Skip to main content

Advertisement

Log in

Oxidative stress and the pathogenesis of scleroderma: the Murrell’s hypothesis revisited

  • Review
  • Published:
Seminars in Immunopathology Aims and scope Submit manuscript

Abstract

Systemic sclerosis (SSc, scleroderma) is a devastating, immune-mediated, multisystem disorder characterized by microvasculature damage, circulating autoantibodies, and fibroblast activation, leading to massive fibrosis of skin, vessels, muscles, and visceral organs. Scleroderma causes disability and death as the result of end-stage organ failure. At present, no specific diagnostic nor therapeutic tools are available to handle the disease. In spite of significant effort, the etiology and pathogenesis of SSc remain obscure and, consequently, the disease outcome is unpredictable. Several years ago, Murrell suggested a unifying hypothesis linking the pathogenesis of scleroderma to the generation of a large excess of reactive oxygen species. This hypothesis has been substantiated by several reports indicating the presence of an abnormal redox state in patients with scleroderma. This review will summarize the available evidence supporting the link between free radicals and the main pathological features of scleroderma.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Jimenez AS, Derk TC (2004) Following the molecular pathway toward an understanding of the pathogenesis of systemic sclerosis. Ann Intern Med 140:37–50

    CAS  PubMed  Google Scholar 

  2. Varga J, Abraham D (2007) Systemic sclerosis: a prototypic multisystem fibrotic disorder. J Clin Invest 117:557–567

    CAS  PubMed  Google Scholar 

  3. Murrell DF (1993) A radical proposal for the pathogenesis of scleroderma. J Am Acad Dermatol 28:78–85

    CAS  PubMed  Google Scholar 

  4. Herrick AL, Rieley F, Schofield D et al (1994) Micronutrient antioxidant status in patients with primary Raynaud’s phenomenon and systemic sclerosis. J Rheumatol 21:1477–1483

    CAS  PubMed  Google Scholar 

  5. Lundberg AC, Akesson A, Akesson B (1992) Dietary intake and nutritional status in patients with systemic sclerosis. Ann Rheum Dis 51:1143–1148

    Article  CAS  PubMed  Google Scholar 

  6. Bruckdorfer KR, Hillary JB, Bunce T et al (1995) Increased susceptibility to oxidation of low-density lipoproteins isolated from patients with systemic sclerosis. Arthritis Rheum 38:1060–1067

    CAS  PubMed  Google Scholar 

  7. Iwata Y, Ogawa F, Komura K et al (2007) Autoantibody against peroxiredoxin I, an antioxidant enzyme, in patients with systemic sclerosis: possible association with oxidative stress. Rheumatology 46:790–795

    CAS  PubMed  Google Scholar 

  8. Tikly M, Channa K, Theodorou P et al (2006) Lipid peroxidation and trace elements in systemic sclerosis. Clin Rheumatol 25:320–324

    PubMed  Google Scholar 

  9. Stein CM, Tanner SB, Awad JA et al (1996) Evidence of free radical-mediated injury (isoprostane overproduction) in scleroderma. Arthritis Rheum 39:1146–1150

    CAS  PubMed  Google Scholar 

  10. Cracowski JL, Marpeau C, Carpentier PH et al (2001) Enhanced in vivo lipid peroxidation in scleroderma spectrum disorders. Arthritis Rheum 44:1143–1148

    CAS  PubMed  Google Scholar 

  11. Cracowski JL, Carpentier PH, Imbert B et al (2002) Increased urinary F2-isoprostanes in systemic sclerosis, but not in primary Raynaud’s phenomenon: effect of cold exposure. Arthritis Rheum 4:1319–1323

    Google Scholar 

  12. Volpe A, Biasi D, Caramaschi P et al (2006) Levels of F2-isoprostanes in systemic sclerosis: correlation with clinical features. Rheumatology 45:314–320

    CAS  PubMed  Google Scholar 

  13. Balbir-Gurman A, Braun-Moscovici Y, Livshitz V et al (2007) Antioxidant status after iloprost treatment in patients with Raynaud’s phenomenon secondary to systemic sclerosis. Clin Rheumatol 26:1517–1521

