Thromb Haemost 2009; 101(04): 656-664
DOI: 10.1160/TH08-12-0779
Theme Issue Article
Schattauer GmbH

Serine proteases, inhibitors and receptors in renal fibrosis

Allison A. Eddy
1   Seattle Children’s Hospital Research Institute, Center for Tissue and Cell Sciences, and Department of Pediatrics, University of Washington, Seattle, Washington, USA
› Author Affiliations
Financial support:The author acknowledges support from the National Institutes of Health
Further Information

Publication History

Received: 01 December 2008

Accepted after minor revision: 16 January 2009

Publication Date:
23 November 2017 (online)

Summary

Chronic kidney disease (CKD) is estimated to affect one in eight adults. Their kidney function progressively deteriorates as inflammatory and fibrotic processes damage nephrons. New therapies to prevent renal functional decline must build on basic research studies that identify critical cellular and molecular mediators. Plasminogen activator inhibitor-1 (PAI-1), a potent fibrosis-promoting glycoprotein, is one promising candidate. Absent from normal kidneys, PAI-1 is frequently expressed in injured kidneys. Studies in genetically engineered mice have demonstrated its potency as a pro-fibrotic molecule. Somewhat surprising, its ability to inhibit serine protease activity does not appear to be its primary pro-fibrotic effect in CKD. Both tissue-type plasminogen activator and plasminogen deficiency significantly reduced renal fibrosis severity after ureteral obstruction, while genetic urokinase (uPA) deficiency had no effect. PAI-1 expression is associated with enhanced recruitment of key cellular effectors of renal fibrosis – interstitial macrophages and myofibroblasts. The ability of PAI-1 to promote cell migration involves interactions with the low-density lipoprotein receptor-associate protein-1 and also complex interactions with uPA bound to its receptor (uPAR) and several leukocyte and matrix integrins that associate with uPAR as co-receptors. uPAR is expressed by several cell types in damaged kidneys, and studies in uPAR-deficient mice have shown that its serves a protective role. uPAR mediates additional anti-fibrotic effects – it interacts with specific co-receptors to degrade PAI-1 and extracellular collagens, and soluble uPAR has leukocyte chemoattractant properties. Molecular pathways activated by serine proteases and their inhibitor, PAI-1, are promising targets for future anti-fibrotic therapeutic agents.

 
  • References

  • 1 Coresh J, Selvin E, Stevens LA. et al. Prevalence of chronic kidney disease in the United States. J Am Med Assoc 2007; 298: 2038-2047.
  • 2 Eddy AA. Progression in chronic kidney disease. Adv Chronic Kidney Dis 2005; 12: 353-365.
  • 3 Collins AJ, Foley R, Herzog C. et al. Excerpts from the United States Renal Data System 2007 annual data report. Am J Kidney Dis 2008; 51: S1-320.
  • 4 Neugarten J, Gallo G, Silbiger S. et al. Glomerulosclerosis in aging humans is not influenced by gender. Am J Kidney Dis 1999; 34: 884-888.
  • 5 Levey A, Coresh J. Part 4. Definition and classification of stages of chronic kideny disease. Am J Kidney Dis 2002; 39: S46-S75.
  • 6 Eddy AA, Fogo AB. PAI-1 in chronic kidney disease: evidence and mechanisms of action. J Am Soc Nephrol 2006; 17: 2999-3012.
  • 7 Wang Y, Wang YP, Zheng G. et al. Ex vivo programmed macrophages ameliorate experimental chronic inflammatory renal disease. Kidney international 2007; 72: 290-299.
  • 8 Ricardo S, van Goor H, Eddy A. Macrophage diversity in renal injury and repair. J Clin Invest 2008; 118: 3522-3530.
  • 9 Nishida M, Fujinaka H, Matsusaka T. et al. Absence of angiotensin II type 1 receptor in bone marrow-derived cells is detrimental in the evolution of renal fibrosis. J Clin Invest 2002; 110: 1859-1868.
  • 10 Zhang G, Kim H, Cai X. et al. Urokinase receptor deficiency accelerates fibrosis in obstructive nephropathy. J Am Soc Nephrol 2003; 14: 1254-1271.
  • 11 Engelholm LH, List K, Netzel-Arnett S. et al. uPARAP/Endo180 is essential for cellular uptake of collagen and promotes fibroblast collagen adhesion. J Cell Biol 2003; 160: 1009-1015.
  • 12 Everts V, van der Zee E, Creemers L. et al. Phagocytosis and intracellular digestion of collagen, its role in turnover and remodelling. Histochem J 1996; 28: 229-245.
  • 13 Singh DK, Winocour P, Farrington K. Mechanisms of disease: the hypoxic tubular hypothesis of diabetic nephropathy. Nat Clin Pract Nephrol 2008; 4: 216-226.
  • 14 Nangaku M. Chronic hypoxia and tubulointerstitial injury: A final common pathway to end-stage renal failure. J Am Soc Nephrol 2006; 17: 17-25.
  • 15 Zandi-Nejad K, Eddy AA, Glassock RJ. et al. Why is proteinuria an ominous biomarker of progressive kidney disease?. Kidney Int Suppl 2004: S76-89.
  • 16 Liu Y. Epithelial to mesenchymal transition in renal fibrogenesis: pathologic significance, molecular mechanism, and therapeutic intervention. J Am Soc Nephrol 2004; 15: 1-12.
  • 17 Zeisberg M, Kalluri R. The role of epithelial-tomesenchymal transition in renal fibrosis. J Mol Med 2004; 82: 175-181.
  • 18 Theilig F, Kriz W, Jerichow T. et al. Abrogation of protein uptake through megalin-deficient proximal tubules does not safeguard against tubulointerstitial injury. J Am Soc Nephrol 2007; 18: 1824-1834.
  • 19 Okamura DM, Lopez-Guisa JM, Koelsch K. et al. Atherogenic scavenger receptor modulation in the tubulointerstitium in response to chronic renal injury. Am J Physiol 2007; 293: F575-585.
  • 20 Okamura DM, Pennathur S, Koelsch K. et al. CD36 regulates oxidative stress and inflammation in hyper-cholesterolemicchronic kidney disease. J Am Soc Nephrol. 2009 epub ahead of print.
  • 21 Dellas C, Loskutoff DJ. Historical analysis of PAI-1 from its discovery to its potential role in cell motility and disease. Thromb Haemost 2005; 93: 631-640.
  • 22 Eddy AA. Plasminogen activator inhibitor-1 and the kidney. American journal of physiology 2002; 283: F209-220.
  • 23 Reis K, Arinsoy T, Derici U. et al. Angiotensinogen and plasminogen activator inhibitor-1 gene polymorphism in relation to chronic allograft dysfunction. Clin Transplant 2005; 19: 10-14.
  • 24 Wang AY, Poon P, Lai FM. et al. Plasminogen activator inhibitor-1 gene polymorphism 4G/4G genotype and lupus nephritis in Chinese patients. Kidney Int 2001; 59: 1520-1528.
  • 25 Estelles A, Dalmau J, Falco C. et al. Plasma PAI-1 levels in obese children--effect of weight loss and influence of PAI-1 promoter 4G/5G genotype. Thromb Haemost 2001; 86: 647-652.
  • 26 Gils A, Declerck PJ. The structural basis for the pathophysiological relevance of PAI-I in cardiovascular diseases and the development of potential PAI-I inhibitors. Thromb Haemost 2004; 91: 425-437.
  • 27 Hoekstra T, Geleijnse JM, Schouten EG. et al. Plasminogen activator inhibitor-type 1: its plasma determinants and relation with cardiovascular risk. Thromb Haemost 2004; 91: 861-872.
  • 28 Oda T, Jung YO, Kim H. et al. PAI-1 deficiency attenuates the fibrogenic response to ureteral obstruction. Kidney Int 2001; 30: 587-596.
  • 29 Matsuo S, Lopez-Guisa JM, Cai X. et al. Multi-functionality of PAI-1 in fibrogenesis: evidence from obstructive nephropathy in PAI-1-overexpressing mice. Kidney Int 2005; 67: 2221-2238.
  • 30 Huang Y, Haraguchi M, Lawrence DA. et al. A mutant, noninhibitory plasminogen activator inhibitor type 1 decreases matrix accumulation in experimental glomerulonephritis. J Clin Invest 2003; 112: 379-388.
  • 31 Huang Y, Border WA, Yu L. et al. A PAI-1 mutant, PAI-1R, slows progression of diabetic nephropathy. J Am Soc Nephrol 2008; 19: 329-338.
  • 32 Montes R, Declerck PJ, Calvo A. et al. Prevention of renal fibrin deposition in endotoxin-induced DIC through inhibition of PAI-1. Thromb Haemost 2000; 84: 65-70.
  • 33 Taneda S, Hudkins KL, Muhlfeld AS. et al. Pro-tease nexin-1, tPA, and PAI-1 are upregulated in cryoglobulinemic membranoproliferative glomerulonephritis. J Am Soc Nephrol 2008; 19: 243-251.
  • 34 Moll S, Schaeren-Wiemers N, Wohlwend A. et al. Protease nexin 1 in the murine kidney: Glomerular localization and up-regulation in glomerulopathies. Kidney Int 1996; 50: 1936-1945.
  • 35 Strehlow D, Jelaska A, Strehlow K. et al. A potential role for protease nexin 1 overexpression in the pathogenesis of scleroderma. J Clin Invest 1999; 103: 1179-1190.
  • 36 Herz J, Strickland DK. LRP: a multifunctional scavenger and signaling receptor. J Clin Invest 2001; 108: 779-784.
  • 37 Zhang G, Kernan KA, Collins SJ. et al. Plasmin(ogen) promotes renal interstitial fibrosis by promoting epithelial-to-mesenchymal transition: role of plasmin-activated signals. J Am Soc Nephrol 2007; 18: 846-859.
  • 38 Romer J, Bugge TH, Pyke C. et al. Plasminogen and wound healing. Nat Med 1996; 2: 725.
  • 39 Lyons RM, Gentry LE, Purchio AF. et al. Mechanism of activation of latent recombinant transforming growth factor beta 1 by plasmin. J Cell Biol 1990; 110: 1361-1367.
  • 40 Oda T, Kim H, Wing D. et al. Effects of genetic PAI-1 deficiency in mice with protein-overload proteinuria. J Am Soc Nephrol 1999; 10: 578A [Abstract].
  • 41 Gold LI, Schwimmer R, Quigley JP. Human plasma fibronectin as a substrate for human urokinase. Biochem J 1989; 262: 529-534.
  • 42 Naldini L, Vigna E, Bardelli A. et al. Biological activation of pro-HGF (hepatocyte growth factor) by urokinase is controlled by a stoichiometric reaction. J Biol Chem 1995; 270: 603-611.
  • 43 Liu Y. Hepatocyte growth factor in kidney fibrosis: therapeutic potential and mechanisms of action. Am J Physiol 2004; 287: F7-16.
  • 44 Yamaguchi I, Lopez-Guisa JM, Cai X. et al. Endogenous urokinase lacks antifibrotic activity during progressive renal injury. Am J Physiol 2007; 293: F12-19.
  • 45 Zhang G, Eddy AA. Urokinase and its receptors in chronic kidney disease. Front Biosci 2008; 13: 5462-5478.
  • 46 Zhang G, Kernan KA, Collins SJ. et al. The novel ligand-receptor relationship identified between urokinase and the nicotinic receptor (nAChRalpha1): its signaling and function in cell growth and chronic renal injury. J Am Soc Nephrol 2007; 18: 7A.
  • 47 Yang J, Shultz RW, Mars WM. et al. Disruption of tissue-type plasminogen activator gene in mice reduces renal interstitial fibrosis in obstructive nephropathy. J Clin Invest 2002; 110: 1525-1538.
  • 48 Hu K, Wu C, Mars WM. et al. Tissue-type plasminogen activator promotes murine myofibroblast activation through LDL receptor-related protein 1-mediated integrin signaling. J Clin Invest 2007; 117: 3821-3832.
  • 49 Hu K, Lin L, Tan X. et al. tPA protects renal interstitial fibroblasts and myofibroblasts from apoptosis. J Am Soc Nephrol 2008; 19: 503-514.
  • 50 Cheng S, Lovett DH. Gelatinase A (MMP-2) is necessary and sufficient for renal tubular cell epithelial-mesenchymal transformation. Am J Pathol 2003; 162: 1937-1949.
  • 51 Cochrane AL, Kett MM, Samuel CS. et al. Renal structural and functional repair in a mouse model of reversal of ureteral obstruction. J Am Soc Nephrol 2005; 16: 3623-3630.
  • 52 Eddy A. Can renal fibrosis be reversed?. Pediatr Nephrol 2005; 20: 1369-1375.
  • 53 Kriz W, Hahnel B, Hosser H. et al. Pathways to recovery and loss of nephrons in anti-Thy-1 nephritis. J Am Soc Nephrol 2003; 14: 1904-1926.
  • 54 Ma LJ, Nakamura S, Aldigier JC. et al. Regression of glomerulosclerosis with high-dose angiotensin inhibition is linked to decreased plasminogen activator inhibitor-1. J Am Soc Nephrol 2005; 16: 966-976.
  • 55 Degryse B, Neels JG, Czekay RP. et al. The low density lipoprotein receptor-related protein is a motogenic receptor for plasminogen activator inhibitor-1. J Biol Chem 2004; 279: 22595-22604.
  • 56 Zhang G, Kim H, Cai X. et al. Urokinase receptor modulates cellular and angiogenic responses in obstructive uropathy. J Am Soc Nephrol 2003; 14: 1234-1253.
  • 57 Wei Y, Waltz DA, Rao N. et al. Identification of the urokinase receptor as an adhesion receptor for vitronectin. J Biol Chem 1994; 269: 32380-32388.
  • 58 Colman RW, Pixley RA, Najamunnisa S. et al. Binding of high molecular weight kininogen to human endothelial cells is mediated via a site within domains 2 and 3 of the urokinase receptor. J Clin Invest 1997; 100: 1481-1487.
  • 59 Binder BR, Mihaly J, Prager GW. uPAR-uPA-PAI-1 interactions and signaling: a vascular biologist’s view. Thromb Haemost 2007; 97: 336-342.
  • 60 Wei C, Moller CC, Altintas MM. et al. Modification of kidney barrier function by the urokinase receptor. Nat Med 2008; 14: 55-63.
  • 61 Roelofs JJ, Rouschop KM, Teske GJ. et al. The urokinase plasminogen activator receptor is crucially involved in host defense during acute pyelonephritis. Kidney Int 2006; 70: 1942-1947.
  • 62 Sitrin RG, Pan PM, Harper HA. et al. Clustering of urokinase receptors (uPAR; CD87) induces proinflammatory signaling in human polymorphonuclear neutrophils. J Immunol 2000; 165: 3341-3349.
  • 63 Alfano M, Mariani SA, Elia C. et al. Ligand-engaged urokinase-type plasminogen activator receptor (uPAR) and activation of the CD11b/CD18 (Mac1) integrin inhibit late events of HIV expression in monocytic cells. Blood 2009; 113: 1699-1709.
  • 64 Montuori N, Carriero MV, Salzano S. et al. The cleavage of the urokinase receptor regulates its multiple functions. J Biol Chem 2002; 277: 46932-46939.
  • 65 Resnati M, Pallavicini I, Wang JM. et al. The fibrinolytic receptor for urokinase activates the G protein-coupled chemotactic receptor FPRL1/LXA4R. Proc Natl Acad Sci USA 2002; 99: 1359-1364.
  • 66 Sitrin RG, Pan PM, Blackwood RA. et al. Cutting edge: evidence for a signaling partnership between urokinase receptors (CD87) and L-selectin (CD62L) in human polymorphonuclear neutrophils. J Immunol 2001; 166: 4822-4825.
  • 67 Xue W, Kindzelskii AL, Todd RF. 3rd, et al. Physical association of complement receptor type 3 and urokinase-type plasminogen activator receptor in neutrophil membranes. J Immunol 1994; 152: 4630-4640.
  • 68 Shushakova N, Eden G, Dangers M. et al. The urokinase/urokinase receptor system mediates the IgG immune complex-induced inflammation in lung. J Immunol 2005; 175: 4060-4068.
  • 69 Shushakova N, Tkachuk N, Dangers M. et al. Urokinase-induced activation of the gp130/Tyk2/Stat3 pathway mediates a pro-inflammatory effect in human mesangial cells via expression of the anaphylatoxin C5a receptor. J Cell Sci 2005; 118: 2743-2753.
  • 70 Kjoller L. The urokinase plasminogen activator receptor in the regulation of the actin cytoskeleton and cell motility. Biol Chem 2002; 383: 5-19.
  • 71 Stefansson S, Lawrence DA. Old dogs and new tricks: proteases, inhibitors, and cell migration. Sci STKE 2003; 2003: pe24.
  • 72 Mazzieri R, Blasi F. The urokinase receptor and the regulation of cell proliferation. Thromb Haemost 2005; 93: 641-646.
  • 73 Behrendt N, Ronne E, Dano K. A novel, specific pro-urokinase complex on monocyte-like cells, detected by transglutaminase-catalyzed cross-linking. FEBS letters 1993; 336: 394-396.
  • 74 López-Guisa JM, Bugge TH, Isacke CH. et al. Endo180/uPAR-associated protein is an important regular of renal fibrogenesis. J Am Soc Nephrol 2008; 19: 32A (Abstract).
  • 75 Kjoller L, Engelholm LH, Hoyer-Hansen M. et al. uPARAP/endo180 directs lysosomal delivery and degradation of collagen IV. Exp Cell Res 2004; 293: 106-116.
  • 76 Wienke D, MacFadyen JR, Isacke CM. Identification and characterization of the endocytic transmembrane glycoprotein endo180 as a novel collagen receptor. Mol Biol Cell 2003; 14: 3592-3604.
  • 77 East L, McCarthy A, Wienke D. et al. A targeted deletion in the endocytic receptor gene Endo180 results in a defect in collagen uptake. EMBO Rep 2003; 4: 710-716.
  • 78 Kjaergaard M, Gardsvoll H, Hirschberg D. et al. Solution structure of recombinant somatomedin B domain from vitronectin produced in Pichia pastoris. Protein Sci 2007; 16: 1934-1945.
  • 79 Cao DJ, Guo YL, Colman RW. Urokinase-type plasminogen activator receptor is involved in mediating the apoptotic effect of cleaved high molecular weight kininogen in human endothelial cells. Circulation research 2004; 94: 1227-1234.
  • 80 Khan MM, Bradford HN, Isordia-Salas I. et al. High-molecular-weight kininogen fragments stimulate the secretion of cytokines and chemokines through uPAR, Mac-1, and gC1qR in monocytes. Arterioscl Thromb Vasc Biol 2006; 26: 2260-2266.
  • 81 Colman RW, Jameson BA, Lin Y. et al. Domain 5 of high molecular weight kininogen (kininostatin) down-regulates endothelial cell proliferation and migration and inhibits angiogenesis. Blood 2000; 95: 543-550.
  • 82 Schanstra JP, Neau E, Drogoz P. et al. In vivo bradykinin B2 receptor activation reduces renal fibrosis. J Clin Invest 2002; 110: 371-379.
  • 83 Okada H, Watanabe Y, Kikuta T. et al. Bradykinin decreases plasminogen activator inhibitor-1 expression and facilitates matrix degradation in the renal tubulointerstitium under angiotensin-converting enzyme blockade. J Am Soc Nephrol 2004; 15: 2404-2413.
  • 84 Azizi M, Wuerzner G. Rationale for combining blockers of the renin-angiotensin system. Seminars Nephrol 2007; 27: 544-554.
  • 85 Liang OD, Chavakis T, Linder M. et al. Binding of urokinase plasminogen activator to gp130 via a putative urokinase-binding consensus sequence. Biol Chem 2003; 384: 229-236.
  • 86 Zhang G, Cai X, Lopez-Guisa JM. et al. Mitogenic signaling of urokinase receptor-deficient kidney fibroblasts: actions of an alternative urokinase receptor and LDL receptor-related protein. J Am Soc Nephrol 2004; 15: 2090-2102.