Literature
首页医源资料库在线期刊美国病理学杂志2007年第169卷第6期

Proangiogenic Cytokines as Hypoxia-Dependent Factors Stimulating Migration of Human Hepatic Stellate Cells

来源:《美国病理学杂志》
摘要:【摘要】Pathologicalangiogenesisisassociatedwiththefibrogenicprogressionofchronicliverdiseases。Experimentaldatasuggestthathypoxiaandvascularendothelialgrowthfactor(VEGF)maystimulateproliferationandsynthesisoftypeIcollageninactivated,myofibroblast-likerat......

点击显示 收起

【摘要】  Pathological angiogenesis is associated with the fibrogenic progression of chronic liver diseases. Experimental data suggest that hypoxia and vascular endothelial growth factor (VEGF) may stimulate proliferation and synthesis of type I collagen in activated, myofibroblast-like rat hepatic stellate cells (HSC/MFs). In this study, we investigated whether hypoxia, recombinant VEGF, or angiopoietin 1 (Ang-1) may affect other crucial profibrogenic features. In human HSC/MFs, which constitutively express VEGF receptor-1 and -2 (VEGFR-1, VEGFR-2) and the Ang-1 receptor Tie-2, exposure to hypoxia, VEGF, or Ang-1 resulted in a Ras/Erk-dependent stimulation of chemokinesis and chemotaxis. Migration of human HSC/MFs under hypoxic conditions involved up-regulation of VEGF-A, Ang-1, and related receptors and was mainly dependent on VEGFR-2 (Flk-1). In specimens from either cirrhotic rat livers or from patients with hepatitis C virus-related cirrhosis, HSC/MFs expressed proangiogenic factors and related receptors in areas of active fibrogenesis (ie, at the leading or lateral edge of developing incomplete fibrotic septa). Data presented herein suggest that VEGF and Ang-1 may contribute to fibrogenesis by acting as hypoxia-inducible, autocrine, and paracrine factors able to recruit myofibroblast-like cells. Moreover, HSC/MFs, in addition to their established profibrogenic role, may also contribute to neoangiogenesis during chronic hepatic wound healing.
--------------------------------------------------------------------------------
Angiogenesis is a hypoxia-stimulated and growth factor-dependent process consisting in the formation of new vascular structures from pre-existing blood vessels. Formation of new vessels is known to occur in several organs and to be critical for both growth and repair of tissues in several pathophysiological conditions.1-6 However, it has become increasingly clear that angiogenesis occurring during chronic wound healing and fibrogenesis provides a key contribution to disease progression. Pathological angiogenesis, as recently reviewed,7 has indeed been described in chronic inflammatory/fibrotic liver diseases of different etiology.
Hepatic angiogenesis differs from homologous processes in other tissues for a number of reasons, including the existence of two different types of microvascular structures in the liver (ie, large vessels lined by a continuous endothelium versus sinusoids lined by a fenestrated endothelium),8 the apparent production of the liver-specific angiogenic factor AN-GPTL3,9 and the unique but not homogenous phenotypic profile and functional role of hepatic stellate cells (HSCs).10-13 HSCs are also regarded as liver-specific pericytes, but their role in modulating angiogenesis, particularly in pathological conditions, may substantially differ from the role attributed to microcapillary pericytes.7 During the fibrotic progression of chronic liver diseases (CLDs), activated and myofibroblast-like HSCs (HSC/MFs) play a major profibrogenic role together with portal (myo)fibroblast and, possibly, bone marrow-derived stem cells, giving rise to hepatic populations of highly proliferative, profibrogenic, and contractile myofibroblast-like cells (MFs).10-16
Possible interplay and/or association between fibrogenesis and angiogenesis in CLDs is now suggested and supported by several findings: 1) angiogenesis and up-regulation of vascular endothelial growth factor (VEGF) expression has been documented in different models of acute and chronic liver injury7,17-21 as well as in specimens from human fibrotic/cirrhotic liver and hepatocellular carcinoma7,22-24 ; 2) in HSCs, hypoxia has been shown to up-regulate expression of VEGF,20,25-27 VEGF receptor type I (fms-like tyrosine kinase receptor or Flt-1),20,25 and collagen type I20 ; 3) VEGF has been proposed to directly stimulate proliferation and expression of 1(I)-procollagen mRNA in activated rat HSCs21 ; and 4) paracrine expression of VEGF by rat HSCs as well as by hepatocytes has been shown to regulate the phenotype (ie, fenestration and CD-31 expression) of liver sinusoidal endothelial cells,28 a feature of possible relevance in CLDs. Data concerning expression of angiopoietins are, at present, much more limited.7,22 Recent work has demonstrated expression of angiopoietin 1 (Ang-1) in human activated HSC/MFs and its up-regulation by hypoxia.27
In the present study, we report that VEGF-A and Ang-1 can stimulate migration and chemotaxis of human HSC/MFs and that, in liver tissue obtained either from cirrhotic rats or from patients with hepatitis C virus (HCV)-related cirrhosis, -smooth muscle actin (-SMA)-positive cells in areas of active fibrogenesis express VEGF-A and Ang-1 and their related receptors. These novel data suggest that hypoxia-dependent synthesis and release of VEGF and Ang-1 by activated HSC/MFs may contribute to both fibrogenesis and neovascularization by their actions on MF-like cells and sinusoidal endothelial cells.

【关键词】  proangiogenic cytokines hypoxia-dependent stimulating migration stellate



Materials and Methods


Materials


Enhanced chemiluminescence reagents, nitrocellulose membranes (Hybond-C extra), and secondary Cy3-conjugated antibodies were from Amersham Pharmacia Biotech (Cologno Monzese, Milano, Italy). Human recombinant growth factors and cytokines, including VEGF and Angiopoietin-1, were from PeproTech Inc. (Rocky Hill, NJ). Antibodies against phosphorylated and unphosphorylated Erk1/2 were from Upstate Biotechnology (Lake Placid, NY). Monoclonal and polyclonal antibodies were from Santa Cruz Biotechnology (Santa Cruz, CA), except those against -SMA and fluorescein isothiocyanate-conjugated antibodies (obtained from Sigma Aldrich Spa, Milano, Italy) and against CD-31 (BD Pharmingen, Erembodegem, Belgium). The monoclonal neutralizing antibody against Flk-1 was obtained from ImClone (New York, NY); although originally raised against mouse epitope, this antibody was found to also cross-react with human Flk-1, as confirmed by the block of Erk1/2 phosphorylation in human HSC/MFs treated with VEGF (data not shown). All of the other reagents were of analytical grade and obtained from Sigma Chemical Co. (Sigma Aldrich Spa).


Cell Isolation and Culture


The use of human material was approved by Human Research Review Committee of the Universit

【参考文献】
  Yancopoulos GD, Davis S, Gale N, Rudge J, Wiegand S, Holash J: Vascular specific growth factors and blood vessel formation. Nature 2000, 407:242-248

Carmeliet P: Mechanisms of angiogenesis and arteriogenesis. Nat Med 2000, 6:389-395

Ferrara N, Gerber H, LeCouter J: The biology of VEGF and its receptors. Nat Med 2003, 9:669-676

Pugh CW, Ratcliffe P: Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 2003, 9:677-684

Jain RK: Molecular regulation of vessel maturation. Nat Med 2003, 9:685-693

Carmeliet P: Angiogenesis in health and disease. Nat Med 2003, 9:653-660

Medina J, Arroyo AG, Sanchez-Madrid F, Moreno-Otero R: Angiogenesis in chronic inflammatory liver disease. Hepatology 2004, 39:1185-1195

McCuskey RS, Reilly F: Hepatic microvasculature: dynamic structure and its regulation. Semin Liver Dis 1993, 13:1-12

Camenisch G, Pisabarro M, Sherman D, Kowalski J, Nagel M, Hass P, Xie MH, Gurney A, Bodary S, Liang XH, Clark K, Beresini M, Ferrara N, Gerber HP: ANGPTL3 stimulates endothelial cell adhesion and migration via integrin vß3 and induces blood vessel formation in vivo. J Biol Chem 2002, 277:17281-17290

Friedman SL: Liver fibrosis: from bench to bedside. J Hepatol 2003, 38:S38-S53

Friedman SL: A moving target in hepatic fibrogenesis. Hepatology 2004, 40:1041-1043