    PubMed  Google Scholar 

  14. Beckman JS, Beckman TW, Chen J et al (1990) Apparent hydroxyl radical production by peroxynitrite: implications for endothelial injury from nitric oxide and superoxide. Proc Natl Aca Sci U S A 87:1620–24

    CAS  Google Scholar 

  15. Clancy RM, Amin AR, Abramson SB (1998) The role of nitric oxide in inflammation and immunity. Arthritis Rheum 41:1141–1151

    CAS  PubMed  Google Scholar 

  16. Matucci CM, Kahaleh MB (2002) Beauty and the beast. The nitric oxide paradox in systemic sclerosis. Rheumatology 41:843–847

    Google Scholar 

  17. Yamamoto Y, Katayama I, Nishioka K (1998) Nitric oxide production and inducible nitric oxide synthase expression in systemic sclerosis. J Rheumatol 25:314–317

    CAS  PubMed  Google Scholar 

  18. Dooley A, Gao B, Bradley N et al (2006) Abnormal nitric oxide metabolism in systemic sclerosis: increased levels of nitrated proteins and asymmetric dimethylarginine. Rheumatology 45:676–684

    CAS  PubMed  Google Scholar 

  19. Takagi K, Kawaguchi Y, Hara M et al (2003) Serum nitric oxide (NO) levels in systemic sclerosis patients: correlation between NO levels and clinical features. Clin Exp Immunol 134:538–544

    CAS  PubMed  Google Scholar 

  20. Allanore Y, Borderie D, Hilliquin P et al (2001) Low levels of nitric oxide (NO) in systemic sclerosis: inducible NO synthase production is decreased in cultured peripheral blood monocyte/macrophage cells. Rheumatology 40:1089–1096

    CAS  PubMed  Google Scholar 

  21. Cotton A, Herrick AL, Jayson MI et al (1999) Endothelial expression of nitric oxide synthases and nitrotyrosine in systemic sclerosis. J Pathol 189:273–278

    CAS  PubMed  Google Scholar 

  22. Kharitonov SA, Cailes JB, Black CM et al (1997) Decreased nitric oxide in the exhaled air of patients with systemic sclerosis with pulmonary hypertension. Thorax 52:1051–1055

    Article  CAS  PubMed  Google Scholar 

  23. Moodley YP, Lalloo UG (2001) Exhaled nitric oxide is elevated in patients with progressive systemic sclerosis without interstitial lung disease. Chest 119:1449–1454

    CAS  PubMed  Google Scholar 

  24. Malerba M, Radaeli A, Ragnoli B et al (2007) Exhaled nitric oxide levels in systemic sclerosis with and without pulmonary involvement. Chest 132:575–580

    CAS  PubMed  Google Scholar 

  25. Tiev KP, Cabane J, Aubourg F et al (2007) Severity of scleroderma lung disease is related to alveolar concentration of nitric oxide. Eur Respir J 30:26–30

    CAS  PubMed  Google Scholar 

  26. Rolla G, Colagrande P, Scappaticci E et al (2000) Exhaled nitric oxide in systemic sclerosis: relationships with lung involvement and pulmonary hypertension. J Rheumatol 27:1693–1698

    CAS  PubMed  Google Scholar 

  27. Sambo P, Amico D, Giacomelli R et al (2001) Intravenous N-acetylcysteine for treatment of Raynaud’s phenomenon secondary to systemic sclerosis: a pilot study. J Rheumatol 28:2257–2262

    CAS  PubMed  Google Scholar 

  28. Herrick AL, Hollis S, Schofield D et al (2000) A double-blind placebo-controlled trial of antioxidant therapy in limited cutaneous systemic sclerosis. Clin Exp Rheumatol 18:349–356

    CAS  PubMed  Google Scholar 

  29. Denton CP, Bunce TD, Dorado MB et al (1999) Probucol improves symptoms and reduces lipoprotein oxidation susceptibility in patients with Raynaud’s phenomenon. Rheumatology 38:309–315

    CAS  PubMed  Google Scholar 

  30. Becker LB (2004) New concepts in reactive oxygen species and cardiovascular reperfusion physiology. Cardiovasc Res 61:461–470

    CAS  PubMed  Google Scholar 

  31. Sambo P, Jannino L, Candela M et al (1999) Monocytes of patients with systemic sclerosis (scleroderma) spontaneously release in vitro increased amounts of superoxide anion. J Invest Dermatol 112:78–84

    CAS  PubMed  Google Scholar 

  32. Cracowski JL, Kom GD, Salvat-Melis M et al (2006) Postocclusive reactive hyperemia inversely correlates with urinary 15-F2t-isoprostane levels in systemic sclerosis. Free Radic Biol Med 40:1732–1737

    CAS  PubMed  Google Scholar 

  33. Sambo P, Svegliati Baroni S, Luchetti M et al (2001) Oxidative stress in scleroderma. Maintenance of scleroderma fibroblast phenotype by the constitutive up-regulation of reactive oxygen species generation through the NADPH oxidase complex pathway. Arthritis Rheum 44:2653–2664

    CAS  PubMed  Google Scholar 

  34. Svegliati Baroni S, Cancello R, Sambo P et al (2005) PDGF and reactive oxygen species (ROS) regulate Ras protein levels in primary human fibroblasts via ERK 1/2. Amplification of ROS-ERK-Ras signalling in systemic sclerosis fibroblasts. J Biol Chem 280:36474–36482

    Google Scholar 

  35. Allanore Y, Borderie D, Périanin A et al (2005) Nifedipine protects against overproduction of superoxide anion by monocytes from patients with systemic sclerosis. Arthritis Res Ther 7:R93–R100

    CAS  PubMed  Google Scholar 

  36. Failli P, Palmieri L, D’Alfonso C et al (2002) Effect of N-acetyl-l-cysteine on peroxynitrite and superoxide anion production pf lung alveolar macrophage in systemic sclerosis. Nitric Oxide 7:277–282

    CAS  PubMed  Google Scholar 

  37. Yamamoto T, Sawada Y, Katayama I et al (1998) Increased production of nitric oxide stimulated by interleukin-1beta in peripheral blood mononuclear cells in patients with systemic sclerosis. Br J Rheumatol 37:1123–1125

    CAS  PubMed  Google Scholar 

  38. Bedard K, Krause KE (2007) The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 87:245–313

    CAS  PubMed  Google Scholar 

  39. Lassegue B, Sorescu D, Szocs K et al (2001) Novel gp91 (phox) homologues in vascular smooth muscle cells: nox1 mediates angiotensin II-induced superoxide formation and redox-sensitive signalling pathway. Circ Res 88:888–894

    CAS  PubMed  Google Scholar 

  40. Touyz RM, Chen X, Tabet F et al (2002) Expression of a functionally active gp91phox-containing neutrophil-type NAD(P)H oxidase in smooth muscle cells from human resistance arteries: regulation by angiotensin II. Circ Res 90:1205–1213

    CAS  PubMed  Google Scholar 

  41. Cucoranu I, Clempus R, Dikalova A et al (2005) NAD(P)H oxidase 4 mediates transforming growth factor-beta-1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ Res 97:900–907

    CAS  PubMed  Google Scholar 

  42. Sturrock A, Cahill B, Norman K et al (2005) Transforming growth factor beta1 induces Nox4 NAD(P)H oxidase and reactive oxygen species-dependent proliferation in human pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 290:L661–L673

    PubMed  Google Scholar 

  43. Neufeld G, Cohen T, Gengrinovitch S et al (1999) Vascular endothelial growth factor (VEGF) and its receptors. FASEB J 13:9–22

    CAS  PubMed  Google Scholar 

  44. Thannickal VJ, Fanburg BL (1995) Activation of an H2O2-generating NADH in human lung fibroblasts by transforming growth factor-beta 1. J Biol Chem 270:30334–30338

    CAS  PubMed  Google Scholar 

  45. Chapple ILC (1997) Reactive oxygen species and antioxidants in inflammatory diseases. J Clin Periodontol 24:287–296

    CAS  PubMed  Google Scholar 

  46. Datla RS, Peshavariya H, Dusting GJ et al (2007) Important role of Nox4 type NADPH oxidase in angiogenic responses in human microvascular endothelial cells in vitro. Arterioscler Thromb Vasc Biol 27:2319–2324

