breast size and were asked to participate in a detailed cosmetic evaluation. RESULTS As of September 2001, median follow-up was 14.5 years. African-American patients presented with an earlier age of onset than white patients (51.1 African-American patients vs 56.5 white). By age groups, 42.5% of African-American patients were older than 50 years, compared with 68.6% of white patients. African-American patients presented with larger tumors (mean pathological size, 1.87 cm in African-American patients vs 1.57 cm in white patients) and were more frequently estrogen receptor negative (51% estrogen receptor-negative African-American patients vs 37% estrogen receptor-negative white patients). However, nodal status was similar in the two populations (27% node-positive African-American patients vs 24% node-positive white patients). Given their younger age and estrogen receptor negativity, African-American patients were more likely to receive chemotherapy, whereas white patients were more likely to receive adjuvant tamoxifen. Despite the earlier age of onset and larger tumor size, there were no significant differences between the African-American and white populations with respect to overall survival (82% in the African-American population vs 79% in the white) or cause-specific survival at 10 years (88% in the African-American population vs 86% in the white). African-American patients had a significantly lower breast relapse-free rate at 10 years (81% in the African-American population vs 87% in the white). Although this may be in part related to the younger age in the African-American patients, a multivariate analysis including age, race, margin status, and treatment parameters revealed young age and African-American race to be significantly associated with local relapse. With respect to overall cosmetic outcome and all specific cosmetic measures (edema, fibrosis, and pigmentation), African-American patients fared more poorly than white patients. Overall cosmesis was good to excellent in 55% of African Americans, compared with 90% of whites. CONCLUSIONS Despite a younger age of onset and a larger tumor size, outcome in African-American patients was similar to that of white patients with respect to overall and cause-specific survival. The explanation for a slightly higher local relapse rate and poorer cosmetic result requires further investigation.
Collapse
Affiliation(s)
- Nimi L Tuamokumo
- Department of Therapeutic Radiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8040, USA
| | | |
Collapse
|
139
|
Abstract
Historically, post acne scarring has not been well treated. New techniques have been added and older ones modified to manage this hitherto refractory problem. The patient, his or her expectations and overall appearance as well as the morphology of each scar must be assessed and treatment designed accordingly. Upon reaching an understanding of what the pathology of the scar is and where it resides in the skin, the most pertinent treatment for that scar may be devised. Post acne scars are polymorphous and include superficial macules, dermal troughs, ice picks, multi-channelled fistulous tracts and subcutaneous atrophy. The wide variety of new methods includes the latest resurfacing tools such as CO(2) and erbium infrared lasers, dermasanding and possibly some future techniques such as non-ablative and radiofrequency resurfacing. Dermal and subcutaneous augmentation with autologous (including fat and blood transfer) and non-autologous tissue augmentation and the advent of tissue undermining has greatly improved the treatment of atrophic scars. Use of punch techniques for sharply marginated scars (such as ice picks) is necessary if this scar morphology is to be treated well. One should attempt to match each scar against an available treatment as far as possible. Many of these techniques may be performed in a single treatment session but repeat treatments are often necessary. The treatment of hypertrophic acne scarring remains difficult, but silastic sheeting, vascular laser and intralesional cytotoxics are interesting developments. Most often occurring extra-facially and in males, these distressing scars often require multiple treatments and modalities before adequate improvement is achieved.
Collapse
Affiliation(s)
- Greg Goodman
- Skin and Cancer Foundation of Victoria, Carlton, Victoria, Australia.
