Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Chondromodulin-I maintains cardiac valvular function by preventing angiogenesis

Abstract

The avascularity of cardiac valves is abrogated in several valvular heart diseases (VHDs). This study investigated the molecular mechanisms underlying valvular avascularity and its correlation with VHD. Chondromodulin-I, an antiangiogenic factor isolated from cartilage, is abundantly expressed in cardiac valves. Gene targeting of chondromodulin-I resulted in enhanced Vegf-A expression, angiogenesis, lipid deposition and calcification in the cardiac valves of aged mice. Echocardiography showed aortic valve thickening, calcification and turbulent flow, indicative of early changes in aortic stenosis. Conditioned medium obtained from cultured valvular interstitial cells strongly inhibited tube formation and mobilization of endothelial cells and induced their apoptosis; these effects were partially inhibited by chondromodulin-I small interfering RNA. In human VHD, including cases associated with infective endocarditis, rheumatic heart disease and atherosclerosis, VEGF-A expression, neovascularization and calcification were observed in areas of chondromodulin-I downregulation. These findings provide evidence that chondromodulin-I has a pivotal role in maintaining valvular normal function by preventing angiogenesis that may lead to VHD.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Immunohistochemistry and immunofluorescence staining of Chm-I protein in developing and adult mouse heart.
Figure 2: Abnormal angiogenesis, inflammatory cell infiltration and mineralization in cardiac valves of aged Chm1−/− mice.
Figure 3: Echocardiography of Chm1−/− mouse hearts.
Figure 4: Immunohistochemistry, immunofluorescence staining and in situ hybridization of Chm-I and Vegf-A in sclerotic lesions of aged Apoe−/− mouse hearts.
Figure 5: Expression of Chm-I in VICs and its effect on tube formation, migration and apoptosis in HCAECs.
Figure 6: Histology and immunohistochemistry of cardiac valves from human autopsies and surgical samples.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Hammon, J.W., Jr ., O'Sullivan, M.J., Oury, J. & Fosburg, R.G. Allograft cardiac valves. A view through the scanning electron microscope. J. Thorac. Cardiovasc. Surg. 68, 352–360 (1974).

    PubMed  Google Scholar 

  2. Soini, Y., Salo, T. & Satta, J. Angiogenesis is involved in the pathogenesis of nonrheumatic aortic valve stenosis. Hum. Pathol. 34, 756–763 (2003).

    Article  CAS  Google Scholar 

  3. Yamauchi, R. et al. Upregulation of SR-PSOX/CXCL16 and recruitment of CD8+ T cells in cardiac valves during inflammatory valvular heart disease. Arterioscler. Thromb. Vasc. Biol. 24, 282–287 (2004).

    Article  CAS  Google Scholar 

  4. Filip, D.A., Radu, A. & Simionescu, M. Interstitial cells of the heart valves possess characteristics similar to smooth muscle cells. Circ. Res. 59, 310–320 (1986).

    Article  CAS  Google Scholar 

  5. Lester, W., Rosenthal, A., Granton, B. & Gotlieb, A.I. Porcine mitral valve interstitial cells in culture. Lab. Invest. 59, 710–719 (1988).

    CAS  PubMed  Google Scholar 

  6. Gotlieb, A.I., Rosenthal, A. & Kazemian, P. Fibroblast growth factor 2 regulation of mitral valve interstitial cell repair in vitro. J. Thorac. Cardiovasc. Surg. 124, 591–597 (2002).

    Article  CAS  Google Scholar 

  7. Akiyama, H. et al. Essential role of Sox9 in the pathway that controls formation of cardiac valves and septa. Proc. Natl. Acad. Sci. USA 101, 6502–6507 (2004).

    Article  CAS  Google Scholar 

  8. Ranger, A.M. et al. The transcription factor NF-ATc is essential for cardiac valve formation. Nature 392, 186–190 (1998).

    Article  CAS  Google Scholar 

  9. Rajamannan, N.M. et al. Human aortic valve calcification is associated with an osteoblast phenotype. Circulation 107, 2181–2184 (2003).

