Generic placeholder image

Current Molecular Medicine

Editor-in-Chief

ISSN (Print): 1566-5240
ISSN (Online): 1875-5666

Research Article

Hepatocyte Growth Factor Secreted from Human Adipose-Derived Stem Cells Inhibits Fibrosis in Hypertrophic Scar Fibroblasts

Author(s): Ji Ma, Xin Yan, Yue Lin and Qian Tan*

Volume 20, Issue 7, 2020

Page: [558 - 571] Pages: 14

DOI: 10.2174/1566524020666200106095745

Price: $65

Abstract

Aims: To study the effect of Adipose-derived stem cells (ADSCs) on fibrosis of hypertrophic scar-derived fibroblasts (HSFs) and its concrete mechanism.

Background: ADSCs have been reported to reduce collagen production and fibroblast proliferation in co-culture experiments. Conditioned medium from adipose-derived stem cells (ADSCs-CM) has successfully inhibited fibrosis by decreasing the expression of collagen type І (Col1) and α-smooth muscle actin (α-SMA) in rabbit ear scar models. Hepatocyte growth factor (HGF), the primary growth factor in ADSCs-CM, has been shown to reverse fibrosis in various fibrotic diseases.

Objective: To test the hypothesis that ADSCs inhibit fibrosis of HSFs through the secretion of HGF.

Methods: HSFs were treated with DMEM containing 0%, 10%, 50% and 100% concentration of ADSCs-CM. The effect of ADSCs-CM on the viability was determined by cell viability assay, and the collagen production in HSFs was examined by Sirius red staining. Expression and secretion of fibrosis and degradation proteins were detected separately. After measuring the concentration of HGF in ADSCs-CM, the same number of HSFs were treated with 50% ADSCs-CM or HGF. HGF activity in ADSCs-CM was neutralized with a goat anti-human HGF antibody.

Results: The results demonstrated that ADSCs-CM dose-dependently decreased cell viability, expression of fibrosis molecules, and tissue inhibitor of metalloproteinases-1 (TIMP-1), and significantly increased matrix metalloproteinase-1 (MMP-1) expression in HSFs. Collagen production and the ratio of collagen type І and type III (Col1/Col3) were also suppressed by ADSCs-CM in a dose-dependent manner. When HSFs were cultured with either 50% ADSCs-CM or HGF (1 ng/ml), a similar trend was observed in gene expression and protein secretion. Adding an HGF antibody to both groups returned protein expression and secretion to basal levels but did not significantly affect the fibrosis factors in the control group.

Conclusion: Our findings revealed that adipose-derived stem cell-secreted HGF effectively inhibits fibrosis-related factors and regulates extracellular matrix (ECM) remodeling in hypertrophic scar fibroblasts.

Keywords: Adipose-derived stem cells (ADSCs), Hypertrophic scar-derived fibroblasts (HSFs), Conditioned medium from ADSCs (ADSCs-CM), Hepatocyte growth factor (HGF), Collagen, extralcellular matrix.

