Skip to main content
Log in

Dysregulation of renal MMP-3 and MMP-7 in canine X-linked Alport syndrome

  • Original Article
  • Published:
Pediatric Nephrology Aims and scope Submit manuscript

Abstract

Matrix metalloproteinases (MMPs) play an important regulatory role in many biological and pathological processes and their specific role in Alport syndrome (AS) is not yet clearly defined. In this study, the naturally occurring canine X-linked AS was used to demonstrate a potential role for MMP-3 and MMP-7 in Alport renal pathogenesis. Recently, we demonstrated that the expression of MMP-2, MMP-9 and MMP-14 was upregulated in the renal cortex of dogs with a spontaneous form of XLAS. In the present study, we examined necropsy samples of renal cortex from normal and XLAS dogs for MMP-3 and MMP-7 as they have the potential to activate MMP-2 and MMP-9. Immunohistochemical analysis showed strong immunostaining for both MMP-3 and MMP-7 in the interstitial space of XLAS kidneys, while virtually no immunostaining was observed in similar fields from normal dogs. RT-PCR and casein zymography confirmed that both mRNA transcripts and activities of MMP-3 and MMP-7 are elevated in XLAS kidneys. The induction of these MMPs likely contributes to tissue destruction associated with the fibrogenic process, while augmenting the activation of MMP-2 and MMP-9 by MMP-3 and MMP-7 in XLAS. Thus, these data further implicate a role for the MMPs in progressive renal pathogenesis associated with AS.

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.

Fig. 1A–F
Fig. 2A,B
Fig. 3A–C
Fig. 4A–C
Fig. 5 A
Fig. 6 A

Similar content being viewed by others

References

  1. Pescucci C, Longo I, Bruttini M, Mari F, Renieri A (2003) Type-IV collagen related diseases. J Nephrol 16:314–316

    Google Scholar 

  2. Kashtan CE (1999) Alport syndrome: An inherited disorder of renal, ocular and cochlear basement membrane. Medicine (Baltimore) 78:338–360

    Google Scholar 

  3. Gross O, Schulze-Lohoff E, Koepke ML, Beirowski B, Addicks K, Bloch W, Smyth N, Weber M (2004) Antifibrotic, nephroprotective potential of ACE inhibitor vs AT antagonist in a murine model of renal fibrosis. Nephrol Dial Transplant 19:1716–1723

    Google Scholar 

  4. Chen D, Jefferson B, Harvey SJ, Zheng K, Gartlay CJ, Jacobs RM, Thorner PS (2003) Cyclosporine a slows the progressive renal disease of Alport syndrome (X-linked hereditary nephritis): results from a canine model. J Am Soc Nephrol 14:14:690–698

    Google Scholar 

  5. Harvey SJ, Zheng K, Jefferson B, Moak P, Sado Y, Naito I, Ninomiya Y, Jacobs R, Thorner PS (2003) Transfer of the α5(IV) collagen chain gene to smooth muscle restores in vivo expression of the α6(IV) collagen chain in a canine model of Alport syndrome. Am J Pathol 162:873–885

    Google Scholar 

  6. Mochizuki T, Lemmink H, Mariyama M, Antignac C, Gubler M-C, Pirson Y, Verellen-Dumoulin C, Chan B, Schroder C, Smeets H, Reeders S (1994) Identification of mutations in the α3(IV) and α4(IV) collagen genes in autosomal recessive Alport syndrome. Nat Genet 8:77–82

    Google Scholar 

  7. Miner JH, Sanes JR (1994) Collagen IV α3, α4, and α5 chains in rodent basal lamina: Sequence, distribution, association with laminins, and developmental switches. J Cell Biol 127:879–891

    Google Scholar 

  8. Kalluri R, Shield CF, Todd P, Hudson BG, Neilson EG (1997) Isoform switching of type IV collagen is developmentally arrested in X-linked Alport syndrome leading to increased susceptibility of renal basement membranes to endoproteolysis. J Clin Invest 15:2470–2478

    Google Scholar 

  9. Cosgrove D, Meehan DT, Grunkemeyer JA, Kornak JM, Sayers R, Hunter WJ, Samuelson GC (1996) Collagen COL4A3 knockout: A mouse model for autosomal Alport syndrome. Genes Dev 10:2981–2992

    Google Scholar 

  10. Lees G, Helman RG, Kashtan CE, Michael AF, Homco LD, Millichamp NJ, Camacho ZT, Templeton JW, Ninomiya Y, Sado Y, Kim Y (1999) A new form of X-linked dominant hereditary nephritis in dogs. Am J Vet Res 60:373–383

    Google Scholar 

  11. Rheault MN, Kren SM, Thielen BK, Mess HA, Crossen JT, Thomas W, Sado Y, Kashtan CE, Segal Y (2004) Mouse model of X-linked Alport syndrome. J Am Soc Nephrol 15:1466–1474

    Google Scholar 

  12. Fine LG, Ong AC, Norman JT (1993) Mechanisms of tubulo-interstitial injury in progressive renal diseases. Eur J Clin Invest 23:259–265

    Google Scholar 

  13. Smeglin A, Frishman WH (2004) Elastolytic matrix metalloproteinases and their inhibitors as therapeutic targets in atherosclerotic plaque instability. Cardiol Rev 12:141–150

    Google Scholar 

  14. Lopez B, Gonzalez A, Diez J (2004) Role of matrix metalloproteinases in hypertension-associated cardiac fibrosis. Curr Opin Nephrol Hypertension 13:197–204

    Google Scholar 

  15. Noel A, Maillard C, Rocks N, Jost M, Chabottaux V, Sounni NE, Maquoi E, Cataldo D, Foidart JM (2004) Membrane associated proteases and their inhibitors in tumor angiogenesis. J Clin Pathol 57:577–584

    Google Scholar 

  16. Rao VH, Lees GE, Kashten CE, Singh RE, Meehan DT, Rodgers K, Berridge BR, Bhattacharya G, Cosgrove D (2003) Increased expression of MMP-2, MMP-9 (type IV collagenases/gelatinases) and MT1-MMP in canine X-linked Alport syndrome (XLAS). Kidney Int 63:1736–1748

    Google Scholar 

  17. Rodgers KD, Rao V, Meehan DT, Fager N, Gotwals P, Ryan ST, Koteliansky V, Nemori R, Cosgrove D (2003) Monocytes may promote myofibroblast accumulation and apoptosis in Alport renal fibrosis. Kidney Int 63:1338–1355

    Google Scholar 

  18. Klahr S, Schreiner G, Ichikawa I (1998) The progression of renal disease. N Engl J Med 318:1657–1666

    Google Scholar 

  19. Bruijin JA, Hogendoorn PCW, Hoedenmaeker PJ, Fleuren GJ (1998) The extracellular matrix in pathology. J Lab Clin Med 111:140–149

    Google Scholar 

  20. Sternlicht MD, Werb Z (2000) How matrix metalloproteinases regulate cell behavior. Ann Rev Cell Dev Biol 17:463–516

    Google Scholar 

  21. Murphy G, Knauper V, Atkinson S, Gavrilovic J, Edwards D (2000) Cellular mechanisms for focal proteolysis and the regulation of microenvironment. Fibrinolysis Proteolysis 14:165–174

    Google Scholar 

  22. Ries C, Petrides PD (1995) Cytokine regulation of matrix metalloproteinases activity and its regulatory dysfunction in disease. Biol Chem Hoppe Seyler 376:345–355

    Google Scholar 

  23. Liotta LA, Kohn E (2004) Anoikis: Cancer and the homeless cell. Nature 430:973–974

    Google Scholar 

  24. Martin SS, Vuori K (2004) Regulation of Bcl-2 proteins during anoikis and amorphosis. Biochim Biophys Acta 1694:145–157

    Google Scholar 

  25. Sires UI, Murphy G, Baragi VM, Flizar CJ, Welgus HG, Senior RM (1994) Matrilysin is much more efficient than other metalloproteinases in the proteolytic inactivation of α-1 antitrypsin. Biochem Biophys Res Commun 204:613–620

    Google Scholar 

  26. Wilson CL, Matrician ML (1998) Matrilysin. In: Parks WC, Mecham RP (eds) Metalloproteinases. Academic Press, San Diego, pp 149–184

  27. Murphy G, Cockett MI, Ward RV, Docherty AJ (1991) Matrix metalloproteinase degradation of elastin, type IV collagen and proteoglycan. A quantitative comparison of the activities of 95 kDa gelatinases, stromelysins-1 and -2 and punctuated metalloproteinase (PUMP). Biochem J 277:277–279

    Google Scholar 

  28. Ramos-DeSimone N, Hahn-Dantona E, Sipley J, Nagase H, French Dl, Quigley JP (1999) Activation of matrix metalloproteinase-9 (MMP-9) via a converging plasmin/stromelysin-1 cascade enhances tumor cell invasion. J Biol Chem 274:13066–13076

    Google Scholar 

  29. Knauper V, Wilhelm SM, Seperack PK, DeClerck YA, Langley KE, Osthues A, Tschesche H (1993) Direct activation of human neutrophil procollagenase by recombinant stromelysin. Biochem J 295:581–586

    Google Scholar 

  30. Miyazaki K, Umenishi F, Funahashi K, Koshikawa N, Yasumitsu H, Umeda M (1992) Activation of TIMP-2/progelatinase A complex by stromelysin. Biochem Biophys Res Commun 185:852–859

    Google Scholar 

  31. Imai K, Yokohama Y, Nakanishi I, Ohuchi E, Fujii Y, Nakai N, Okada Y (1995) Matrix metalloproteinase 7 (matrilysin) from human rectal carcinoma cells. Activation of the precursor, interaction with other matrix metalloproteinases and enzymic properties. J Biol Chem 270:6691–6697

    Google Scholar 

  32. Barille S, Bataille R, Rapp MJ, Harasseau JL, Amiot M (1999) Production of metalloproteinase-7 (matrilysin) by human myeloma cells and its potential involvement in metalloproteinase-2 activation. J Immunol 163:5723–5728

    Google Scholar 

  33. Crabbe T, Smith B, O’Connell J, Docherty A (1994) Human progelatinase A can be activated by matrilysin. FEBS Lett 345:14–16

    Google Scholar 

  34. Cosgrove DE, Rodgers KD, Meehan DT, Miller C, Bovard K, Gilroy A, Gardner H, Kotelianski V, Gotwals P, Amatucci A, Kalluri R (2000) Integrin α1β1 and TGF-β1 play distinct roles in Alport glomerular pathogenesis and serve as dual targets for metabolic therapy. Am J Pathol 157:1649–1659

    Google Scholar 

Download references

Acknowledgements

This research was supported in part by NIH R01 DK55000 to D.C., NIH R01 DK57676 to C.E.K., and the tobacco settlement fund from the State of Nebraska. We thank Q. Kong for her help in preparing the histograms and statistical analysis, and Rene Rodgers for sequence analysis. We also thank John (Skip) Kennedy for help in figure preparation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dominic Cosgrove.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rao, V.H., Lees, G.E., Kashtan, C.E. et al. Dysregulation of renal MMP-3 and MMP-7 in canine X-linked Alport syndrome. Pediatr Nephrol 20, 732–739 (2005). https://doi.org/10.1007/s00467-004-1805-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00467-004-1805-5

Keywords

Navigation