The IL-6 trans-signaling-STAT3 pathway mediates ECM and cellular proliferation in fibroblasts from hypertrophic scar

Sutapa Ray, Xiaoxi Ju, Hong Sun, Celeste C Finnerty, David N Herndon, Allan R Brasier, Sutapa Ray, Xiaoxi Ju, Hong Sun, Celeste C Finnerty, David N Herndon, Allan R Brasier

Abstract

The molecular mechanisms behind the pathogenesis of postburn hypertrophic scar (HS) remain unclear. Here, we investigate the role of the IL-6 trans-signaling-signal transducer and activator of transcription (STAT)3 pathway in HS fibroblasts (HSFs) derived from post-burn HS skin. HSF showed increased Tyr 705 STAT3 phosphorylation compared with normal fibroblast (NF) after IL-6•IL-6Rα stimulation by immunoassays. The endogenous STAT3 target gene, SOCS3, was upregulated in HSFs and showed increased STAT3 binding on its promoter relative to NFs in a chromatin immunoprecipitation assay. We observed that the cell-surface signaling transducer glycoprotein 130 is upregulated in HSFs by quantitative real-time reverse-transcriptase-PCR and flow cytometry. The production of excessive extracellular matrix (ECM), including the expression of alpha2 (1) procollagen (Col1A2) and fibronectin 1 (FN), was seen in HSFs. A STAT3 peptide inhibitor abrogated FN and Col1A2 gene expression in HSFs indicating involvement of STAT3 in ECM production. The cellular proliferation markers Cyclin D1, Bcl-Xl, and c-Myc were also upregulated in HSF, and knockdown of STAT3 by small interfering RNA attenuated c-Myc expression indicating the essential role of STAT3 in fibroblast proliferation. Taken together, our results suggest that the IL-6 trans-signaling-STAT3 pathway may have an integral role in HS pathogenesis, and disruption of this pathway could be a potential therapeutic strategy for the treatment of post-burn HS.

Trial registration: ClinicalTrials.gov NCT00675714.

Conflict of interest statement

CONFLICT OF INTERSEST

The authors state no conflict of interest.

Figures

Figure 1
Figure 1
(a) Immunohistochemical analysis of phospho-STAT3 (Tyr 705) expression in burned HSskin. ‘A-F’ and G are HS skin stained with anti-phosphoTyr705-STAT3 Ab and control IgG respectively. Panel H-I are normal skin stained with anti-phosphoTyr705-STAT3 Ab. Arrowheads (D-F) show the strong nuclear staining for activated STAT3. A-C, G-H and D-F, I are X10 and X20 magnifications respectively, scale bar: 10µm. (b) Intensity of nuclear staining in the dermis of HS (n=10) and normal skin (n=5) (MetaMorph 7.3). *, p<0.01. (c) Western Immunoblot analysis of phospho-STAT3 (Tyr 705) in HSFs. WCE of NF and HSF were fractionated and Western immunoblot analysis with phospho-STAT3 (Tyr 705) Ab is shown. The numbers at the bottom of the lane indicate the fold induction. β-actin blot (lower panel) shows loading control.
Figure 1
Figure 1
(a) Immunohistochemical analysis of phospho-STAT3 (Tyr 705) expression in burned HSskin. ‘A-F’ and G are HS skin stained with anti-phosphoTyr705-STAT3 Ab and control IgG respectively. Panel H-I are normal skin stained with anti-phosphoTyr705-STAT3 Ab. Arrowheads (D-F) show the strong nuclear staining for activated STAT3. A-C, G-H and D-F, I are X10 and X20 magnifications respectively, scale bar: 10µm. (b) Intensity of nuclear staining in the dermis of HS (n=10) and normal skin (n=5) (MetaMorph 7.3). *, p<0.01. (c) Western Immunoblot analysis of phospho-STAT3 (Tyr 705) in HSFs. WCE of NF and HSF were fractionated and Western immunoblot analysis with phospho-STAT3 (Tyr 705) Ab is shown. The numbers at the bottom of the lane indicate the fold induction. β-actin blot (lower panel) shows loading control.
Figure 1
Figure 1
(a) Immunohistochemical analysis of phospho-STAT3 (Tyr 705) expression in burned HSskin. ‘A-F’ and G are HS skin stained with anti-phosphoTyr705-STAT3 Ab and control IgG respectively. Panel H-I are normal skin stained with anti-phosphoTyr705-STAT3 Ab. Arrowheads (D-F) show the strong nuclear staining for activated STAT3. A-C, G-H and D-F, I are X10 and X20 magnifications respectively, scale bar: 10µm. (b) Intensity of nuclear staining in the dermis of HS (n=10) and normal skin (n=5) (MetaMorph 7.3). *, p<0.01. (c) Western Immunoblot analysis of phospho-STAT3 (Tyr 705) in HSFs. WCE of NF and HSF were fractionated and Western immunoblot analysis with phospho-STAT3 (Tyr 705) Ab is shown. The numbers at the bottom of the lane indicate the fold induction. β-actin blot (lower panel) shows loading control.
Figure 2
Figure 2
(a) Activation of IL-6 trans-signaling in HSF. NF and HSF cells were treated with IL-6•sIL-6Rα (8ng/ml •25ng/ml) for 30 minutes, or left untreated. Total cellular RNA was subjected to Q-RT-PCR for hSOCS3 transcript. Shown is fold change mRNA expression relative to GAPDH. Data represents mean±SD, *, p<0.01. (b) Inhibition of phospho-STAT3 with STAT3 siRNA. NFs and HSFs were transfected with either control or STAT3 siRNA. 72 hours after transfection, cells were stimulated as above and WCE were measured for phosphoTry705-STAT3 by immunoassay (Bio-Rad). Shown is the representative fluorescence intensity of phospho-STAT3 relative to total protein. (c) Inhibition of hSOCS3 gene with STAT3 silencing. Cells were treated as in (b) and total cellular RNA was subjected to Q-RT-PCR for hSOCS3 expression relative to GAPDH. *, p<0.05.
Figure 2
Figure 2
(a) Activation of IL-6 trans-signaling in HSF. NF and HSF cells were treated with IL-6•sIL-6Rα (8ng/ml •25ng/ml) for 30 minutes, or left untreated. Total cellular RNA was subjected to Q-RT-PCR for hSOCS3 transcript. Shown is fold change mRNA expression relative to GAPDH. Data represents mean±SD, *, p<0.01. (b) Inhibition of phospho-STAT3 with STAT3 siRNA. NFs and HSFs were transfected with either control or STAT3 siRNA. 72 hours after transfection, cells were stimulated as above and WCE were measured for phosphoTry705-STAT3 by immunoassay (Bio-Rad). Shown is the representative fluorescence intensity of phospho-STAT3 relative to total protein. (c) Inhibition of hSOCS3 gene with STAT3 silencing. Cells were treated as in (b) and total cellular RNA was subjected to Q-RT-PCR for hSOCS3 expression relative to GAPDH. *, p<0.05.
Figure 2
Figure 2
(a) Activation of IL-6 trans-signaling in HSF. NF and HSF cells were treated with IL-6•sIL-6Rα (8ng/ml •25ng/ml) for 30 minutes, or left untreated. Total cellular RNA was subjected to Q-RT-PCR for hSOCS3 transcript. Shown is fold change mRNA expression relative to GAPDH. Data represents mean±SD, *, p<0.01. (b) Inhibition of phospho-STAT3 with STAT3 siRNA. NFs and HSFs were transfected with either control or STAT3 siRNA. 72 hours after transfection, cells were stimulated as above and WCE were measured for phosphoTry705-STAT3 by immunoassay (Bio-Rad). Shown is the representative fluorescence intensity of phospho-STAT3 relative to total protein. (c) Inhibition of hSOCS3 gene with STAT3 silencing. Cells were treated as in (b) and total cellular RNA was subjected to Q-RT-PCR for hSOCS3 expression relative to GAPDH. *, p<0.05.
Figure 3
Figure 3
hSOCS3 promoter occupancy of STAT3 in HSF and NF. Protein-DNA crosslinked extracts of IL-6•sIL-6Rα (8ng/ml •25ng/ml) stimulated NF and HSF cells were immunoprecipitated with IgG or anti-STAT3 Ab. SOCS3 promoter occupancy of STAT3 were detected by two step ChIP assays as described in the method. Shown is the fold change in quantitative-genomic PCR (Q-gPCR) normalized to input DNA. *, p<0.01, students t- test.
Figure 4
Figure 4
(a) Activation of gp130 in HSF. NFs and HSFs were treated with IL-6•sIL-6Rα for 30 min. Total RNA was subjected to Q-RT-PCR for human gp130 mRNA expression, carried out in triplicate. Shown is fold change mRNA expression relative to GAPDH as internal control. Data represents mean±SD *, p<0.01, students t test. (b) Cell surface activation of gp130 in HSF. Cultured NF and HSF cells were left untreated or IL-6•sIL-6Rα (8ng/ml •25ng/ml) stimulated for 30 min. The expression of cell surface gp130 was analyzed by flow cytometry after staining with anti-gp130-PE. Events were plotted as a function of fluorescence intensity (x-axis). Shaded histograms represent isotype antibody control and open histograms represents either unstimulated anti-gp130-PE stained cells (dotted line) or IL-6•sIL-6Rα stimulated anti-gp130-PE (solid line) as indicated.
Figure 4
Figure 4
(a) Activation of gp130 in HSF. NFs and HSFs were treated with IL-6•sIL-6Rα for 30 min. Total RNA was subjected to Q-RT-PCR for human gp130 mRNA expression, carried out in triplicate. Shown is fold change mRNA expression relative to GAPDH as internal control. Data represents mean±SD *, p<0.01, students t test. (b) Cell surface activation of gp130 in HSF. Cultured NF and HSF cells were left untreated or IL-6•sIL-6Rα (8ng/ml •25ng/ml) stimulated for 30 min. The expression of cell surface gp130 was analyzed by flow cytometry after staining with anti-gp130-PE. Events were plotted as a function of fluorescence intensity (x-axis). Shaded histograms represent isotype antibody control and open histograms represents either unstimulated anti-gp130-PE stained cells (dotted line) or IL-6•sIL-6Rα stimulated anti-gp130-PE (solid line) as indicated.
Figure 5
Figure 5
(a) ECM expression in HSF. NFs and HSFs were stimulated with IL-6•sIL-6Rα and total cellular RNA was subjected to Q-RT-PCR for Col1A2 and FN expression. Shown is fold change mRNA expression relative to GAPDH. *, p<0.01. (b) STAT3 peptide inhibitor attenuates hSOCS3 expression. NF and HSFs were treated with 500 nM STAT3 inhibitor peptide for 6 hoursrs or left untreated. Total cellular RNA (stimulated) was subjected to Q-RT-PCR for hSOCS3 transcript. *, p<0.01. (c) STAT3 peptide inhibitor abrogates FN production in HSF. Cells were treated as in (b) and FN gene expression was measured relative to GAPDH.*, p<0.05. (d) STAT3 peptide inhibitor abrogates Col1A2 production in HSF. Cells were treated as in (b) and Col1A2 gene expression was measured relative to GAPDH. *, p<0.05.
Figure 5
Figure 5
(a) ECM expression in HSF. NFs and HSFs were stimulated with IL-6•sIL-6Rα and total cellular RNA was subjected to Q-RT-PCR for Col1A2 and FN expression. Shown is fold change mRNA expression relative to GAPDH. *, p<0.01. (b) STAT3 peptide inhibitor attenuates hSOCS3 expression. NF and HSFs were treated with 500 nM STAT3 inhibitor peptide for 6 hoursrs or left untreated. Total cellular RNA (stimulated) was subjected to Q-RT-PCR for hSOCS3 transcript. *, p<0.01. (c) STAT3 peptide inhibitor abrogates FN production in HSF. Cells were treated as in (b) and FN gene expression was measured relative to GAPDH.*, p<0.05. (d) STAT3 peptide inhibitor abrogates Col1A2 production in HSF. Cells were treated as in (b) and Col1A2 gene expression was measured relative to GAPDH. *, p<0.05.
Figure 5
Figure 5
(a) ECM expression in HSF. NFs and HSFs were stimulated with IL-6•sIL-6Rα and total cellular RNA was subjected to Q-RT-PCR for Col1A2 and FN expression. Shown is fold change mRNA expression relative to GAPDH. *, p<0.01. (b) STAT3 peptide inhibitor attenuates hSOCS3 expression. NF and HSFs were treated with 500 nM STAT3 inhibitor peptide for 6 hoursrs or left untreated. Total cellular RNA (stimulated) was subjected to Q-RT-PCR for hSOCS3 transcript. *, p<0.01. (c) STAT3 peptide inhibitor abrogates FN production in HSF. Cells were treated as in (b) and FN gene expression was measured relative to GAPDH.*, p<0.05. (d) STAT3 peptide inhibitor abrogates Col1A2 production in HSF. Cells were treated as in (b) and Col1A2 gene expression was measured relative to GAPDH. *, p<0.05.
Figure 5
Figure 5
(a) ECM expression in HSF. NFs and HSFs were stimulated with IL-6•sIL-6Rα and total cellular RNA was subjected to Q-RT-PCR for Col1A2 and FN expression. Shown is fold change mRNA expression relative to GAPDH. *, p<0.01. (b) STAT3 peptide inhibitor attenuates hSOCS3 expression. NF and HSFs were treated with 500 nM STAT3 inhibitor peptide for 6 hoursrs or left untreated. Total cellular RNA (stimulated) was subjected to Q-RT-PCR for hSOCS3 transcript. *, p<0.01. (c) STAT3 peptide inhibitor abrogates FN production in HSF. Cells were treated as in (b) and FN gene expression was measured relative to GAPDH.*, p<0.05. (d) STAT3 peptide inhibitor abrogates Col1A2 production in HSF. Cells were treated as in (b) and Col1A2 gene expression was measured relative to GAPDH. *, p<0.05.
Figure 6
Figure 6
(a) IL-6 trans-signaling increases expression of cellular proliferation markers in HSF. NF and HSF cells were stimulated with IL-6•sIL-6Rα and total cellular RNA was subjected to QPCR for Cyclin D1, c-Myc and Bcl-XL gene expression. Shown is fold change mRNA expression relative to GAPDH as internal control performed in triplicate. Data represents mean ± SD.*, p<0.05, students t test. (b) STAT3 siRNA inhibits expression of cellular proliferation markers in HSF. NF and HSF cells were transfected with either STAT3 siRNA or control siRNA. 72 h after transfection cells were stimulated with IL-6•sIL-6Rα for 30 min and total cellular RNA was subjected to Q-PCR for c-Myc, Bcl-XL and Cyclin D1 gene expression. *, p<0.01, students t test.
Figure 6
Figure 6
(a) IL-6 trans-signaling increases expression of cellular proliferation markers in HSF. NF and HSF cells were stimulated with IL-6•sIL-6Rα and total cellular RNA was subjected to QPCR for Cyclin D1, c-Myc and Bcl-XL gene expression. Shown is fold change mRNA expression relative to GAPDH as internal control performed in triplicate. Data represents mean ± SD.*, p<0.05, students t test. (b) STAT3 siRNA inhibits expression of cellular proliferation markers in HSF. NF and HSF cells were transfected with either STAT3 siRNA or control siRNA. 72 h after transfection cells were stimulated with IL-6•sIL-6Rα for 30 min and total cellular RNA was subjected to Q-PCR for c-Myc, Bcl-XL and Cyclin D1 gene expression. *, p<0.01, students t test.

References

    1. Abergel RP, Pizzurro D, Meeker CA, Lask G, Matsuoka LY, Minor RR, Chu ML, Uitto J. Biochemical composition of the connective tissue in keloids and analysis of collagen metabolism in keloid fibroblast cultures. J Invest Dermatol. 1985;84:384–390.
    1. Bombaro KM, Engrav LH, Carrougher GJ, Wiechman SA, Faucher L, Costa BA, Heimbach DM, Rivara FP, Honari S. What is the prevalence of hypertrophic scarring following burns? Burns. 2003;29:299–302.
    1. Bowman T, Broome MA, Sinibaldi D, Wharton W, Pledger WJ, Sedivy JM, Irby R, Yeatman T, Courtneidge SA, Jove R. Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc Natl Acad Sci U S A. 2001;98:7319–7324.
    1. Castagnoli C, Trombotto C, Ondei S, Stella M, Calcagni M, Magliacani G, Alasia ST. Characterization of T-cell subsets infiltrating post-burn hypertrophic scar tissues. Burns. 1997;23:565–572.
    1. Dauer DJ, Ferraro B, Song LX, Yu B, Mora L, Buettner R, Enkemann S, Jove R, Haura EB. Stat3 regulates genes common to both wound healing and cancer. Oncogene. 2005;24:3397–3408.
    1. Deitch EA, Wheelahan TM, Rose MP, Clothier J, Cotter J. Hypertrophic burn scars: analysis of variables. J Trauma. 1983;23:895–898.
    1. Ebihara N, Matsuda A, Nakamura S, Matsuda H, Murakami A. Role of the IL-6 classicand trans-signaling pathways in corneal sterile inflammation and wound healing. Invest Ophthalmol Vis Sci. 2011;52:8549–8557.
    1. Eckner R, Ewen ME, Newsome D, Gerds M, Decaprio JA, Lawrence JB, Livingston DM. Molecular cloning and functional ananlysis of the adenovirus E1A-associated 300 kD protein (p300) reveals a protein with properties of a transcriptional adaptor. Genes Dev. 1994;8:869–884.
    1. Feghali CA, Bost KL, Boulware DW, Levy LS. Control of IL-6 expression and response in fibroblasts from patients with systemic sclerosis. Autoimmunity. 1994;17:309–318.
    1. Finnerty CC, Herndon DN, Przkora R, Pereira CT, Oliveira HM, Queiroz DM, Rocha AM, Jeschke MG. Cytokine expression profile over time in severely burned pediatric patients. Shock. 2006;26:13–19.
    1. Finnerty CC, Jeschke MG, Herndon DN, Gamelli R, Gibran N, Klein M, Silver G, Arnoldo B, Remick D, Tompkins RG. Temporal cytokine profiles in severely burned patients: a comparison of adults and children. Mol Med. 2008;14:553–560.
    1. Ghazizadeh M, Tosa M, Shimizu H, Hyakusoku H, Kawanami O. Functional implications of the IL-6 signaling pathway in keloid pathogenesis. J Invest Dermatol. 2007;127:98–105.
    1. Haga S, Terui K, Zhang HQ, Enosawa S, Ogawa W, Inoue H, Okuyama T, Takeda K, Akira S, Ogino T, Irani K, Ozaki M. Stat3 protects against Fas-induced liver injury by redox-dependent and -independent mechanisms. J Clin Invest. 2003;112:989–998.
    1. Hou T, Ray S, Brasier AR. The functional role of an interleukin 6-inducible CDK9.STAT3 complex in human gamma-fibrinogen gene expression. J Biol Chem. 2007;282:37091–37102.
    1. Hou T, Ray S, Lee C, Brasier AR. The STAT3 NH2-terminal domain stabilizes enhanceosome assembly by interacting with the p300 bromodomain. J Biol Chem. 2008a;283:30725–30734.
    1. Hou T, Tieu BC, Ray S, Recinos IA, Cui R, Tilton RG, Brasier AR. Roles of IL-6-gp130 Signaling in Vascular Inflammation. Curr Cardiol Rev. 2008b;4:179–192.
    1. Igarashi A, Nashiro K, Kikuchi K, Sato S, Ihn H, Fujimoto M, Grotendorst GR, Takehara K. Connective tissue growth factor gene expression in tissue sections from localized scleroderma, keloid, and other fibrotic skin disorders. J Invest Dermatol. 1996;106(4):729–733.
    1. Jeschke MG, Chinkes DL, Finnerty CC, Kulp G, Suman OE, Norbury WB, Branski LK, Gauglitz GG, Mlcak RP, Herndon DN. Pathophysiologic response to severe burn injury. Ann Surg. 2008;248:387–401.
    1. Jones SA, Horiuchi S, Topley N, Yamamoto N, Fuller GM. The soluble interleukin 6 receptor: mechanisms of production and implications in disease. FASEB Journal. 2001;15:43–58.
    1. Jones SA, Richards PJ, Scheller J, Rose-John S. IL-6 transsignaling: the in vivo consequences. J Interferon Cytokine Res. 2005;25:241–253.
    1. Kushner I. The acute phase response: an overview. Methods in Enzymology. 1988;163:373–383.
    1. Lim CP, Phan TT, Lim IJ, Cao X. Stat3 contributes to keloid pathogenesis via promoting collagen production, cell proliferation and migration. Oncogene. 2006;25:5416–5425.
    1. Lim CP, Phan TT, Lim IJ, Cao X. Cytokine profiling and Stat3 phosphorylation in epithelialmesenchymal interactions between keloid keratinocytes and fibroblasts. J Invest Dermatol. 2009;129:851–861.
    1. Matsui T, Ito C, Oda M, Itoigawa M, Yokoo K, Okada T, Furukawa H. Lapachol suppresses cell proliferation and secretion of interleukin-6 and plasminogen activator inhibitor-1 of fibroblasts derived from hypertrophic scars. J Pharm Pharmacol. 2011;63:960–966.
    1. Nowak DE, Tian B, Brasier AR. Two-step cross-linking method for identification of NFkappaB gene network by chromatin immunoprecipitation. Biotechniques. 2005;39:715–725.
    1. O'Brien CA, Manolagas SC. Isolation and characterization of the human gp130 promoter. Regulation by STATS. J Biol Chem. 1997;272:15003–15010.
    1. 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.
    1. Ray S, Boldogh I, Brasier AR. STAT3 NH2-terminal acetylation is activated by the hepatic acute-phase response and required for IL-6 induction of angiotensinogen. Gastroenterology. 2005;129:1616–1632.
    1. Ray S, Lee C, Hou T, Bhakat KK, Brasier AR. Regulation of signal transducer and activator of transcription 3 enhanceosome formation by apurinic/apyrimidinic endonuclease 1 in hepatic acute phase response. Mol Endocrinol. 2010;24:391–401.
    1. Ray S, Lee C, Hou T, Boldogh I, Brasier AR. Requirement of histone deacetylase1 (HDAC1) in signal transducer and activator of transcription 3 (STAT3) nucleocytoplasmic distribution. Nucleic Acids Res. 2008;36:4510–4520.
    1. Sano S, Chan KS, Digiovanni J. Impact of Stat3 activation upon skin biology: a dichotomy of its role between homeostasis and diseases. J Dermatol Sci. 2008;50:1–14.
    1. Scheller J, Rose-John S. Updating interleukin-6 classic- and trans-signaling. Signal Transduction. 2006;6:240–259.
    1. Scott PG, Ghahary A, Tredget EE. Molecular and cellular aspects of fibrosis following thermal injury. Hand Clin. 2000;16:271–287.
    1. Shahar I, Fireman E, Topilsky M, Grief J, Kivity S, Spirer Z, Ben ES. Effect of IL-6 on alveolar fibroblast proliferation in interstitial lung diseases. Clin Immunol Immunopathol. 1996;79:244–251.
    1. Sheridan RL, Tompkins RG. What's new in burns and metabolism. J Am Coll Surg. 2004;198:243–263.
    1. Sidgwick GP, Bayat A. Extracellular matrix molecules implicated in hypertrophic and keloid scarring. J Eur Acad Dermatol Venereol. 2012;26:141–152.
    1. Sinibaldi D, Wharton W, Turkson J, Bowman T, Pledger WJ, Jove R. Induction of p21WAF1/CIP1 and cyclin D1 expression by the Src oncoprotein in mouse fibroblasts: role of activated STAT3 signaling. Oncogene. 2000;19:5419–5427.
    1. Sporri B, Muller KM, Wiesmann U, Bickel M. Soluble IL-6 receptor induces calcium flux and selectively modulates chemokine expression in human dermal fibroblasts. Int Immunol. 1999;11:1053–1058.
    1. Tan PL, Farmiloe S, Yeoman S, Watson JD. Expression of the interleukin 6 gene in rheumatoid synovial fibroblasts. J Rheumatol. 1990;17:1608–1612.
    1. Tao L, Liu J, Li Z, Dai X, Li S. Role of the JAK-STAT pathway in proliferation and differentiation of human hypertrophic scar fibroblasts induced by connective tissue growth factor. Mol Med Report. 2010;3(6):941–5.
    1. Tieu BC, Lee C, Sun H, Lejeune W, Recinos A, III, Ju X, Spratt H, Guo DC, Milewicz D, Tilton RG, Brasier AR. An adventitial IL-6/MCP1 amplification loop accelerates macrophage-mediated vascular inflammation leading to aortic dissection in mice. J Clin Invest. 2009;119:3637–3651.
    1. Timofeeva OA, Gaponenko V, Lockett SJ, Tarasov SG, Jiang S, Michejda CJ, Perantoni AO, Tarasova NI. Rationally designed inhibitors identify STAT3 N-domain as a promising anticancer drug target. ACS Chem Biol. 2007;2:799–809.
    1. van Zuijlen PP, Vloemans JF, van Trier AJ, Suijker MH, van UE, Groenevelt F, Kreis RW, Middelkoop E. Dermal substitution in acute burns and reconstructive surgery: a subjective and objective long-term follow-up. Plast Reconstr Surg. 2001;108:1938–1946.
    1. Vermes C, Jacobs JJ, Zhang J, Firneisz G, Roebuck KA, Glant TT. Shedding of the interleukin-6 (IL-6) receptor (gp80) determines the ability of IL-6 to induce gp130 phosphorylation in human osteoblasts. J Biol Chem. 2002;277:16879–16887.
    1. Yamauchi-Takihara K, Kishimoto T. A role for STAT3 in cardiac remodeling. Trends Cardiovasc Med. 2000;10:298–303.
    1. Zhang Z, Finnerty CC, He J, Herndon DN. Smad ubiquitination regulatory factor 2 expression is enhanced in hypertrophic scar fibroblasts from burned children. Burns. 2011
    1. Zunwen L, Shizhen Z, Dewu L, Yungui M, Pu N. Effect of tetrandrine on the TGF-β- induced smad signal transduction pathway in human hypertrophic scar fibroblasts in vitro. Burns. 2012;38(3):404–413.

Source: PubMed

3
订阅