[1]
Zhu Z, Ding J, Tredget EE. The molecular basis of hypertrophic scars. Burns Trauma 2016; 4: 2.
[http://dx.doi.org/10.1186/s41038-015-0026-4] [PMID: 27574672]
[2]
Kerwin LY, El Tal AK, Stiff MA, Fakhouri TM. Scar prevention and remodeling: a review of the medical, surgical, topical and light treatment approaches. Int J Dermatol 2014; 53(8): 922-36.
[http://dx.doi.org/10.1111/ijd.12436] [PMID: 24697346]
[3]
Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med 1999; 341(10): 738-46.
[http://dx.doi.org/10.1056/NEJM199909023411006] [PMID: 10471461]
[4]
Bai X, He T, Liu J, et al. Loureirin B inhibits fibroblast proliferation and extracellular matrix deposition in hypertrophic scar via TGF-β/Smad pathway. Exp Dermatol 2015; 24(5): 355-60.
[http://dx.doi.org/10.1111/exd.12665] [PMID: 25683490]
[5]
Spiekman M, Przybyt E, Plantinga JA, Gibbs S, van der Lei B, Harmsen MC. Adipose tissue-derived stromal cells inhibit TGF-β1-induced differentiation of human dermal fibroblasts and keloid scar-derived fibroblasts in a paracrine fashion. Plast Reconstr Surg 2014; 134(4): 699-712.
[http://dx.doi.org/10.1097/PRS.0000000000000504] [PMID: 25357030]
[6]
Penn JW, Grobbelaar AO, Rolfe KJ. The role of the TGF-β family in wound healing, burns and scarring: a review. Int J Burns Trauma 2012; 2(1): 18-28.
[PMID: 22928164]
[7]
Gauglitz GG, Korting HC, Pavicic T, Ruzicka T, Jeschke MG. Hypertrophic scarring and keloids: pathomechanisms and current and emerging treatment strategies. Mol Med 2011; 17(1-2): 113-25.
[http://dx.doi.org/10.2119/molmed.2009.00153] [PMID: 20927486]
[8]
Pakyari M, Farrokhi A, Maharlooei MK, Ghahary A. Critical role of transforming growth factor beta in different phases of wound healing. Adv Wound Care (New Rochelle) 2013; 2(5): 215-24.
[http://dx.doi.org/10.1089/wound.2012.0406] [PMID: 24527344]
[9]
Tan J, Wu J. Current progress in understanding the molecular pathogenesis of burn scar contracture. Burns Trauma 2017; 5: 14.
[http://dx.doi.org/10.1186/s41038-017-0080-1] [PMID: 28546987]
[10]
Mehta M, Branford OA, Rolfe KJ. The evidence for natural therapeutics as potential anti-scarring agents in burn-related scarring. Burns Trauma 2016; 4: 15.
[http://dx.doi.org/10.1186/s41038-016-0040-1] [PMID: 27574685]
[11]
Klinger M, Caviggioli F, Klinger FM, et al. Autologous fat graft in scar treatment. J Craniofac Surg 2013; 24(5): 1610-5.
[http://dx.doi.org/10.1097/SCS.0b013e3182a24548] [PMID: 24036737]
[12]
Lee WJ, Ahn HM, Roh H, et al. Decorin-expressing adenovirus decreases collagen synthesis and upregulates MMP expression in keloid fibroblasts and keloid spheroids. Exp Dermatol 2015; 24(8): 591-7.
[http://dx.doi.org/10.1111/exd.12719] [PMID: 25865370]
[13]
Bruno A, Delli Santi G, Fasciani L, Cempanari M, Palombo M, Palombo P. Burn scar lipofilling: immunohistochemical and clinical outcomes. J Craniofac Surg 2013; 24(5): 1806-14.
[http://dx.doi.org/10.1097/SCS.0b013e3182a148b9] [PMID: 24036785]
[14]
Klinger M, Marazzi M, Vigo D, Torre M. Fat injection for cases of severe burn outcomes: a new perspective of scar remodeling and reduction. Aesthetic Plast Surg 2008; 32(3): 465-9.
[http://dx.doi.org/10.1007/s00266-008-9122-1] [PMID: 18305985]
[15]
Caviggioli F, Klinger F, Villani F, Fossati C, Vinci V, Klinger M. Correction of cicatricial ectropion by autologous fat graft. Aesthetic Plast Surg 2008; 32(3): 555-7.
[http://dx.doi.org/10.1007/s00266-008-9117-y] [PMID: 18293030]
[16]
Maione L, Memeo A, Pedretti L, et al. Autologous fat graft as treatment of post short stature surgical correction scars. Injury 2014; 45(Suppl. 6): S126-32.
[http://dx.doi.org/10.1016/j.injury.2014.10.036] [PMID: 25457332]
[17]