    CAS  PubMed  Google Scholar 

  47. Ago T, Kitazono T, Ooboshi H et al (2004) Nox4 as the major catalytic component of an endothelial NAD(P)H oxidase. Circulation 109:227–233

    CAS  PubMed  Google Scholar 

  48. Ago T, Kitazono T, Kuroda J et al (2005) NAD(P)H oxidase in rat basilar arterial endothelial cells. Stroke 36:1040–1046

    CAS  PubMed  Google Scholar 

  49. Banfi B, Malgrange B, Knisz J et al (2004) Nox3: a superoxide-generating NADPH oxidase of the inner ear. J Biol Chem 279:46065–46072

    CAS  PubMed  Google Scholar 

  50. Furst R, Brueckl C, Kuebler WM et al (2005) Atrial natriuretic peptide induces mitogen-activated protein kinase phosphatase-1 in human endothelial cells via Rac1 and NAD(P)H oxidase/Nox2-activation. Circ Res 96:43–53

    PubMed  Google Scholar 

  51. Goyal P, Weissmann N, Grimminger F et al (2004) Upregulation of NAD(P)H oxidase 1 in hypoxia activates hypoxia-inducible factor 1 via increase in reactive oxygen species. Free Radic Biol Med 36:1279–1288

    CAS  PubMed  Google Scholar 

  52. Higgings DF, Kimura K, Bernhardt WM et al (2007) Hypoxia promotes fibrogenesis in vivo via HIF-1 stimulation of epithelial-to-mesenchymal transition. J Clin Invest 117:3810–3820

    Google Scholar 

  53. Colston JT, de la Rosa SD, Strader JR et al (2005) H2O2 activates Nox4 through PLA2-dependent arachidonic acid production in adult cardiac fibroblasts. FEBS Lett 579:2533–2540

    CAS  PubMed  Google Scholar 

  54. Dhaunsi GS, Paintlia MK, Kaur J et al (2004) NADPH oxidase in human lung fibroblasts. J Biomed Sci 11:617–622

    CAS  PubMed  Google Scholar 

  55. Chamseddine AH, Miller JFJ (2003) gp91phox contributes to NADPH oxidase activity in aortic fibroblasts, but not smooth muscle cells. Am J Physiol Heart Circ Physiol 285:H2284–H2289

    CAS  PubMed  Google Scholar 

  56. Harris ML, Rosen A (2003) Autoimmunity in scleroderma: the origin, pathogenetic role and clinical significance of autoantibodies. Curr Opin Rheumatol 15:778–784

    CAS  PubMed  Google Scholar 

  57. Hasegawa M, Sato S, Fujimoto M et al (1998) Serum levels of interleukin 6 (IL-6) oncostatin M, soluble IL-6 receptor, and soluble gp130 in patients with systemic sclerosis. J Rheumatol 25:308–313

    CAS  PubMed  Google Scholar 

  58. Hasegawa M, Fujimoto M, Kikuchi K et al (1997) Elevated serum levels of interleukin 4 (IL-4), IL-10 and IL-13 in patients with systemic sclerosis. J Rheumatol 24:328–332

    CAS  PubMed  Google Scholar 

  59. Valentini G, Baroni A, Esposito K et al (2001) Peripheral blood T lymphocytes from systemic sclerosis patients show both Th1 and Th2 activation. J Clin Immunol 21:210–217

    CAS  PubMed  Google Scholar 

  60. Hesegawa M, Sato S, Ihn H et al (1999) Enhanced production of interleukin-6 (IL-6) oncostatin M and soluble IL-6 receptor by cultured peripheral blood mononuclear cells from patients with systemic sclerosis. Rheumatology 38:612–617

    Google Scholar 

  61. Mavalia C, Scaletti C, Romagnani P et al (1997) Type 2 helper T-cell predominance and high CD30 expression in systemic sclerosis. Am J Pathol 151:1751–1758

    CAS  PubMed  Google Scholar 

  62. Li-Weber M, Giaisi M, Treiber MK et al (2002) Vitamin E inhibits IL-4 gene expression in peripheral blood T cells. Eur J Immunol 32:2401–2408

    CAS  PubMed  Google Scholar 

  63. King MR, Ismail AS, Davis LS et al (2006) Oxidative stress promotes polarization of human T cell differentiation toward a T helper 2 phenotype. J Immunol 176:2765–2772

    CAS  PubMed  Google Scholar 

  64. Ishikawa H, Carrasco D, Claudio E et al (1997) Gastric hyperplasia and increased proliferative responses of lymphocytes in mice lacking the COOH-terminal ankyrin domain of NF-kappaB2. J Exp Med 186:999–1014

    CAS  PubMed  Google Scholar 

  65. Casciola-Rosen L, Wigley F, Rosen A (1997) Scleroderma autoantigens are uniquely fragmented by metal-catalyzed oxidation reactions: implication for pathogenesis. J Exp Med 185:71–79

    CAS  PubMed  Google Scholar 

  66. Obata F, Hoshino A, Toyama A (2006) Hydrogen peroxide increases interleukin-12 p40/p70 molecular ratio and induces Th2 predominant responses in mice. Scand J Immunol 63:125–130

    CAS  PubMed  Google Scholar 

  67. Peterson JD, Herzenberg LA, Vasquez K et al (1998) Glutathione levels in antigen-presenting cells modulate Th1 versus Th2 response patterns. Proc Natl Acad Sci U S A 95:3071–3076

    CAS  PubMed  Google Scholar 

  68. Jeannin P, Delneste Y, Lecoanet-Henchoz S et al (1995) Thiols decrease human interleukin (IL) 4 production and IL-4-induced immunoglobulin synthesis. J Exp Med 182:1785–1792

    CAS  PubMed  Google Scholar 

  69. Prescott RJ, Freemont AJ, Jones CJ et al (1992) Sequential dermal microvascular and perivascular changes in the development of scleroderma. J Pathol 166:255–263

    CAS  PubMed  Google Scholar 

  70. Fleischmajer R, Perlish JS, Shaw KV et al (1976) Skin capillary changes in early systemic scleroderma. Electron microscopy and “in vitro” autoradiography with tritiated thymidine. Arch Dermatol 112:1553–1557

    CAS  PubMed  Google Scholar 

  71. Fleischmajer R, Perlish JS (1980) Capillary alterations in scleroderma. J Am Acad Dermatol 2:161–170

    CAS  PubMed  Google Scholar 

  72. Mitchell RN, Libby P (2007) Vascular remodelling in transplant vasculopathy. Circ Res 100:967–978

    CAS  PubMed  Google Scholar 

  73. Kuwana M, Okazaki Y, Yasuoka H et al (2004) Defective vasculogenesis in systemic sclerosis. Lancet 364:603–610

    CAS  PubMed  Google Scholar 

  74. Sgonc R, Gruschwitz MS, Dietrich H et al (1996) Endothelial cell apoptosis is a primary pathogenetic event underlying skin lesions in avian and human scleroderma. J Clin Invest 98:785–792

    CAS  PubMed  Google Scholar 

  75. Herron GS, Luz I, Romero LI (1998) Vascular abnormalities in scleroderma. Semin Cutan Med Surg 17:12–17

    CAS  PubMed  Google Scholar 

  76. Griendling KK, Sorescu D, Ushio-Fukai M (2000) NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res 86:494–501

    CAS  PubMed  Google Scholar 

  77. Lassègue B, Sorescu D, Szöcs K et al (2001) Novel gp91 (phox) homologues in vascular smooth muscle cells: nox1 mediates angiotensin II-induced superoxide formation and redox-sensitive signaling pathways. Circ Res 88:888–894

    PubMed  Google Scholar 

  78. Marumo T, Schini-Kerth VB, Fissilthaler B et al (1997) Platelet-derived growth factor-stimulated superoxide anion production modulates activation of transcription factor NF-kappaB and expression of monocyte chemoattractant protein 1 in human aortic smooth muscle cells. Circulation 96:2361–2367

    CAS  PubMed  Google Scholar 

  79. Wang Z, Castresana MR, Newman WH (2004) Reactive oxygen species-sensitive p38 MAPK controls thrombin-induced migration of vascular smooth muscle cells. J Mol Cell Cardiol 36:49–56

    CAS  PubMed  Google Scholar 

  80. Zhang H, Schmeisser A, Garlichs CD et al (1999) Angiotensin II-induced superoxide anion generation in human vascular endothelial cells: role of membrane-bound NADH-/NADPH-oxidases. Cardiovasc Res 44:215–222

    CAS  PubMed  Google Scholar 

  81. Pedruzzi E, Guichard C, Ollivier V (2004) NAD(P)H oxidase Nox-4 mediates 7-ketocholesterol-induced endoplasmic reticulum stress and apoptosis in human aortic smooth muscle cells. Mol Cell Biol 24:10703–10717

    CAS  PubMed  Google Scholar 

  82. Ushio-Fukai M, Zafari AM, Fukui T, Ishizaka N, Griendling KK (1996) p22phox is a critical component of the superoxide-generating NADH/NADPH oxidase system and regulates angiotensin II-induced hypertrophy in vascular smooth muscle cells. J Biol Chem 271:23317–23321

    CAS  PubMed  Google Scholar 

  83. Rey FE, Pagano PJ (2002) The reactive adventitia: fibroblasts oxidase in vascular function. Thromb Vasc Biol 22:1962–1971

    CAS  Google Scholar 

  84. Kawaguchi Y, Takagi K, Hara M et al (2004) Angiotensin II in the lesional skin of systemic sclerosis patients contributes to tissue fibrosis via angiotensin II type 1 receptors. Arthritis Rheum 50:216–226

    CAS  PubMed  Google Scholar 

  85. Holland JA, Meyer JW, Chang MM et al (1998) Thrombin stimulated reactive oxygen species production in cultured endothelial cells. Endothelium 6:113–121

    CAS  PubMed  Google Scholar 

  86. Bogatkevich GS, Gustio E, Oates JC et al (2004) Distinct PKC isoforms mediate cell survival and DNA synthesis in thrombin-induced myofibroblasts. Am J Physiol Lung Cell Mol Physiol 288:L190–L201

    PubMed  Google Scholar 

  87. Klareskog L, Gustaffson R, Scheynius A et al (1990) Increased expression of platelet-derived growth factor type B receptors in the skin of patients with systemic sclerosis. Arthritis Rheum 33:1534–1541

    CAS  PubMed  Google Scholar 

  88. Xue-yi Z, Jan-zhong Z, Ping T et al (1998) Expression of platelet-derived growth factor B chain and platelet-derived growth factor b-receptor in fibroblasts of scleroderma. J Dermatol Sci 18:90–97

    Google Scholar 

  89. Abraham DJ, Varga J (2005) Scleroderma: from cell and molecular mechanisms to disease models. Trends Immunol 26:587–595

    CAS  PubMed  Google Scholar 

  90. Murrell GAC, Francis MJO, Bromley L (1990) Modulation of fibroblast proliferation by oxygen free radicals. Biochem J 265:659–665

    CAS  PubMed  Google Scholar 

  91. Falanga V, Martin TA, Takagi H et al (1993) Low oxygen tension increases mRNA levels of alpha 1(I) procollagen in human dermal fibroblasts. J Cell Physiol 157:408–412

    CAS  PubMed  Google Scholar 

  92. Parola M, Pinzani M, Casini A et al (1993) Stimulation of lipid peroxidation or 4-hydroxynonenal treatment increases procollagen a(I) gene expression in human liver fat-storing cells. Biochem Biophys Res Commun 194:1044–1050

    CAS  PubMed  Google Scholar 

  93. Kinnula VL, Fattman CL, Tan RJ et al (2005) Oxidative stress in pulmonary fibrosis. A possible role for redox modulatory therapy. Am J Respir Crit Care Med 172:417–422

    PubMed  Google Scholar 

  94. Shi Y, Massagué J (2003) Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 113:685–700

    CAS  PubMed  Google Scholar 

  95. Trojanowska M (2002) Molecular aspects of scleroderma. Front Biosci 7:608–618

    Google Scholar 

  96. Bellocq A, Azoulay E, Marullo S et al (1999) Reactive oxygen species and nitrogen intermediates increase transforming growth factor-beta 1 release from human epithelial alveolar cells through two different mechanisms. Am J Respir Cell Mol Biol 21:128–136

    CAS  PubMed  Google Scholar 

  97. Barcellos-Hoff MH, Dix TA (1996) Redox-mediated activation of latent transforming growth factor beta. Mol Endocrinol 10:1077–1083

    CAS  PubMed  Google Scholar 

  98. Hancock JT, Desikan R, Neill SJ (2001) Role of reactive oxygen species in cell signalling pathways. Biochem Soc Trans 29:345–350

    CAS  PubMed  Google Scholar 

  99. Sementchenko VI, Watson DK (2000) Ets target genes: past, present and future. Oncogene 19(55):6533–6548

    CAS  PubMed  Google Scholar 

  100. Arsalane K, Dubois CM, Muanza T et al (1997) Transforming growth factor-beta 1 is a potent inhibitor of glutathione synthesis in the lung epithelial cell line A 549: transcription effect on the GSH rate-limiting enzyme gamma-glutamylcysteine synthetase. Am J Respir Cell Mol Biol 17:599–607

    CAS  PubMed  Google Scholar 

  101. Factor VM, Kiss A, Woitach JT et al (1998) Disruption of redox homeostasis in the transforming growth factor-alpha/c-myc transgenic mouse model of accelerated hepatocarcinogenesis. J Biol Chem 273:15846–15853

    CAS  PubMed  Google Scholar 

  102. Ask K, Martin GE, Kolb M et al (2006) Targeting genes for treatment in idiopathic pulmonary fibrosis: challenges and opportunities, promises and pitfalls. Proc Am Thorac Soc 3:389–393

    CAS  PubMed  Google Scholar 

  103. Hoyle GW, Li J, Finkelstein JB et al (1999) Emphysematous lesions, inflammation, and fibrosis in the lungs of transgenic mice overexpressing platelet-derived growth factor. Am J Pathol 154:1763–1775

    CAS  PubMed  Google Scholar 

  104. Bae SY, sung JY Kim OS et al (2000) Platelet-derived growth factor-induced H2O2 production requires the activation of phosphatidylinositol 3-kases. J Biol Chem 275:10527–10531

    CAS  PubMed  Google Scholar 

  105. Park J, Ha H, Ahn HJ et al (2005) Sirolimus inhibits platelet-derived growth factor-induced collagen synthesis. Transplant Proc 37:3459–3462

    CAS  PubMed  Google Scholar 

  106. Rajkumar VS, Howelll K, Csiszar K et al (2005) Shared expression of phenotypic markers in systemic sclerosis indicates convergence of pericytes and fibroblasts to a myofibroblast lineage in fibrosis. Arthritis Res Ther 7:R1113–R1123

    CAS  PubMed  Google Scholar 

  107. Distler JH, Jungel A, Huber LC et al (2007) Imatinib mesylate reduces production of extracellular matrix and prevents development of experimental dermal fibrosis. Arthritis Rheum 56:311–322

    CAS  PubMed  Google Scholar 

  108. Svegliati Baroni SS, Santillo MR, Bevilacqua F et al (2006) Stimulatory autoantibodies to the PDGF receptor in systemic sclerosis (scleroderma). N Engl J Med 354:2667–2676

    Google Scholar 

Download references

Acknowledgments

This work has been supported in part by A.I.L.S. (Associazione Italiana per la Lotta alla Sclerodermia), the Ministero Italiano per l’Università e la Ricerca Scientifica (MIUR 2006), and a generous grant from Fondazione Cariverona.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Armando Gabrielli.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Gabrielli, A., Svegliati, S., Moroncini, G. et al. Oxidative stress and the pathogenesis of scleroderma: the Murrell’s hypothesis revisited. Semin Immunopathol 30, 329–337 (2008). https://doi.org/10.1007/s00281-008-0125-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00281-008-0125-4

Keywords

Navigation