当前位置: 首页 > 期刊 > 《动脉硬化血栓血管生物学》 > 2005年第1期 > 正文
编号:11275471
The ADAMTS Proteases, Extracellular Matrix, and Vascular Disease
http://www.100md.com 《动脉硬化血栓血管生物学》
     From The Hope Heart Program at the Benaroya Research Institute at Virginia Mason, Seattle, Wash.

    Correspondence to Thomas N. Wight, PhD, Hope Heart Program, Benaroya Research Institute at Virginia Mason, 1124 Columbia Street, Seattle, WA 98104-2046. E-mail twight@hopeheart.org

    The importance of proteases as mediators of extracellular matrix (ECM) degradation and vascular cell phenotype in the pathogenesis of vascular disease is indisputable. In fact, excellent cases can be made for the matrix metalloproteinases (MMPs) (see review1), the serine proteases (see review2), and the cysteine and aspartic proteases (see review3) being involved in many of the events in vascular disease. In this issue of Arteriosclerosis, Thrombosis, and Vascular Biology, Jonsson-Rylander and her colleagues show us that ADAMTS1, a member of another family of proteases, is also involved.4 Descendants from the ADAM family of proteases, the ADAMTS members (a disintegrin-like and metalloproteinase with thrombospondin type 1 motifs), currently numbering 19, evolved as nonintegral membrane proteins that associate with the cell surface and ECM through specific protein domains (see reviews5,6). Like the ADAM family, the ADAMTS proteases are multidomain proteins with common structural motifs that include an N-terminal signal sequence, a prodomain, a catalytic domain with Zn binding site, an ancillary domain, a disintegrin-like domain, a central thrombospondin repeat domain (TSR), a cysteine rich domain, a cysteine free spacer domain, and usually one or more TSRs. Some of the ADAMTS members also contain unique C-terminal domains that can contain PLAC and/or CUB sequences. These proteases are synthesized as zymogens that undergo processing by convertases such as furin and by metalloproteinases such as MT-4 MMP.7,8 Like the MMPs, the ADAMTS members can be inhibited by tissue inhibitors of matrix metalloproteinases or TIMPs. For example TIMP-3 is a potent inhibitor of ADAMTS4 and 5.9 Other inhibitors exist as well. For example, C-terminal truncation of ADAMTS4 can be blocked by TIMP-1.7 Proteins that share structural homology to the ADAMTS members also may function as ADAMTS inhibitors. One example is papilin, a protein produced by the fruit fly Drosophila melanogaster which shares a set of homologous domains with ADAMTS members including the ancillary domain but excluding the catalytic domain. Papilin is capable of interacting with ADAMTS2 and inhibiting the activity of this protease.10 BLAST searches reveal homologous but not identical proteins in mouse. In fact, other mammalian proteins that appear to be similar to the invertebrate papilin have recently been found and termed punctin 1 or ADAMTSL-1 and punctin 2 or ADAMTSL-3.11,12 The name punctin derives from its punctate distribution associated with the cells and within the ECM.11 Whether these proteins influence ADAMTS activity remains to be shown.

    See page 180

    The roles of the different ADAMTS family members are just starting to be uncovered.5,6 However, considerable attention has focused on ADAMTS1, the founding member of this family and one that is induced by inflammatory mediators such as lipopolysaccharide and tumor necrosis factor alpha.13 Perhaps one of the most dramatic roles for ADAMTS1 and associated ADAMTS members is their involvement in the degradation of the ECM in cartilage leading to inflammation and arthritis (see review14). In 1991, Sandy et al reported that when bovine articular cartilage is treated with interleukin-1, the principle proteoglycan in cartilage, aggrecan, is cleaved at Glu373-Ala374 and not where the MMPs are known to cleave.15 This activity was identified as distinct from the activity of the MMPs and given the name aggrecanase.16 This activity was later identified by cloning to be caused by ADAMTS4 and ADAMTS5.17,18 Later studies showed ADAMTS1 to be an aggrecanase as well.19,20 These early studies also identified at least 4 other sites in the aggrecan molecule that are cleaved by ADAMTS1, 4, 5.20–23

    Aggrecan is found mostly in cartilage and is a member of a gene family of proteoglycans that share the property of interacting with hyaluronan in a specific fashion so as to form high molecular weight aggregates that resist compressive forces and entrap water.24 Versican is a member of this gene family and bears high resemblance to aggrecan.25 A number of years ago, we identified versican as a major chondroitin sulfate proteoglycan present in developing blood vessels26 and synthesized by arterial smooth muscle cells.27–29 Furthermore, a number of studies over the past few years, reviewed by Wight and Merrilees30 have shown versican to be involved in various aspects of vascular lesion development and prominent in different lesion types including early and late atherosclerotic plaques, restenotic lesions, lesions arising during graft repair, and in aneurysmal lesions. Furthermore, because both aggrecan and versican have highly homologous N-terminal and C-terminal domains, we wondered whether the aggrecanases could work as versicanases as well. We found that human aorta contains a 70-kDa fragment of versican whose cleavage site is similar to the known N-terminal cleavage site of aggrecan.31 Furthermore, this fragment was present in thickened layers of human intimas and in human aortic explants. Using purified aortic versican, we found that recombinant ADAMTS1 and 4 were capable of generating this 70-kDa fragment, indicating that these proteases were indeed "versicanases" as well. In a more recent study, we found that this 70-kDA fragment of versican was increased in a graft repair model when the graft was subjected to high blood flow, indicating that activity of the ADAMTS proteases can be regulated by shear stress in blood vessels.32 The report of Jonsson-Rylander et al in this issue extends these observations and highlights the importance of this protease in the pathogenesis of atherosclerosis.4 ADAMTS1 mRNA transcript is shown to be abundant in human aorta and increases as arterial smooth muscle cells migrate and proliferate in vitro (Figure). Furthermore, the authors show that this enzyme localizes to smooth muscle cells and macrophages as well as endothelial cells in vascular lesions suggesting multiple cellular sources for this enzyme. Evidence for multiple cellular sources for this enzyme is supported by studies that show that ADAMTS1 is also produced by endothelial cells in response to inflammatory stimuli.33 In addition, ADAMTS1 appears to inhibit neovascularization34 through its ability to sequester vascular endothelial growth factor and limit the bioavailability of this angiogenic factor.35 Furthermore, Jonsson-Rylander et al show that apoE-deficient mice crossed to Adamts1 overexpressing mice develop enhanced intimal thickening when compared with apoE-deficient only, suggesting that Adamts1 contributes to lesion expansion, possibly through its effects on arterial smooth muscle cell proliferation and migration.4 Finally, evidence is presented to indicate that ADAMTS1 is capable of cleaving versican at more than 1 site as has been described for aggrecan.20

    Schematic model of ADAMTS1 being deposited in the ECM and associated with the surface of vascular cells including endothelial cells (EC), smooth muscle cells (SMC), and monocyte/macrophages (M). One substrate identified for ADAMTS1 is versican, a chondroitin sulfate proteoglycan that accumulates in the ECM in different vascular lesions. ADAMTS1 is known to cleave versican at multiple sites and generate fragments of versican that may possess bioactivity and in turn influence the phenotype of the endothelial and smooth muscle cells (white arrows) and possibly the macrophages as well. Given the multiple ways versican has been shown to influence the events associated with vascular lesion development, a key question to be resolved is whether intact versican or versican fragments are more proatherosclerotic.