Gadelkarim M, Abushouk AI, Ghanem E, Hamaad AM, Saad AM, Abdel-Daim MM. Adipose-derived stem cells: Effectiveness and advances in delivery in diabetic wound healing. Biomed Pharmacother 2018; 107: 625-33.
[http://dx.doi.org/10.1016/j.biopha.2018.08.013] [PMID: 30118878]
[18]
Nie C, Yang D, Xu J, Si Z, Jin X, Zhang J. Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell Transplant 2011; 20(2): 205-16.
[http://dx.doi.org/10.3727/096368910X520065] [PMID: 20719083]
[19]
Zhou ZQ, Chen Y, Chai M, et al. Adipose extracellular matrix promotes skin wound healing by inducing the differentiation of adipose derived stem cells into fibroblasts. Int J Mol Med 2019; 43(2): 890-900.
[PMID: 30535488]
[20]
Kuo YR, Wang CT, Cheng JT, Kao GS, Chiang YC, Wang CJ. Adipose-derived stem cells accelerate diabetic wound healing through the induction of autocrine and paracrine effects. Cell Transplant 2016; 25(1): 71-81.
[http://dx.doi.org/10.3727/096368915X687921] [PMID: 25853951]
[21]
Navone SE, Pascucci L, Dossena M, et al. Decellularized silk fibroin scaffold primed with adipose mesenchymal stromal cells improves wound healing in diabetic mice. Stem Cell Res Ther 2014; 5(1): 7.
[http://dx.doi.org/10.1186/scrt396] [PMID: 24423450]
[22]
Yuan F, Lei YH, Fu XB, Sheng ZY, Cai S, Sun TZ. [Promotive effect of adipose-derived stem cells on the wound model of human epidermal keratinocytes in vitro] Zhonghua Wai Ke Za Zhi 2008; 46(20): 1575-8.
[PMID: 19094656]
[23]
Nambu M, Kishimoto S, Nakamura S, et al. Accelerated wound healing in healing-impaired db/db mice by autologous adipose tissue-derived stromal cells combined with atelocollagen matrix. Ann Plast Surg 2009; 62(3): 317-21.
[http://dx.doi.org/10.1097/SAP.0b013e31817f01b6] [PMID: 19240532]
[24]
Shen T, Pan ZG, Zhou X, Hong CY. Accelerated healing of diabetic wound using artificial dermis constructed with adipose stem cells and poly (L-glutamic acid)/chitosan scaffold. Chin Med J (Engl) 2013; 126(8): 1498-503.
[PMID: 23595384]
[25]
Wang X, Ma Y, Gao Z, Yang J. Human adipose-derived stem cells inhibit bioactivity of keloid fibroblasts. Stem Cell Res Ther 2018; 9(1): 40.
[http://dx.doi.org/10.1186/s13287-018-0786-4] [PMID: 29467010]
[26]
Yun IS, Jeon YR, Lee WJ, et al. Effect of human adipose derived stem cells on scar formation and remodeling in a pig model: a pilot study. Dermatol Surg 2012; 38(10): 1678-88.
[http://dx.doi.org/10.1111/j.1524-4725.2012.02495.x] [PMID: 22804839]
[27]
Zhang Q, Liu LN, Yong Q, Deng JC, Cao WG. Intralesional injection of adipose-derived stem cells reduces hypertrophic scarring in a rabbit ear model. Stem Cell Res Ther 2015; 6: 145.
[http://dx.doi.org/10.1186/s13287-015-0133-y] [PMID: 26282394]
[28]
Rehman J, Traktuev D, Li J, et al. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation 2004; 109(10): 1292-8.
[http://dx.doi.org/10.1161/01.CIR.0000121425.42966.F1] [PMID: 14993122]
[29]
Komaki Y, Kanmura S, Sasaki F, et al. Hepatocyte growth factor facilitates esophageal mucosal repair and inhibits the submucosal fibrosis in a rat model of esophageal ulcer. Digestion 2019; 99(3): 227-38.
[http://dx.doi.org/10.1159/000491876] [PMID: 30227416]
[30]
Liu Y. Hepatocyte growth factor in kidney fibrosis: therapeutic potential and mechanisms of action. Am J Physiol Renal Physiol 2004; 287(1): F7-F16.
[http://dx.doi.org/10.1152/ajprenal.00451.2003] [PMID: 15180923]
[31]
Jinnin M, Ihn H, Mimura Y, Asano Y, Yamane K, Tamaki K. Matrix metalloproteinase-1 up-regulation by hepatocyte growth factor in human dermal fibroblasts via ERK signaling pathway involves Ets1 and Fli1. Nucleic Acids Res 2005; 33(11): 3540-9.
[http://dx.doi.org/10.1093/nar/gki648] [PMID: 15972796]
[32]
Arpino V, Brock M, Gill SE. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biol 2015; 44-46: 247-54.
[http://dx.doi.org/10.1016/j.matbio.2015.03.005] [PMID: 25805621]
[33]
Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol 2007; 8(3): 221-33.
[http://dx.doi.org/10.1038/nrm2125] [PMID: 17318226]
[34]
Mehrvarz S, Ebrahimi A, Sahraei H, et al. Effects of topical tamoxifen on wound healing of burned skin in rats. Arch Plast Surg 2017; 44(5): 378-83.
[http://dx.doi.org/10.5999/aps.2017.44.5.378] [PMID: 28946718]
[35]
Chrysanthopoulou A, Mitroulis I, Apostolidou E, et al. Neutrophil extracellular traps promote differentiation and function of fibroblasts. J Pathol 2014; 233(3): 294-307.
[http://dx.doi.org/10.1002/path.4359] [PMID: 24740698]
[36]
Li Y, Zhang W, Gao J, et al. Adipose tissue-derived stem cells suppress hypertrophic scar fibrosis via the p38/MAPK signaling pathway. Stem Cell Res Ther 2016; 7(1): 102.
[http://dx.doi.org/10.1186/s13287-016-0356-6] [PMID: 27484727]
[37]
Altalhi W, Hatkar R, Hoying JB, Aghazadeh Y, Nunes SS. Type I diabetes delays perfusion and engraftment of 3d constructs by impinging on angiogenesis; which can be rescued by hepatocyte growth factor supplementation. Cell Mol Bioeng 2019; 12(5): 443-54.
[http://dx.doi.org/10.1007/s12195-019-00574-3] [PMID: 31719926]
[38]
Suga H, Eto H, Shigeura T, et al. IFATS collection: Fibroblast growth factor-2-induced hepatocyte growth factor secretion by adipose-derived stromal cells inhibits postinjury fibrogenesis through a c-Jun N-terminal kinase-dependent mechanism. Stem Cells 2009; 27(1): 238-49.
[http://dx.doi.org/10.1634/stemcells.2008-0261] [PMID: 18772314]
[39]
Zhu XY, Zhang XZ, Xu L, et al. Transplantation of adipose-derived stem cells overexpressing hHGF into cardiac tissue. Biochem and Biophysical Res Communications 2009; 379: 0-1090.
[http://dx.doi.org/10.1016/j.bbrc.2009.01.019]
[40]
Ueki T, Kaneda Y, Tsutsui H, et al. Hepatocyte growth factor gene therapy of liver cirrhosis in rats. Nat Med 1999; 5(2): 226-30.
[http://dx.doi.org/10.1038/5593] [PMID: 9930873]
[41]
Tang WP, Akahoshi T, Piao JS, et al. Basic fibroblast growth factor-treated adipose tissue-derived mesenchymal stem cell infusion to ameliorate liver cirrhosis via paracrine hepatocyte growth factor. J Gastroenterol Hepatol 2015; 30(6): 1065-74.
[http://dx.doi.org/10.1111/jgh.12893] [PMID: 25639333]
[42]
Dong LH, Jiang YY, Liu YJ, et al. The anti-fibrotic effects of mesenchymal stem cells on irradiated lungs via stimulating endogenous secretion of HGF and PGE2. Sci Rep 2015; 5: 8713.
[http://dx.doi.org/10.1038/srep08713] [PMID: 25736907]
[43]
Rawlins JM, Lam WL, Karoo RO, Naylor IL, Sharpe DT. Quantifying collagen type in mature burn scars: a novel approach using histology and digital image analysis. J Burn Care Res 2006; 27(1): 60-5.
[http://dx.doi.org/10.1097/01.bcr.0000192266.14329.7b] [PMID: 16566538]
[44]
Lee G, Hunter-Smith DJ, Rozen WM. Autologous fat grafting in keloids and hypertrophic scars: a review. Scars Burn Heal 2017; 32059513117700157
[http://dx.doi.org/10.1177/2059513117700157] [PMID: 29799555]
[45]
Zhan W, Tan SS, Lu F. Adipose-derived stem cell delivery for adipose tissue engineering: current status and potential applications in a tissue engineering chamber model. Stem Cell Rev Rep 2016; 12(4): 484-91.
[http://dx.doi.org/10.1007/s12015-016-9653-9] [PMID: 27075632]
[46]
Pham PV. Adipose stem cells in the clinic. Biomed Res Ther 2014; 1: 57-70.
[47]
Wu YY, Jiao YP, Xiao LL, et al. Experimental study on effects of adipose-derived stem cell-seeded silk fibroin chitosan film on wound healing of a diabetic rat model. Ann Plast Surg 2018; 80(5): 572-80.