Pinzani M, Marra F: Cytokine receptors and signaling in hepatic stellate cells. Semin Liver Dis 2001, 21:397-416

Cassiman D, Libbrecht L, Desmet V, Denef C, Roskams T: Hepatic stellate cell/myofibroblast subpopulations in fibrotic human and rat livers. J Hepatol 2002, 36:200-209

Bataller R, Brenner DA: Liver fibrosis. J Clin Invest 2005, 115:209-218

Ramadori G, Saile B: Mesenchymal cells in the liver??one cell type or two? Liver 2002, 22:283-294

Russo FP, Alison MR, Bigger BW, Amofah E, Florou A, Amin F, Bou-Gharios G, Jeffery R, Iredale JP, Forbes SJ: The bone marrow functionally contributes to liver fibrosis. Gastroenterology 2006, 130:1807-1821

Ankoma-Sey V, Matli M, Chang KB, Lalazar A, Donner DB, Wong L, Warren RS, Friedman SL: Coordinated induction of VEGF receptors in mesenchymal cell types during rat hepatic wound healing. Oncogene 1998, 17:115-121

Ishikawa K, Mochida S, Mashiba S, Inao M, Matsui A, Ikeda H, Ohno A, Shibuya M, Fujiwara K: Expression of vascular endothelial growth factor in nonparenchymal as well as parenchymal cells in rat liver after necrosis. Biochem Biophys Res Commun 1999, 254:587-593

Rosmorduc O, Wendum D, Corpechot C, Galy B, Sebbagh N, Raleigh J, Housset C, Poupon R: Hepatocellular hypoxia-induced vascular endothelial growth factor expression and angiogenesis in experimental biliary fibrosis. Am J Pathol 1999, 155:1065-1073

Corpechot C, Barbu V, Wendum D, Kinnman N, Rey C, Poupon R, Housset C, Rosmorduc O: Hypoxia-induced VEGF and collagen I expressions are associated with angiogenesis and fibrogenesis in experimental cirrhosis. Hepatology 2002, 35:1010-1021

Yoshiji H, Kuriyama S, Yoshii J, Ikenaka Y, Noguchi R, Hicklin DJ, Wu Y, Yanase K, Namisaki T, Yamazaki M, Tsujinoue H, Imazu H, Masaki T, Fukui H: Vascular endothelial growth factor and receptor interaction is a prerequisite for murine hepatic fibrogenesis. Gut 2003, 52:1347-1354

Medina J, Caveda L, Sanz-Cameno P, Arroyo A, Martin-Vilchez S, Majano P, Garcia-Buey L, Sanchez-Madrid F, Moreno-Otero R: Hepatocyte growth factor activates endothelial pro-angiogenic mechanisms relevant in chronic hepatitis C-associated neoangiogenesis. J Hepatol 2003, 38:660-667

Shimoda K, Mori M, Shibuta K, Banner B, Barnard G: Vascular endothelial growth factor/vascular permeability factor mRNA expression in patients with chronic hepatitis C and hepatocellular carcinoma. Int J Oncol 1999, 14:353-359

Medina J, Sanz-Cameno P, Garcia-Buey L, Martin-Vilchez S, Lopez-Cabrera M, Moreno-Otero R: Evidence of angiogenesis in primary biliary cirrhosis: an immunohistochemical descriptive study. J Hepatol 2005, 42:124-131

Ankoma-Sey V, Wang Y, Dai Z: Hypoxic stimulation of vascular endothelial growth factor expression in activated rat hepatic stellate cells. Hepatology 2000, 31:141-148

Wang YQ, Luk JM, Ikeda K, Man K, Chu AC, Kaneda K, Fan ST: Regulatory role of vHL/HIF-1 in hypoxia-induced VEGF production in hepatic stellate cells. Biochem Biophys Res Commun 2004, 317:358-362

Aleffi S, Petrai I, Bertolani C, Parola M, Colombatto S, Novo E, Vizzutti F, Anania FA, Milani S, Rombouts K, Laffi G, Pinzani M, Marra F: Upregulation of proinflammatory and proangiogenic cytokines by leptin in human hepatic stellate cells. Hepatology 2005, 42:1339-1348

DeLeve LD, Wang X, Hu L, McCuskey MK, McCuskey RS: Rat liver sinusoidal endothelial cell phenotype is maintained by paracrine and autocrine regulation. Am J Physiol 2004, 287:G757-G763

Casini A, Pinzani M, Milani S, Grappone C, Galli G, Jezequel AM, Schuppan D, Rotella CM, Surrenti C: Regulation of extracellular matrix synthesis by transforming growth factor ß1 in human fat-storing cells. Gastroenterology 1993, 105:245-253

Cassiman D, Roskams T: Beauty is in the eye of the beholder: emerging concepts and pitfalls in hepatic stellate cell research. J Hepatol 2002, 37:527-535

Failli P, De Franco RM, Caligiuri A, Gentilini A, Romanelli RG, Marra F, Batignani G, Guerra CT, Laffi G, Gentilini P, Pinzani M: Nitrovasodilators inhibit platelet-derived growth factor-induced proliferation and migration of activated human hepatic stellate cells. Gastroenterology 2000, 119:479-492

Caligiuri A, De Franco RM, Romanelli RG, Gentilini A, Meucci M, Failli P, Mazzetti L, Rombouts K, Geerts A, Vanesia M, Gentilini P, Marra F, Pinzani M: Antifibrogenic effects of canrenone, an antialdosteronic drug, on human hepatic stellate cells. Gastroenterology 2003, 124:504-520

Novo E, Marra F, Zamara E, Valfr di Bonzo L, Monitillo L, Cannito S, Petrai I, Mazzocca A, Bonacchi A, De Franco RSM, Colombatto S, Autelli R, Pinzani, Parola M: Overexpression of Bcl-2 by activated human hepatic stellate cells: resistance to apoptosis as a mechanism of progressive hepatic fibrogenesis in humans. Gut 2006, 55:1174-1182

Zamara E, Novo E, Marra F, Gentilini A, Romanelli RG, Caligiuri A, Robino G, Tamagno E, Aragno M, Danni O, Autelli R, Colombatto S, Dianzani MU, Pinzani M, Parola M: 4-Hydroxynonenal as a selective pro-fibrogenic stimulus for activated human hepatic stellate cells. J Hepatol 2004, 40:60-68

Parola M, Robino G, Marra F, Pinzani M, Bellomo G, Leonarduzzi G, Chiarugi P, Camandola S, Poli G, Gentilini P, Dianzani MU: HNE interacts directly with JNK isoforms in human hepatic stellate cells. J Clin Invest 1998, 102:1942-1950

Robino G, Parola M, Marra F, Caligiuri A, De Franco RM, Zamara E, Bellomo G, Gentilini P, Pinzani M, Dianzani MU: Interaction between 4-hydroxy-2,3-alkenals and the platelet-derived growth factor-beta receptor. Reduced tyrosine phosphorylation and downstream signaling in hepatic stellate cells. J Biol Chem 2000, 275:40561-40567

Marra F, Gentilini A, Pinzani M, Choudury GG, Parola M, Herbst H, Dianzani MU, Laffi G, Abboud HE, Gentilini P: Phosphatidylinositol 3-kinase is required for platelet-derived growth factor??s actions on hepatic stellate cells. Gastroenterology 1997, 112:1297-1306

Proctor E, Chatamra K: High yield micronodular cirrhosis in the rat. Gastroenterology 1982, 83:1183-1190

Sanso G, Aragno M, Mastrocola R, Restivo F, Mengozzi G, Smedile A, Rosina F, Danni O, Parola M, Rizzetto M: Neutral endopeptidase (EC 3.4.24.11) in cirrhotic liver: a new target to treat portal hypertension? J Hepatol 2005, 43:791-798

Novo E, Marra F, Zamara E, Valfr di Bonzo L, Caligiuri A, Cannito S, Antonaci C, Colombatto S, Pinzani M, Parola M: Dose dependent and divergent effects of superoxide anion on cell death, proliferation and migration of activated human hepatic stellate cells. Gut 2006, 55:90-97


作者单位:From the Dipartimento Medicina e Oncologia Sperimentale,* Universit

作者: Erica Novo*, Stefania Cannito*, Elena Zamara*, Lor 2008-5-29
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具