    As with any good study, more questions are generated than possibly can be answered at this time. For example, it will be important to further define the nature of the intimal thickenings in the ADAMTS1 overexpressing mice; are more cells involved and of what type? Is the protease regulated differently in different proinflammatory–proatherosclerotic conditions? How do the levels of active versus inactive enzyme compare in these conditions? Is ADAMTS1 activity elevated during atherosclerotic lesion formation? Are other ADAMTS family members involved? Are there natural inhibitors of ADAMTS1 activity present in blood vessels, and do they influence atherosclerotic lesion development? What is the nature of the breakdown products of versican and do they have biological activity? Indeed, is the phenotypic change associated with ADAMTS1 expression actually due to versican degradation or to some other substrate? What other substrates exist for ADAMTS1? The list can go on and on. However, what is clear, given the importance of versican in human atherosclerosis, is that the involvement of specific enzymes that degrade versican in diseased blood vessels, induced by inflammatory stimuli, needs further attention. These studies also should alert us to considering the biological importance of specific ECM degradation products regulating key events in the pathogenesis of vascular disease. There is some precedent to think that parts of the versican molecule can exhibit specific bioactivity. For example, Burton Yang’s group in Toronto has been overexpressing parts of the versican molecule using versican minigenes and finding interesting phenotypic changes in cells. For example, expressing only the G3 carboxy-terminal region of the versican molecule stimulates the proliferation of NIH 3T3 cells.36 Furthermore, this group has shown that the C-terminal fragment of versican is capable of binding to cell surface integrins influencing growth and cell survival.37,38 Interestingly, one of the spliced variants of versican, V3, contains only the N-terminal and C-terminal parts of the molecule and completely lacks the protein domain that carries the GAG chains and the ADAMTS cleavage sites. This versican variant has a dramatic effect on the adhesive, proliferative, and migratory capacities of arterial smooth muscle cells and the capacity of these cells to assemble an elastin rich ECM.39,40 Whether versican fragments generated by ADAMTS1 proteolytic activity affect events associated with the development of atherosclerosis will have to await further study. The group in Sweden, however, has made an excellent start!

    Acknowledgments

    This manuscript was supported in part by a grant from the National Institutes of Health HK 18645.

    References

    Galis ZS, Khatri JJ. Matrix metalloproteinases in vascular remodeling and atherogenesis: the good, the bad, and the ugly. Circ Res. 2002; 90: 251–262.

    Luttun A, Carmeliet P. Genetic studies on the role of proteinases and growth factors in atherosclerosis and aneurysm formation. Ann N Y Acad Sci. 2001; 947: 124–132;discussion 132–123.

    Liu J, Sukhova GK, Sun JS, Xu WH, Libby P, Shi GP. Lysosomal cysteine proteases in atherosclerosis. Arterioscler Thromb Vasc Biol. 2004; 24: 1359–1366.

    J?nsson-Rylander A-C, Nilsson T, Fritsche-Danielson R, Hammarstr?m A, Behrendt M, Andersson JO, Lindgren K, Andersson A-K, Wallbrandt P, Rosengren B, Brodin P, Thelin A, Westin A, Hurt-Camejo E, Lee-S?gaard C-H. Role of ADAMTS-1 in atherosclerosis: remodeling of carotid artery, immunohistochemistry, and proteolysis of versican. Arterioscler Thromb Vasc Biol. 2005; 25: 180–185.

    Tang BL. ADAMTS. a novel family of extracellular matrix proteases. Int J Biochem Cell Biol. 2001; 33: 33–44.

    Apte SS. A disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motifs: the ADAMTS family. Int J Biochem Cell Biol. 2004; 36: 981–985.

    Gao G, Westling J, Thompson VP, Howell TD, Gottschall PE, Sandy JD. Activation of the proteolytic activity of ADAMTS4 (aggrecanase-1) by C-terminal truncation. J Biol Chem. 2002; 277: 11034–11041.

    Gao G, Plaas A, Thompson VP, Jin S, Zuo F, Sandy JD. ADAMTS4 (aggrecanase-1) activation on the cell surface involves C-terminal cleavage by glycosylphosphatidyl inositol-anchored membrane type 4-matrix metalloproteinase and binding of the activated proteinase to chondroitin sulfate and heparan sulfate on syndecan-1. J Biol Chem. 2004; 279: 10042–10051.

    Kashiwagi M, Tortorella M, Nagase H, Brew K. TIMP-3 is a potent inhibitor of aggrecanase 1 (ADAM-TS4) and aggrecanase 2 (ADAM-TS5). J Biol Chem. 2001; 276: 12501–12504.

    Kramerova IA, Kawaguchi N, Fessler LI, Nelson RE, Chen Y, Kramerov AA, Kusche-Gullberg M, Kramer JM, Ackley BD, Sieron AL, Prockop DJ, Fessler JH. Papilin in development; a pericellular protein with a homology to the ADAMTS metalloproteinases. Development. 2000; 127: 5475–5485.

    Hirohata S, Wang LW, Miyagi M, Yan L, Seldin MF, Keene DR, Crabb JW, Apte SS. Punctin, a novel ADAMTS-like molecule, ADAMTSL-1, in extracellular matrix. J Biol Chem. 2002; 277: 12182–12189.

    Hall NG, Klenotic P, Anand-Apte B, Apte SS. ADAMTSL-3/punctin-2, a novel glycoprotein in extracellular matrix related to the ADAMTS family of metalloproteases. Matrix Biol. 2003; 22: 501–510.

    Kuno K, Kanada N, Nakashima E, Fujiki F, Ichimura F, Matsushima K. Molecular cloning of a gene encoding a new type of metalloproteinase-disintegrin family protein with thrombospondin motifs as an inflammation associated gene. J Biol Chem. 1997; 272: 556–562.

    Nagase H, Kashiwagi M. Aggrecanases and cartilage matrix degradation. Arthritis Res Ther. 2003; 5: 94–103.

    Sandy JD, Neame PJ, Boynton RE, Flannery CR. Catabolism of aggrecan in cartilage explants. Identification of a major cleavage site within the interglobular domain. J Biol Chem. 1991; 266: 8683–8685.

    Plaas AN, Sandy JD. A cartilage explant system for studies of aggrecan structure, biosynthesis, and metabolism in discrete zones of the mammalian growth plate. Matrix. 1993; 13: 135–147.

    Tortorella MD, Burn TC, Pratta MA, Abbaszade I, Hollis JM, Liu R, Rosenfeld SA, Copeland RA, Decicco CP, Wynn R, Rockwell A, Yang F, Duke JL, Solomon K, George H, Bruckner R, Nagase H, Itoh Y, Ellis DM, Ross H, Wiswall BH, Murphy K, Hillman MC Jr, Hollis GF, Arner EC, et al. Purification and cloning of aggrecanase-1: a member of the ADAMTS family of proteins. Science. 1999; 284: 1664–1666.

    Abbaszade I, Liu RQ, Yang F, Rosenfeld SA, Ross OH, Link JR, Ellis DM, Tortorella MD, Pratta MA, Hollis JM, Wynn R, Duke JL, George HJ, Hillman MC, Jr., Murphy K, Wiswall BH, Copeland RA, Decicco CP, Bruckner R, Nagase H, Itoh Y, Newton RC, Magolda RL, Trzaskos JM, Burn TC. Cloning and characterization of ADAMTS11, an aggrecanase from the ADAMTS family. J Biol Chem. 1999; 274: 23443–23450.

    Kuno K, Okada Y, Kawashima H, Nakamura H, Miyasaka M, Ohno H, Matsushima K. ADAMTS-1 cleaves a cartilage proteoglycan, aggrecan. FEBS Lett. 2000; 478: 241–245.

    Rodriguez-Manzaneque JC, Westling J, Thai SN, Luque A, Knauper V, Murphy G, Sandy JD, Iruela-Arispe ML. ADAMTS1 cleaves aggrecan at multiple sites and is differentially inhibited by metalloproteinase inhibitors. Biochem Biophys Res Commun. 2002; 293: 501–508.

    Tortorella MD, Pratta M, Liu RQ, Austin J, Ross OH, Abbaszade I, Burn T, Arner E. Sites of aggrecan cleavage by recombinant human aggrecanase-1 (ADAMTS-4). J Biol Chem. 2000; 275: 18566–18573.

    Tortorella MD, Liu RQ, Burn T, Newton RC, Arner E. Characterization of human aggrecanase 2 (ADAM-TS5): substrate specificity studies and comparison with aggrecanase 1 (ADAM-TS4). Matrix Biol. 2002; 21: 499–511.

    Vankemmelbeke MN, Holen I, Wilson AG, Ilic MZ, Handley CJ, Kelner GS, Clark M, Liu C, Maki RA, Burnett D, Buttle DJ. Expression and activity of ADAMTS-5 in synovium. Eur J Biochem. 2001; 268: 1259–1268.

    Margolis RU, Margolis RK. Aggrecan-versican-neurocan family proteoglycans. Methods Enzymol. 1994; 245: 105–126.

    Zimmermann D. Versican. In: Iozzo R, ed. Proteoglycans-Structure, Biology and Molecular Interactions. New York: Marcel Dekker, Inc; 2000.