[http://dx.doi.org/10.1097/SAP.0000000000001355] [PMID: 29443833]
[48]
Kim JH, Jung M, Kim HS, Kim YM, Choi EH. Adipose-derived stem cells as a new therapeutic modality for ageing skin. Exp Dermatol 2011; 20(5): 383-7.
[http://dx.doi.org/10.1111/j.1600-0625.2010.01221.x] [PMID: 21355887]
[49]
Shen X, Du Y, Shen W, Xue B, Zhao Y. Adipose-derived stem cells promote human dermal fibroblast function and increase senescence-associated β galactosidase mRNA expression through paracrine effects. Mol Med Rep 2014; 10(6): 3068-72.
[http://dx.doi.org/10.3892/mmr.2014.2627] [PMID: 25310816]
[50]
Wang T, Guo S, Liu X, Xv N, Zhang S. Protective effects of adipose-derived stem cells secretome on human dermal fibroblasts from ageing damages. Int J Clin Exp Pathol 2015; 8(12): 15739-48.
[PMID: 26884843]
[51]
Kim WS, Park BS, Sung JH, et al. Wound healing effect of adipose-derived stem cells: a critical role of secretory factors on human dermal fibroblasts. J Dermatol Sci 2007; 48(1): 15-24.
[http://dx.doi.org/10.1016/j.jdermsci.2007.05.018] [PMID: 17643966]
[52]
Jinnin M, Ihn H, Mimura Y, Asano Y, Yamane K, Tamaki K. Effects of hepatocyte growth factor on the expression of type I collagen and matrix metalloproteinase-1 in normal and scleroderma dermal fibroblasts. J Invest Dermatol 2005; 124(2): 324-30.
[http://dx.doi.org/10.1111/j.0022-202X.2004.23601.x] [PMID: 15675950]
[53]
Mukhopadhyay A, Fan S, Dang VD, et al. The role of hepatocyte growth factor/c-Met system in keloid pathogenesis. J Trauma 2010; 69(6): 1457-66.
[http://dx.doi.org/10.1097/TA.0b013e3181f45f71] [PMID: 21150524]
[54]
Pierce GF, Tarpley JE, Yanagihara D, Mustoe TA, Fox GM, Thomason A. Platelet-derived growth factor (BB homodimer), transforming growth factor-beta 1, and basic fibroblast growth factor in dermal wound healing. Neovessel and matrix formation and cessation of repair. Am J Pathol 1992; 140(6): 1375-88.
[PMID: 1376557]
[55]
Ichiki Y, Smith E, LeRoy EC, Trojanowska M. Different effects of basic fibroblast growth factor and transforming growth factor-beta on the two platelet-derived growth factor receptors’ expression in scleroderma and healthy human dermal fibroblasts. J Invest Dermatol 1995; 104(1): 124-7.
[http://dx.doi.org/10.1111/1523-1747.ep12613617] [PMID: 7798629]
[56]
Kach J, Sandbo N, Sethakorn N, et al. Regulation of myofibroblast differentiation and bleomycin-induced pulmonary fibrosis by adrenomedullin. Am J Physiol Lung Cell Mol Physiol 2013; 304(11): L757-64.
[http://dx.doi.org/10.1152/ajplung.00262.2012] [PMID: 23585227]
[57]
Li L, Zhang S, Zhang Y, Yu B, Xu Y, Guan Z. Paracrine action mediate the antifibrotic effect of transplanted mesenchymal stem cells in a rat model of global heart failure. Mol Biol Rep 2009; 36(4): 725-31.
[http://dx.doi.org/10.1007/s11033-008-9235-2] [PMID: 18368514]
[58]
Verma SK, Garikipati VNS, Krishnamurthy P, et al. Interleukin-10 inhibits bone marrow fibroblast progenitor cell-mediated cardiac fibrosis in pressure-overloaded myocardium. Circulation 2017; 136(10): 940-53.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.117.027889] [PMID: 28667100]
[59]
Németh K, Leelahavanichkul A, Yuen PS, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med 2009; 15(1): 42-9.
[http://dx.doi.org/10.1038/nm.1905] [PMID: 19098906]
[60]
Carceller MC, Guillén MI, Ferrándiz ML, Alcaraz MJ. Paracrine in vivo inhibitory effects of adipose tissue-derived mesenchymal stromal cells in the early stages of the acute inflammatory response. Cytotherapy 2015; 17(9): 1230-9.
[http://dx.doi.org/10.1016/j.jcyt.2015.06.001] [PMID: 26276006]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy