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:

Regulation of cardiovascular development and integrity by the heart of glass–cerebral cavernous malformation protein pathway

A Corrigendum to this article was published on 01 May 2009

This article has been updated

Abstract

Cerebral cavernous malformations (CCMs) are human vascular malformations caused by mutations in three genes of unknown function: KRIT1, CCM2 and PDCD10. Here we show that the heart of glass (HEG1) receptor, which in zebrafish has been linked to ccm gene function, is selectively expressed in endothelial cells. Heg1−/− mice showed defective integrity of the heart, blood vessels and lymphatic vessels. Heg1−/−; Ccm2lacZ/+ and Ccm2lacZ/lacZ mice had more severe cardiovascular defects and died early in development owing to a failure of nascent endothelial cells to associate into patent vessels. This endothelial cell phenotype was shared by zebrafish embryos deficient in heg, krit1 or ccm2 and reproduced in CCM2-deficient human endothelial cells in vitro. Defects in the hearts of zebrafish lacking heg or ccm2, in the aortas of early mouse embryos lacking CCM2 and in the lymphatic vessels of neonatal mice lacking HEG1 were associated with abnormal endothelial cell junctions like those observed in human CCMs. Biochemical and cellular imaging analyses identified a cell-autonomous pathway in which the HEG1 receptor couples to KRIT1 at these cell junctions. This study identifies HEG1-CCM protein signaling as a crucial regulator of heart and vessel formation and integrity.

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: Heart and blood vessel integrity defects in HEG1-deficient mouse embryos and neonates.
Figure 2: Lymphatic vessel dilatation and leakage in Heg1−/− neonatal mice.
Figure 3: Heg1−/−; Ccm2lacZ/+ mouse embryos do not establish a patent blood vascular network.
Figure 4: Endothelial cells of zebrafish lacking heg or ccm2 form vessels that are normally patterned but not patent.
Figure 5: HEG1 is required to form normal endothelial junctions in vivo.
Figure 6: HEG1 receptor intracellular tails associate with CCM2 through KRIT1.

Similar content being viewed by others

Change history

  • 27 January 2009

    In the version of the supplementary information for this article originally posted online, the descriptions of Supplementary Videos 1–6 were incorrect. The errors have been corrected as of 27 January 2009.

  • 12 February 2009

    In this version of the article initially published, Shawn M. Sweeney was not included in the list of authors. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Rigamonti, D. et al. Cerebral cavernous malformations. Incidence and familial occurrence. N. Engl. J. Med. 319, 343–347 (1988).

    Article  CAS  Google Scholar 

  2. Revencu, N. & Vikkula, M. Cerebral cavernous malformation: new molecular and clinical insights. J. Med. Genet. 43, 716–721 (2006).

    Article  CAS  Google Scholar 

  3. Sahoo, T. et al. Mutations in the gene encoding KRIT1, a Krev-1/rap1a binding protein, cause cerebral cavernous malformations (CCM1). Hum. Mol. Genet. 8, 2325–2333 (1999).

    Article  CAS  Google Scholar 

  4. Eerola, I. et al. KRIT1 is mutated in hyperkeratotic cutaneous capillary-venous malformation associated with cerebral capillary malformation. Hum. Mol. Genet. 9, 1351–1355 (2000).

    Article  CAS  Google Scholar 

  5. Liquori, C.L. et al. Mutations in a gene encoding a novel protein containing a phosphotyrosine-binding domain cause type 2 cerebral cavernous malformations. Am. J. Hum. Genet. 73, 1459–1464 (2003).

    Article  CAS  Google Scholar 

  6. Denier, C. et al. Mutations within the MGC4607 gene cause cerebral cavernous malformations. Am. J. Hum. Genet. 74, 326–337 (2004).

    Article  CAS  Google Scholar 

  7. Bergametti, F. et al. Mutations within the programmed cell death 10 gene cause cerebral cavernous malformations. Am. J. Hum. Genet. 76, 42–51 (2005).

    Article  CAS  Google Scholar 

  8. Zawistowski, J.S. et al. CCM1 and CCM2 protein interactions in cell signaling: implications for cerebral cavernous malformations pathogenesis. Hum. Mol. Genet. 14, 2521–2531 (2005).

    Article  CAS  Google Scholar 

  9. Voss, K. et al. CCM3 interacts with CCM2 indicating common pathogenesis for cerebral cavernous malformations. Neurogenetics 8, 249–256 (2007).

    Article  CAS  Google Scholar 

  10. Zhang, J., Rigamonti, D., Dietz, H.C. & Clatterbuck, R.E. Interaction between krit1 and malcavernin: implications for the pathogenesis of cerebral cavernous malformations. Neurosurgery 60, 353–359 (2007).