    Yao LY, Moody C, Sch?nherr E, Wight TN, Sandell LJ. Identification of the proteoglycan versican in aorta and smooth muscle cells by DNA sequence analysis, in situ hybridization and immunohistochemistry. Matrix Biol. 1994; 14: 213–225.

    Chang Y, Yanagishita M, Hascall VC, Wight TN. Proteoglycans synthesized by smooth muscle cells derived from monkey (Macaca nemestrina) aorta. J Biol Chem. 1983; 258: 5679–5688.

    Sch?nherr E, J?rvel?inen HT, Sandell LJ, Wight TN. Effects of platelet-derived growth factor and transforming growth factor-beta 1 on the synthesis of a large versican-like chondroitin sulfate proteoglycan by arterial smooth muscle cells. J Biol Chem. 1991; 266: 17640–17647.

    Sch?nherr E, Kinsella MG, Wight TN. Genistein selectively inhibits platelet-derived growth factor stimulated versican biosynthesis in monkey arterial smooth muscle cells. Arch Biochem Biophys. 1997; 339: 353–361.

    Wight TN, Merrilees MJ. Proteoglycans in atherosclerosis and restenosis: key roles for versican. Circ Res. 2004; 94: 1158–1167.

    Sandy JD, Westling J, Kenagy RD, Iruela-Arispe ML, Verscharen C, Rodriguez-Mazaneque JC, Zimmermann DR, Lemire JM, Fischer JW, Wight TN, Clowes AW. Versican V1 proteolysis in human aorta in vivo occurs at the Glu441-Ala442 bond, a site that is cleaved by recombinant ADAMTS-1 and ADAMTS-4. J Biol Chem. 2001; 276: 13372–13378.

    Kenagy R, Fischer J, Lara S, Sandy JD, Clowes AW, Wight TN. Accumulation and loss of extracellular matrix during shear stress mediated intimal growth and regression in baboon vascular grafts. J Histochem Cytochem. In Press.

    Norata GD, Bjork H, Hamsten A, Catapano AL, Eriksson P. High-density lipoprotein subfraction 3 decreases ADAMTS-1 expression induced by lipopolysaccharide and tumor necrosis factor-alpha in human endothelial cells. Matrix Biol. 2004; 22: 557–560.

    Vazquez F, Hastings G, Ortega MA, Lane TF, Oikemus S, Lombardo M, Iruela-Arispe ML. METH-1, a human ortholog of ADAMTS-1, and METH-2 are members of a new family of proteins with angio-inhibitory activity. J Biol Chem. 1999; 274: 23349–23357.

    Luque A, Carpizo DR, Iruela-Arispe ML. ADAMTS1/METH1 inhibits endothelial cell proliferation by direct binding and sequestration of VEGF165. J Biol Chem. 2003; 278: 23656–23665.

    Yang BL, Zhang Y, Cao L, Yang BB. Cell adhesion and proliferation mediated through the G1 domain of versican. J Cell Biochem. 1999; 72: 210–220.

    Wu Y, Chen L, Zheng PS, Yang BB. beta 1-Integrin-mediated glioma cell adhesion and free radical-induced apoptosis are regulated by binding to a C-terminal domain of PG-M/versican. J Biol Chem. 2002; 277: 12294–12301.

    Wu Y, Chen L, Cao L, Sheng W, Yang BB. Overexpression of the C-terminal PG-M/versican domain impairs growth of tumor cells by intervening in the interaction between epidermal growth factor receptor and beta1-integrin. J Cell Sci. 2004; 117: 2227–2237.

    Lemire JM, Merrilees MJ, Braun KR, Wight TN. Overexpression of the V3 variant of versican alters arterial smooth muscle cell adhesion, migration, and proliferation in vitro. J Cell Physiol. 2002; 190: 38–45.

    Merrilees MJ, Lemire JM, Fischer JW, Kinsella MG, Braun KR, Clowes AW, Wight TN. Retrovirally mediated overexpression of versican v3 by arterial smooth muscle cells induces tropoelastin synthesis and elastic fiber formation in vitro and in neointima after vascular injury. Circ Res. 2002; 90: 481–487.(Thomas N. Wight)