| |
Collapse
|
140
|
Hasegawa T, Nakao A, Sumiyoshi K, Tsuboi R, Ogawa H. IFN-gamma fails to antagonize fibrotic effect of TGF-beta on keloid-derived dermal fibroblasts. J Dermatol Sci 2003; 32:19-24. [PMID: 12788525 DOI: 10.1016/s0923-1811(03)00044-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Interferon-gamma (IFN-gamma) has been noted as a potential therapeutic agent for various fibrotic disorders, in part, through its antagonistic effect on a fibrogenic cytokine, transforming growth factor-beta (TGF-beta). Keloid is a fibrotic skin disorder that results in an excessive deposition of extracellular matrix, which is associated with altered-expression of or -responses to TGF-beta in dermal fibroblasts. OBJECTIVE We sought to determine whether IFN-gamma antagonized TGF-beta-mediated fibrotic response in keloid-derived dermal fibroblasts. METHODS Type I collagen production, fibroblast contractile activity, and alpha-smooth muscle actin (alpha-SMA) expression were assessed by using Western blotting, an in vitro type I collagen gel contraction assay, and immunofluorescence study in normal and keloid-derived human dermal fibroblasts in the presence or absence of IFN-gamma and/or TGF-beta. RESULTS In contrast to normal dermal fibroblasts, IFN-gamma did not inhibit TGF-beta-induced type I collagen production, contractile activity, and alpha-SMA expression in keloid-derived dermal fibroblasts. In addition, keloid-derived dermal fibroblasts constitutively expressed type I collagen and alpha-SMA with increased capacity to contract a collagen matrix. CONCLUSION IFN-gamma failed to antagonize TGF-beta-mediated fibrotic response in keloid-derived dermal fibroblasts. Thus, IFN-gamma may not be therapeutically useful for keloid and clarification of the molecular mechanisms underlying the IFN-gamma resistance should be investigated for therapeutic application of IFN-gamma for keloid.
Collapse
Affiliation(s)
- Toshio Hasegawa
- Department of Dermatology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | | | | | | | | |
Collapse
|
141
|
Daian T, Ohtsuru A, Rogounovitch T, Ishihara H, Hirano A, Akiyama-Uchida Y, Saenko V, Fujii T, Yamashita S. Insulin-like growth factor-I enhances transforming growth factor-beta-induced extracellular matrix protein production through the P38/activating transcription factor-2 signaling pathway in keloid fibroblasts. J Invest Dermatol 2003; 120:956-62. [PMID: 12787120 DOI: 10.1046/j.1523-1747.2003.12143.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Keloids are benign dermal tumors, characterized by invasive growth of fibroblasts and concomitant increased biosynthesis of extracellular matrix components, with unclear etiology. We previously demonstrated that keloid fibroblasts overexpress insulin-like growth factor-I receptor. In investigating the role of insulin-like growth factor-I receptor overexpression, insulin-like growth factor-I and transforming growth factor-beta interaction was examined in relation to extracellular matrix protein production in cultured human and mouse fibroblasts. Western blotting revealed that collagen type I was expressed in keloid and normal fibroblasts, and its expression was increased by transforming growth factor-beta stimulation more significantly in keloid rather than in normal fibroblasts. Insulin-like growth factor-I and transforming growth factor-beta1 costimulation markedly increased extracellular matrix proteins (collagen type I, fibronectin, and plasminogen activator inhibitor-1) compared with cultures with transforming growth factor-beta1 alone. Insulin-like growth factor-I treatment alone had no stimulatory effect. Real-time reverse transcription-polymerase chain reaction confirmed parallel collagen type I messenger RNA level changes. Luciferase assays were conducted to investigate intracellular signaling pathways in this synergistic stimulation using a mouse fibroblast cell line. Transforming growth factor-beta1 (1 or 10 ng per ml) increased the specific signaling activity approximately 10-fold, whereas the increase with insulin-like growth factor-I (100 ng per ml) was less than 2-fold compared with basal activity; however, the combination of transforming growth factor-beta1 and insulin-like growth factor-I resulted in an approximately 25-fold increase. Insulin-like growth factor-I markedly enhanced transforming growth factor-beta-induced phosphorylation of p38 mitogen-activated protein kinase and activating transcription factor-2. Luciferase assay showed that this synergistic effect was attenuated by the p38 mitogen-activated protein kinase specific inhibitor SB203580 or phosphatidylinositol 3-kinase inhibitor wortmannin, but not by the mitogen-activated protein kinase/extracellular-signal-regulated protein kinase kinase inhibitor PD98059. These results indicate that insulin-like growth factor-I enhances transforming growth factor-beta-induced keloid formation through transforming growth factor-beta postreceptor signal cross-talk, mainly via the p38 mitogen-activated protein kinase/activating transcription factor-2 pathway.
Collapse
Affiliation(s)
- Takehiro Daian
- Department of Plastic and Reconstructive Surgery, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Tuan TL, Wu H, Huang EY, Chong SSN, Laug W, Messadi D, Kelly P, Le A. Increased plasminogen activator inhibitor-1 in keloid fibroblasts may account for their elevated collagen accumulation in fibrin gel cultures. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 162:1579-89. [PMID: 12707042 PMCID: PMC1851185 DOI: 10.1016/s0002-9440(10)64292-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Proteolytic degradation of the provisional fibrin matrix and subsequent substitution by fibroblast-produced collagen are essential features of injury repair. Immunohistochemical studies revealed that although dermal fibroblasts of normal scars and keloids expressed both urokinase type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1), keloid fibroblasts had a much higher PAI-1 expression. In long-term three-dimensional fibrin gel cultures (the in vitro fibroplasia model), normal fibroblasts expressed moderate and modulated activity levels of uPA and PAI-1. In contrast, keloid fibroblasts expressed a persistently high level of PAI-1 and a low level of uPA. The high PAI-1 activity of keloid fibroblasts correlated with their elevated collagen accumulation in fibrin gel cultures. Substituting collagen for fibrin in the gel matrix resulted in increased uPA activity and reduced collagen accumulation of keloid fibroblasts. Furthermore, decreasing PAI-1 activity of keloid fibroblasts in fibrin gel cultures with anti-PAI-1-neutralizing antibodies also resulted in a reduction in collagen accumulation by keloid fibroblasts. Cumulatively, these results suggest that PAI-1 overexpression is a consistent feature of keloid fibroblasts both in vitro and in vivo, and PAI-1 may play a causative role in elevated collagen accumulation of keloid fibroblasts.
Collapse
Affiliation(s)
- Tai-Lan Tuan
- Department of Surgery, Childrens Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles 90027, USA.
| | | | | | | | | | | | | | | |
Collapse
|
143
|
Abstract
Fibroproliferative scars remain an ongoing clinical challenge. Both hypertrophic scars and keloids require multimodal therapy toachieve partally successful treatment. At the present time incomplete understanding about the pathogenesis of fibroproliferative scars makes targeted, mechanistic treatment impossible. As understanding of these abnormal wound problems increases, more effective treatments will likely be available. Until that time, clinicians must utilize existing knowledge to treat patients while continuing to experiment with new approaches.
Collapse
Affiliation(s)
- Shahrad R Rahban
- University of Southern California Keck School of Medicine, Division of Plastic and Reconstructive Surgery, LAC+ USC Medical Center, 1450 San Pablo Street, Suite #2000, Los Angeles, CA 90033, USA
| | | |
Collapse
|
144
|
|
145
|
Chipev CC, Simon M. Phenotypic differences between dermal fibroblasts from different body sites determine their responses to tension and TGFbeta1. BMC DERMATOLOGY 2002; 2:13. [PMID: 12445328 PMCID: PMC138803 DOI: 10.1186/1471-5945-2-13] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2002] [Accepted: 11/21/2002] [Indexed: 11/10/2022]
Abstract
BACKGROUND Wounds in the nonglabrous skin of keloid-prone individuals tend to cause large disordered accumulations of collagen which extend beyond the original margins of the wound. In addition to abnormalities in keloid fibroblasts, comparison of dermal fibroblasts derived from nonwounded glabrous or nonglabrous skin revealed differences that may account for the observed location of keloids. METHODS Fibroblast apoptosis and the cellular content of alpha-smooth-muscle actin, TGFbeta1 receptorII and ED-A fibronectin were estimated by FACS analysis. The effects of TGFbeta1 and serum were examined. RESULTS In monolayer cultures non-glabrous fibroblasts were slower growing, had higher granularity and accumulated more alpha-smooth-muscle actin than fibroblasts from glabrous tissues. Keloid fibroblasts had the highest level of alpha-smooth-muscle actin in parallel with their expression level of ED-A fibronectin. TGFbeta1 positively regulated alpha-smooth-muscle actin expression in all fibroblast cultures, although its effects on apoptosis in fibroblasts from glabrous and non-glabrous tissues were found to differ. The presence of collagen I in the ECM resulted in reduction of alpha-smooth-muscle actin. A considerable percentage of the apoptotic fibroblasts in attached gels were alpha-smooth-muscle actin positive. The extent of apoptosis correlated positively with increased cell and matrix relaxation. TGFbeta1 was unable to overcome this apoptotic effect of matrix relaxation. CONCLUSION The presence of myofibroblasts and the apoptosis level can be regulated by both TGFbeta1 and by the extracellular matrix. However, reduction of tension in the matrix is the critical determinant. This predicts that the tension in the wound bed determines the type of scar at different body sites.
Collapse
Affiliation(s)
- Constantin C Chipev
- Living Skin Bank, University Hospital, Dept. Oral Biology and Pathology, Dept. of Dermatology, HSC, SUNY at Stony Brook, NY 11794-9702, USA
| | - Marcia Simon
- Living Skin Bank, University Hospital, Dept. Oral Biology and Pathology, Dept. of Dermatology, HSC, SUNY at Stony Brook, NY 11794-9702, USA
| |
Collapse
|
146
|
Varga J. Scleroderma and Smads: dysfunctional Smad family dynamics culminating in fibrosis. ARTHRITIS AND RHEUMATISM 2002; 46:1703-13. [PMID: 12124852 DOI: 10.1002/art.10413] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- John Varga
- Section of Rheumatology, University of Illinois at Chicago, College of Medicine, 60607, USA.
| |
Collapse
|
147
|
Huang M, Sharma S, Zhu LX, Keane MP, Luo J, Zhang L, Burdick MD, Lin YQ, Dohadwala M, Gardner B, Batra RK, Strieter RM, Dubinett SM. IL-7 inhibits fibroblast TGF-beta production and signaling in pulmonary fibrosis. J Clin Invest 2002. [PMID: 11927620 DOI: 10.1172/jci0214685] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Based on studies by our group and others, we hypothesized that IL-7 may possess antifibrotic activities in an IFN-gamma-dependent and independent manner. Here, we have evaluated the antifibrotic therapeutic potential of IL-7 in both in vitro and in vivo pulmonary fibrosis models. IL-7 inhibited both TGF-beta production and signaling in fibroblasts and required an intact JAK1/STAT1 signal transduction pathway. IL-7-mediated inhibition of TGF-beta signaling was found to be associated with an increase in Smad7, a major inhibitory regulator in the SMAD family. In the presence of IL-7, Smad7 dominant negative fibroblasts restored TGF-beta-induced collagen synthesis, indicating that an IL-7-mediated increase in Smad7 suppressed TGF-beta signaling. Consistent with these in vitro findings, recombinant IL-7 decreased bleomycin-induced pulmonary fibrosis in vivo, independent of IFN-gamma. The antifibrotic activities of IL-7 merit further basic and clinical investigation for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Min Huang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California Los Angeles School of Medicine, Los Angeles, California 90095-1690, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Huang M, Sharma S, Zhu LX, Keane MP, Luo J, Zhang L, Burdick MD, Lin YQ, Dohadwala M, Gardner B, Batra RK, Strieter RM, Dubinett SM. IL-7 inhibits fibroblast TGF-beta production and signaling in pulmonary fibrosis. J Clin Invest 2002; 109:931-7. [PMID: 11927620 PMCID: PMC150933 DOI: 10.1172/jci14685] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Based on studies by our group and others, we hypothesized that IL-7 may possess antifibrotic activities in an IFN-gamma-dependent and independent manner. Here, we have evaluated the antifibrotic therapeutic potential of IL-7 in both in vitro and in vivo pulmonary fibrosis models. IL-7 inhibited both TGF-beta production and signaling in fibroblasts and required an intact JAK1/STAT1 signal transduction pathway. IL-7-mediated inhibition of TGF-beta signaling was found to be associated with an increase in Smad7, a major inhibitory regulator in the SMAD family. In the presence of IL-7, Smad7 dominant negative fibroblasts restored TGF-beta-induced collagen synthesis, indicating that an IL-7-mediated increase in Smad7 suppressed TGF-beta signaling. Consistent with these in vitro findings, recombinant IL-7 decreased bleomycin-induced pulmonary fibrosis in vivo, independent of IFN-gamma. The antifibrotic activities of IL-7 merit further basic and clinical investigation for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Min Huang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California Los Angeles School of Medicine, Los Angeles, California 90095-1690, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|