    Article  Google Scholar 

  11. Mably, J.D., Mohideen, M.A., Burns, C.G., Chen, J.N. & Fishman, M.C. Heart of glass regulates the concentric growth of the heart in zebrafish. Curr. Biol. 13, 2138–2147 (2003).

    Article  CAS  Google Scholar 

  12. Mably, J.D. et al. Santa and valentine pattern concentric growth of cardiac myocardium in the zebrafish. Development 133, 3139–3146 (2006).

    Article  CAS  Google Scholar 

  13. Whitehead, K.J., Plummer, N.W., Adams, J.A., Marchuk, D.A. & Li, D.Y. Ccm1 is required for arterial morphogenesis: implications for the etiology of human cavernous malformations. Development 131, 1437–1448 (2004).

    Article  CAS  Google Scholar 

  14. Plummer, N.W. et al. Neuronal expression of the Ccm2 gene in a new mouse model of cerebral cavernous malformations. Mamm. Genome 17, 119–128 (2006).

    Article  CAS  Google Scholar 

  15. Wong, J.H., Awad, I.A. & Kim, J.H. Ultrastructural pathological features of cerebrovascular malformations: a preliminary report. Neurosurgery 46, 1454–1459 (2000).

    Article  CAS  Google Scholar 

  16. Clatterbuck, R.E., Eberhart, C.G., Crain, B.J. & Rigamonti, D. Ultrastructural and immunocytochemical evidence that an incompetent blood-brain barrier is related to the pathophysiology of cavernous malformations. J. Neurol. Neurosurg. Psychiatry 71, 188–192 (2001).

    Article  CAS  Google Scholar 

  17. Tu, J., Stoodley, M.A., Morgan, M.K. & Storer, K.P. Ultrastructural characteristics of hemorrhagic, nonhemorrhagic, and recurrent cavernous malformations. J. Neurosurg. 103, 903–909 (2005).

    Article  Google Scholar 

  18. Glading, A., Han, J., Stockton, R.A. & Ginsberg, M.H. KRIT-1/CCM1 is a Rap1 effector that regulates endothelial cell cell junctions. J. Cell Biol. 179, 247–254 (2007).

    Article  CAS  Google Scholar 

  19. Nakatsu, M.N. et al. Angiogenic sprouting and capillary lumen formation modeled by human umbilical vein endothelial cells (HUVEC) in fibrin gels: the role of fibroblasts and Angiopoietin-1. Microvasc. Res. 66, 102–112 (2003).

    Article  CAS  Google Scholar 

  20. Folkman, J. & Haudenschild, C. Angiogenesis in vitro. Nature 288, 551–556 (1980).

    Article  CAS  Google Scholar 

  21. Jin, S.W., Beis, D., Mitchell, T., Chen, J.N. & Stainier, D.Y. Cellular and molecular analyses of vascular tube and lumen formation in zebrafish. Development 132, 5199–5209 (2005).

    Article  CAS  Google Scholar 

  22. Kamei, M. et al. Endothelial tubes assemble from intracellular vacuoles in vivo. Nature 442, 453–456 (2006).

    CAS  Google Scholar 

  23. Blum, Y. et al. Complex cell rearrangements during intersegmental vessel sprouting and vessel fusion in the zebrafish embryo. Dev. Biol. 316, 312–322 (2008).

    Article  CAS  Google Scholar 

  24. Hogan, B.M., Bussmann, J., Wolburg, H. & Schulte-Merker, S. Ccm1 cell autonomously regulates endothelial cellular morphogenesis and vascular tubulogenesis in zebrafish. Hum. Mol. Genet. 17, 2424–2432 (2008).

    Article  CAS  Google Scholar 

  25. Sebzda, E. et al. Syk and Slp-76 mutant mice reveal a cell-autonomous hematopoietic cell contribution to vascular development. Dev. Cell 11, 349–361 (2006).

    Article  CAS  Google Scholar 

  26. Lawson, N.D. & Weinstein, B.M. In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev. Biol. 248, 307–318 (2002).

    Article  CAS  Google Scholar 

  27. Jin, S.W. et al. A transgene-assisted genetic screen identifies essential regulators of vascular development in vertebrate embryos. Dev. Biol. 307, 29–42 (2007).

    Article  CAS  Google Scholar 

  28. Amsterdam, A. & Hopkins, N. Retroviral-mediated insertional mutagenesis in zebrafish. Methods Cell Biol. 77, 3–20 (2004).

    Article  CAS  Google Scholar 

  29. Weinstein, B.M., Stemple, D.L., Driever, W. & Fishman, M.C. Gridlock, a localized heritable vascular patterning defect in the zebrafish. Nat. Med. 1, 1143–1147 (1995).

    Article  CAS  Google Scholar 

  30. Lim, C.J. et al. Alpha4 integrins are type I cAMP-dependent protein kinase-anchoring proteins. Nat. Cell Biol. 9, 415–421 (2007).

    Article  CAS  Google Scholar 

  31. Pfaff, M., Liu, S., Erle, D.J. & Ginsberg, M.H. Integrin beta cytoplasmic domains differentially bind to cytoskeletal proteins. J. Biol. Chem. 273, 6104–6109 (1998).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank C. Bertozzi, C. Chen, A. Granger, J. Lee, D. Li, P. Mericko, A. Schmaier, E. Sebzda, N. Shanbhag and K. Whitehead for valuable insights; M. Pack and J. He for assistance with zebrafish studies; R. Meade for preparation of the electron microscopy samples; and T. Branson for animal husbandry. This work was supported by the Swiss National Science Foundation, a Network of Excellence grant from the European Community (M.A.) and the US National Institutes of Health grants T32 HL07439 (to B.K.), T32 HL07971 (to X.Z.), HL078784 and AR27214 (to M.G.), HL62454 (to J.K.) and HL075380 and HL095326 (to M.L.K.).

Author information

Authors and Affiliations

Authors

Contributions

B.K. designed and performed all of the studies involving mutant mouse lines, contributed to endothelial cell culture and biochemical studies and wrote the manuscript; X.Z. designed and performed most of the zebrafish and biochemical studies; J.J.L., M.C., Z.Z., S.M.S. and L.G. contributed to the biochemical studies; Y.B. and M.C. contributed to the zebrafish studies; J.J.T. performed the endothelial cell culture studies; M.L. performed the immunohistochemistry studies; D.Z. contributed to the generation of mutant mouse lines; J.K. designed the endothelial cell culture studies and wrote the manuscript; M.A. designed some of the zebrafish studies and wrote the manuscript; M.H.G. designed some of the biochemical studies and wrote the manuscript; M.L.K. contributed to the design of mouse, zebrafish, endothelial and biochemical studies and wrote the manuscript.

Corresponding author

Correspondence to Mark L Kahn.

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–12, Supplementary Tables 1 and 2, and Supplementary Methods (PDF 1210 kb)

Supplementary Movie 1

Intersegmental vessels of 24–26-h.p.f. fli1a:EGFP-cdc42 transgenic zebrafish embryos following injection of scrambled morpholino (control). The GFP-cdc42 fusion protein is expressed in endothelial cells and outlines the vacuoles that form and fuse during lumen formation. (AVI 4038 kb)

Supplementary Movie 2

Intersegmental vessels of 24–26-h.p.f. fli1a:EGFP-cdc42 transgenic zebrafish embryos following injection of morpholinos to block expression of heg. The GFP-cdc42 fusion protein is expressed in endothelial cells and outlines the vacuoles that form and fuse during lumen formation. Endothelial vacuole formation in zebrafish embryos lacking heg is similar to that observed in control embryos (Supplementary Movie 1). (AVI 1894 kb)

Supplementary Movie 3

Intersegmental vessels of 24–26-h.p.f. fli1a:EGFP-cdc42 transgenic zebrafish embryos following injection of morpholinos to block expression of ccm2. The GFP-cdc42 fusion protein is expressed in endothelial cells and outlines the vacuoles that form and fuse during lumen formation. Endothelial vacuole formation in zebrafish embryos lacking ccm2 is similar to that observed in control embryos (Supplementary Movie 1). (AVI 5678 kb)

Supplementary Movie 4

Red fluorescent quantum dots were injected into the dorsal aorta of 24–26-h.p.f. fli1a:EGFP-cdc42 transgenic zebrafish embryos following injection of scrambled morpholino (control). Patent intersegmental vessels are indicated by the presence of red fluorescent dots in the vessels. (AVI 2022 kb)

Supplementary Movie 5

Red fluorescent quantum dots were injected into the dorsal aorta of 24–26-h.p.f. fli1a:EGFP-cdc42 transgenic zebrafish embryos following injection of morpholinos to block expression of heg. The presence of red fluorescent dots in the intersegmental vessels indicates vessel patency. The presence of red fluorescent dots in the intersegmental vessels indicates that the intersegmental vessels of zebrafish embryos lacking heg are patent. (AVI 3405 kb)

Supplementary Movie 6

Red fluorescent quantum dots were injected into the dorsal aorta of 24–26-h.p.f. fli1a:EGFP-cdc42 transgenic zebrafish embryos following injection of morpholinos to block expression of ccm2. The presence of red fluorescent dots in the intersegmental vessels indicates vessel patency. The presence of red fluorescent dots in the intersegmental vessels indicates that the intersegmental vessels of zebrafish embryos lacking ccm2 are patent. (AVI 7446 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kleaveland, B., Zheng, X., Liu, J. et al. Regulation of cardiovascular development and integrity by the heart of glass–cerebral cavernous malformation protein pathway. Nat Med 15, 169–176 (2009). https://doi.org/10.1038/nm.1918

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.1918

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