    Article  Google Scholar 

  10. Chan-Thomas, P.S., Thompson, R.P., Robert, B., Yacoub, M.H. & Barton, P.J. Expression of homeobox genes Msx-1 (Hox-7) and Msx-2 (Hox-8) during cardiac development in the chick. Dev. Dyn. 197, 203–216 (1993).

    Article  CAS  Google Scholar 

  11. Sugi, Y., Yamamura, H., Okagawa, H. & Markwald, R.R. Bone morphogenetic protein-2 can mediate myocardial regulation of atrioventricular cushion mesenchymal cell formation in mice. Dev. Biol. 269, 505–518 (2004).

    Article  CAS  Google Scholar 

  12. Camenisch, T.D. et al. Temporal and distinct TGFbeta ligand requirements during mouse and avian endocardial cushion morphogenesis. Dev. Biol. 248, 170–181 (2002).

    Article  CAS  Google Scholar 

  13. Ikeda, T. et al. The combination of SOX5, SOX6, and SOX9 (the SOX trio) provides signals sufficient for induction of permanent cartilage. Arthritis Rheum. 50, 3561–3573 (2004).

    Article  CAS  Google Scholar 

  14. Hiraki, Y. [Molecular cloning of a novel cartilage-specific functional matrix, chondromodulin-I, and its role in endochondral bone formation]. Seikagaku 63, 1449–1454 (1991).

    CAS  PubMed  Google Scholar 

  15. Funaki, H. et al. Expression and localization of angiogenic inhibitory factor, chondromodulin-I, in adult rat eye. Invest. Ophthalmol. Vis. Sci. 42, 1193–1200 (2001).

    CAS  PubMed  Google Scholar 

  16. Azizan, A., Holaday, N. & Neame, P.J. Post-translational processing of bovine chondromodulin-I. J. Biol. Chem. 276, 23632–23638 (2001).

    Article  CAS  Google Scholar 

  17. Hiraki, Y. et al. Identification of chondromodulin I as a novel endothelial cell growth inhibitor. Purification and its localization in the avascular zone of epiphyseal cartilage. J. Biol. Chem. 272, 32419–32426 (1997).

    Article  CAS  Google Scholar 

  18. Inoue, H., Kondo, J., Koike, T., Shukunami, C. & Hiraki, Y. Identification of an autocrine chondrocyte colony-stimulating factor: chondromodulin-I stimulates the colony formation of growth plate chondrocytes in agarose culture. Biochem. Biophys. Res. Commun. 241, 395–400 (1997).

    Article  CAS  Google Scholar 

  19. Shukunami, C. & Hiraki, Y. Expression of cartilage-specific functional matrix chondromodulin-I mRNA in rabbit growth plate chondrocytes and its responsiveness to growth stimuli in vitro. Biochem. Biophys. Res. Commun. 249, 885–890 (1998).

    Article  CAS  Google Scholar 

  20. Hiraki, Y., Kono, T., Sato, M., Shukunami, C. & Kondo, J. Inhibition of DNA synthesis and tube morphogenesis of cultured vascular endothelial cells by chondromodulin-I. FEBS Lett. 415, 321–324 (1997).

    Article  CAS  Google Scholar 

  21. Zacks, S. et al. Characterization of Cobblestone mitral valve interstitial cells. Arch. Pathol. Lab. Med. 115, 774–779 (1991).

    CAS  PubMed  Google Scholar 

  22. Oshima, Y. et al. Expression and localization of tenomodulin, a transmembrane type chondromodulin-I-related angiogenesis inhibitor, in mouse eyes. Invest. Ophthalmol. Vis. Sci. 44, 1814–1823 (2003).

    Article  Google Scholar 

  23. Hiraki, Y. et al. Molecular cloning of human chondromodulin-I, a cartilage-derived growth modulating factor, and its expression in Chinese hamster ovary cells. Eur. J. Biochem. 260, 869–878 (1999).

    Article  CAS  Google Scholar 

  24. Shukunami, C., Iyama, K., Inoue, H. & Hiraki, Y. Spatiotemporal pattern of the mouse chondromodulin-I gene expression and its regulatory role in vascular invasion into cartilage during endochondral bone formation. Int. J. Dev. Biol. 43, 39–49 (1999).

    CAS  PubMed  Google Scholar 

  25. Shukunami, C., Yamamoto, S., Tanabe, T. & Hiraki, Y. Generation of multiple transcripts from the chicken chondromodulin-I gene and their expression during embryonic development. FEBS Lett. 456, 165–170 (1999).

    Article  CAS  Google Scholar 

  26. Dietz, U.H., Ziegelmeier, G., Bittner, K., Bruckner, P. & Balling, R. Spatio-temporal distribution of chondromodulin-I mRNA in the chicken embryo: expression during cartilage development and formation of the heart and eye. Dev. Dyn. 216, 233–243 (1999).

    Article  CAS  Google Scholar 

  27. Sachdev, S.W. et al. Sequence analysis of zebrafish chondromodulin-1 and expression profile in the notochord and chondrogenic regions during cartilage morphogenesis. Mech. Dev. 105, 157–162 (2001).

    Article  CAS  Google Scholar 

  28. Chalajour, F. et al. Angiogenic activation of valvular endothelial cells in aortic valve stenosis. Exp. Cell Res. 298, 455–464 (2004).

    Article  CAS  Google Scholar 

  29. Takeda, S., Bonnamy, J.P., Owen, M.J., Ducy, P. & Karsenty, G. Continuous expression of Cbfa1 in nonhypertrophic chondrocytes uncovers its ability to induce hypertrophic chondrocyte differentiation and partially rescues Cbfa1-deficient mice. Genes Dev. 15, 467–481 (2001).

    Article  CAS  Google Scholar 

  30. Dawson, D.W. et al. Pigment epithelium-derived factor: a potent inhibitor of angiogenesis. Science 285, 245–248 (1999).

    Article  CAS  Google Scholar 

  31. Shworak, N.W. Angiogenic modulators in valve development and disease: does valvular disease recapitulate developmental signaling pathways? Curr. Opin. Cardiol. 19, 140–146 (2004).

    Article  Google Scholar 

  32. Mohler, E.R., III . et al. Bone formation and inflammation in cardiac valves. Circulation 103, 1522–1528 (2001).

    Article  Google Scholar 

  33. Mazzone, A. et al. Neoangiogenesis, T-lymphocyte infiltration, and heat shock protein-60 are biological hallmarks of an immunomediated inflammatory process in end-stage calcified aortic valve stenosis. J. Am. Coll. Cardiol. 43, 1670–1676 (2004).

    Article  CAS  Google Scholar 

  34. O'Brien, K.D., McDonald, T.O., Chait, A., Allen, M.D. & Alpers, C.E. Neovascular expression of E-selectin, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 in human atherosclerosis and their relation to intimal leukocyte content. Circulation 93, 672–682 (1996).

    Article  CAS  Google Scholar 

  35. Moulton, K.S. et al. Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis. Proc. Natl. Acad. Sci. USA 100, 4736–4741 (2003).

    Article  CAS  Google Scholar 

  36. Otto, C.M., Kuusisto, J., Reichenbach, D.D., Gown, A.M. & O'Brien, K.D. Characterization of the early lesion of 'degenerative' valvular aortic stenosis. Histological and immunohistochemical studies. Circulation 90, 844–853 (1994).

    Article  CAS  Google Scholar 

  37. O'Brien, K.D. et al. Osteopontin is expressed in human aortic valvular lesions. Circulation 92, 2163–2168 (1995).

    Article  CAS  Google Scholar 

  38. Agmon, Y. et al. Aortic valve sclerosis and aortic atherosclerosis: different manifestations of the same disease? Insights from a population-based study. J. Am. Coll. Cardiol. 38, 827–834 (2001).

    Article  CAS  Google Scholar 

  39. O'Brien, K.D. et al. Association of angiotensin-converting enzyme with low-density lipoprotein in aortic valvular lesions and in human plasma. Circulation 106, 2224–2230 (2002).

    Article  CAS  Google Scholar 

  40. Pohle, K. et al. Association of cardiovascular risk factors to aortic valve calcification as quantified by electron beam computed tomography. Mayo Clin. Proc. 79, 1242–1246 (2004).

    Article  Google Scholar 

  41. Rabkin, E. et al. Activated interstitial myofibroblasts express catabolic enzymes and mediate matrix remodeling in myxomatous heart valves. Circulation 104, 2525–2532 (2001).

    Article  CAS  Google Scholar 

  42. Fondard, O. et al. Extracellular matrix remodelling in human aortic valve disease: the role of matrix metalloproteinases and their tissue inhibitors. Eur. Heart J. 26, 1333–1341 (2005).

    Article  CAS  Google Scholar 

  43. Enomoto, H. et al. Vascular endothelial growth factor isoforms and their receptors are expressed in human osteoarthritic cartilage. Am. J. Pathol. 162, 171–181 (2003).

    Article  CAS  Google Scholar 

  44. Fujimoto, N. & Iwata, K. Use of EIA to measure MMPs and TIMPs. Methods Mol. Biol. 151, 347–358 (2001).

    CAS  PubMed  Google Scholar 

  45. Tanaka, K. et al. Age-associated aortic stenosis in apolipoprotein E-deficient mice. J. Am. Coll. Cardiol. 46, 134–141 (2005).

    Article  CAS  Google Scholar 

  46. Nakamichi, Y. et al. Chondromodulin I is a bone remodeling factor. Mol. Cell. Biol. 23, 636–644 (2003).

    Article  CAS  Google Scholar 

  47. Kyuwa, S., Xiao, Y., Toyoda, Y. & Sato, E. Characterization of embryonic stem-like cell lines derived from embryoid bodies. Exp. Anim. 46, 11–16 (1997).

    Article  CAS  Google Scholar 

  48. Litwin, S.E., Katz, S.E., Morgan, J.P. & Douglas, P.S. Serial echocardiographic assessment of left ventricular geometry and function after large myocardial infarction in the rat. Circulation 89, 345–354 (1994).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Y. Nishizaki and S. Kondo for technical support. This study was supported in part by research grants from the Ministry of Education, Science and Culture, Japan, and by the Program for Promotion of Fundamental Studies in Health Science of the National Institute of Biomedical Innovation.

Author information

Authors and Affiliations

Authors

Contributions

This study was designed by K.F.; experiments were performed by M.Y., S.Y., K.M., K.K. and N.K.; the Chm1 knockout mouse and antibody to Chm-I were made by C.S. and Y.H.; surgical specimens were collected by M.M., H.S. and R.Y.; the MMP experiment was performed by T.S. and Y.O.; specimens from Apoe knockout mice were distributed by M.S.; S.O. contributed to the writing of the paper.

Corresponding author

Correspondence to Keiichi Fukuda.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

Temporal and spatial expression of Chm1 in rodent and human heart (PDF 2002 kb)

Supplementary Fig. 2

Immunofluorescent staining for Chm-I in middle- and late-stage embryonic mouse heart. (PDF 287 kb)

Supplementary Fig. 3

Antiangiogenic effect of recombinant human CHM-I on HCAECs in vitro. (PDF 224 kb)

Supplementary Fig. 4

MTT assay for VICs. (PDF 982 kb)

Supplementary Fig. 5

Cbfa1-positive cells were detected in vascular areas in VHD. (PDF 189 kb)

Supplementary Fig. 6

Immunohistochemistry of the cardiac valves for MMP-1, -2, -3, -9 and -13 in humans with various VHDs. (PDF 450 kb)

Supplementary Fig. 7

Activated myofibroblasts were detected in vascular areas in VHD. (PDF 279 kb)

Supplementary Fig. 8

The conceptual framework of the role of Chm-I in cardiac valves. (PDF 1790 kb)

Supplementary Methods (PDF 137 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yoshioka, M., Yuasa, S., Matsumura, K. et al. Chondromodulin-I maintains cardiac valvular function by preventing angiogenesis. Nat Med 12, 1151–1159 (2006). https://doi.org/10.1038/nm1476

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm1476

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing