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.

  • Letter
  • Published:

Integrated genomic approaches implicate osteoglycin (Ogn) in the regulation of left ventricular mass

Abstract

Left ventricular mass (LVM) and cardiac gene expression are complex traits regulated by factors both intrinsic and extrinsic to the heart. To dissect the major determinants of LVM, we combined expression quantitative trait locus1 and quantitative trait transcript2 (QTT) analyses of the cardiac transcriptome in the rat. Using these methods and in vitro functional assays, we identified osteoglycin (Ogn) as a major candidate regulator of rat LVM, with increased Ogn protein expression associated with elevated LVM. We also applied genome-wide QTT analysis to the human heart and observed that, out of 22,000 transcripts, OGN transcript abundance had the highest correlation with LVM. We further confirmed a role for Ogn in the in vivo regulation of LVM in Ogn knockout mice. Taken together, these data implicate Ogn as a key regulator of LVM in rats, mice and humans, and suggest that Ogn modifies the hypertrophic response to extrinsic factors such as hypertension and aortic stenosis.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Quantitative trait transcripts analysis of cis eQTLs with physiological traits in the rat.
Figure 2: Candidate cis-regulated genes within the rat QTL for left ventricular mass.
Figure 3: Correlation between hemodynamic indices and indexed LVM in individuals with aortic stenosis.
Figure 4: Molecular characterization of rat Ogn.
Figure 5: In vivo regulation of LVM in Ogn knockout mice.

Similar content being viewed by others

Accession codes

Accessions

ArrayExpress

Gene Expression Omnibus

References

  1. Hubner, N. et al. Integrated transcriptional profiling and linkage analysis for identification of genes underlying disease. Nat. Genet. 37, 243–253 (2005).

    Article  CAS  Google Scholar 

  2. Passador-Gurgel, G., Hsieh, W.P., Hunt, P., Deighton, N. & Gibson, G. Quantitative trait transcripts for nicotine resistance in Drosophila melanogaster. Nat. Genet. 39, 264–268 (2007).

    Article  CAS  Google Scholar 

  3. Lorell, B.H. & Carabello, B.A. Left ventricular hypertrophy: pathogenesis, detection, and prognosis. Circulation 102, 470–479 (2000).

    Article  CAS  Google Scholar 

  4. Devereux, R.B. et al. Relations of left ventricular mass to demographic and hemodynamic variables in American Indians: The Strong Heart Study. Circulation 96, 1416–1423 (1997).

    Article  CAS  Google Scholar 

  5. Pravenec, M. et al. Mapping of quantitative trait loci for blood pressure and cardiac mass in the rat by genome scanning of recombinant inbred strains. J. Clin. Invest. 96, 1973–1978 (1995).

    Article  CAS  Google Scholar 

  6. Post, W.S., Larson, M.G., Myers, R.H., Galderisi, M. & Levy, D. Heritability of left ventricular mass: the Framingham Heart Study. Hypertension 30, 1025–1028 (1997).

    Article  CAS  Google Scholar 

  7. de Koning, D.J. & Haley, C.S. Genetical genomics in humans and model organisms. Trends Genet. 21, 377–381 (2005).

    Article  CAS  Google Scholar 

  8. Petretto, E. et al. Heritability and tissue specificity of expression quantitative trait loci. PLoS Genet. 2, e172 (2006).

    Article  Google Scholar 

  9. Hoshijima, M. & Chien, K.R. Mixed signals in heart failure: cancer rules. J. Clin. Invest. 109, 849–855 (2002).

    Article  CAS  Google Scholar 

  10. Cerutti, C. et al. Transcriptional alterations in the left ventricle of three hypertensive rat models. Physiol. Genomics 27, 295–308 (2006).

    Article  CAS  Google Scholar 

  11. Yoshioka, J. et al. Thioredoxin-interacting protein controls cardiac hypertrophy through regulation of thioredoxin activity. Circulation 109, 2581–2586 (2004).

    Article  CAS  Google Scholar 

  12. O'Connell, T.D. et al. The α(1A/C)- and α(1B)-adrenergic receptors are required for physiological cardiac hypertrophy in the double-knockout mouse. J. Clin. Invest. 111, 1783–1791 (2003).

    Article  CAS  Google Scholar 

  13. Goring, H.H. et al. Discovery of expression QTLs using large-scale transcriptional profiling in human lymphocytes. Nat. Genet. 39, 1208–1216 (2007).

    Article  Google Scholar 

  14. Cook, S.A., Novikov, M.S., Ahn, Y., Matsui, T. & Rosenzweig, A. A20 is dynamically regulated in the heart and inhibits the hypertrophic response. Circulation 108, 664–667 (2003).

    Article  Google Scholar 

  15. Oka, T. et al. Genetic manipulation of periostin expression reveals a role in cardiac hypertrophy and ventricular remodeling. Circ. Res. 101, 313–321 (2007).

    Article  CAS  Google Scholar 

  16. Berk, B.C., Fujiwara, K. & Lehoux, S. ECM remodeling in hypertensive heart disease. J. Clin. Invest. 117, 568–575 (2007).

    Article  CAS  Google Scholar 

  17. Cheitlin, M.D. et al. ACC/AHA/ASE 2003 guideline update for the clinical application of echocardiography. J. Am. Soc. Echocardiogr. 16, 1091–1110 (2003).

    PubMed  Google Scholar 

  18. Birney, E. et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 447, 799–816 (2007).

    Article  CAS  Google Scholar 

  19. Tasheva, E.S. et al. Mimecan/osteoglycin-deficient mice have collagen fibril abnormalities. Mol. Vis. 8, 407–415 (2002).

    CAS  PubMed  Google Scholar 

  20. Hirota, H. et al. Loss of a gp130 cardiac muscle cell survival pathway is a critical event in the onset of heart failure during biomechanical stress. Cell 97, 189–198 (1999).

    Article  CAS  Google Scholar 

  21. Schroen, B. et al. Lysosomal integral membrane protein 2 is a novel component of the cardiac intercalated disc and vital for load-induced cardiac myocyte hypertrophy. J. Exp. Med. 204, 1227–1235 (2007).

    Article  CAS  Google Scholar 

  22. Tasheva, E.S., Corpuz, L.M., Funderburgh, J.L. & Conrad, G.W. Differential splicing and alternative polyadenylation generate multiple mimecan mRNA transcripts. J. Biol. Chem. 272, 32551–32556 (1997).

    Article  CAS  Google Scholar 

  23. Wang, G.S. & Cooper, T.A. Splicing in disease: disruption of the splicing code and the decoding machinery. Nat. Rev. Genet. 8, 749–761 (2007).

    Article  CAS  Google Scholar 

  24. Kresse, H. & Schonherr, E. Proteoglycans of the extracellular matrix and growth control. J. Cell. Physiol. 189, 266–274 (2001).

    Article  CAS  Google Scholar 

  25. Zhang, D. et al. TAK1 is activated in the myocardium after pressure overload and is sufficient to provoke heart failure in transgenic mice. Nat. Med. 6, 556–563 (2000).

    Article  CAS  Google Scholar 

  26. Hashimoto, J., Imai, Y. & O'Rourke, M.F. Indices of pulse wave analysis are better predictors of left ventricular mass reduction than cuff pressure. Am. J. Hypertens. 20, 378–384 (2007).

    Article  Google Scholar 

  27. Cook, S.A., Sugden, P.H. & Clerk, A. Regulation of bcl-2 family proteins during development and in response to oxidative stress in cardiac myocytes: association with changes in mitochondrial membrane potential. Circ. Res. 85, 940–949 (1999).

    Article  CAS  Google Scholar 

  28. Schroen, B. et al. Thrombospondin-2 is essential for myocardial matrix integrity: increased expression identifies failure-prone cardiac hypertrophy. Circ. Res. 95, 515–522 (2004).

    Article  CAS  Google Scholar 

  29. Caruthers, S.D. et al. Practical value of cardiac magnetic resonance imaging for clinical quantification of aortic valve stenosis: comparison with echocardiography. Circulation 108, 2236–2243 (2003).

    Article  Google Scholar 

  30. Tusher, V.G., Tibshirani, R. & Chu, G. Significance analysis of microarrays applied to the ionizing radiation response. Proc. Natl. Acad. Sci. USA 98, 5116–5121 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Mice used in this study were produced by Eli Lilly, Inc., and made available to the authors. We are grateful to R. Buchan for technical assistance, to P. Froguel, A. Angius, M. Falchi and M. Schneider for comments on the manuscript, to S. Harding (National Heart and Lung Institute, Imperial College, London) for providing the isolated adult rat cardiac myocytes and to J. Sassard (University of Lyon) for providing DNA from the Lyon rat strains. This work was primarily supported by funding from the UK Department of Health (S.A.C. and H.L.) and the British Heart Foundation (R.S., S.A.C.). In addition, studies were supported by research grants from the Medical Research Council of UK (T.J.A., S.A.C.), the Fondation Leducq (T.J.A., S.A.C., M.B.), the EU EURATools award (T.J.A., S.A.C., N.H., M.P.), the Wellcome Trust (I.G.), the Howard Hughes Medical Institute Research Scholars Program (M.P.), the Grant Agency of the Czech Republic (M.P.), the Ministry of Education of the Czech Republic (M.P., V.K.), the 2003T302 grant of the Netherlands Heart Foundation (Y.M.P), the InterCardiology Institute Netherlands (Y.M.P.), a Rubicon grant from the Netherlands Organisation for Scientific Research (NWO, to B.S.), the Wellcome Trust Functional Genomics Initiative and the Biological Atlas of Insulin Resistance (BAIR) (M.K.K.), the German National Genome Research Network (NGFN2, to N.H.), and the US National Institutes of Health's National Eye Institute EY000952 and EY13395 (G.W.C.).

Author information

Authors and Affiliations

Authors

Contributions

The study was designed by S.A.C., E.P. and T.J.A.; S.A.C. obtained funding, supervised the study and coordinated the collaborations; R.S. performed PCR-based experiments and genotyping; H.L. and M.K.K. generated rat microarray data; B.S. and Y.M.P. generated human microarray data; M.B. generated immunofluorescence confocal micrographs; R.S. and H.L. performed cell culture and cloning experiments; R.S., B.S. and M.B. performed immunoblotting; P.J.M. and R.S. performed in vivo analyses in Ogn knockout mice; E.S.T., L.M.C., M.D.W. and G.W.C. provided and genotyped the Ogn knockout mice; N.H. and J.F. carried out sequence analysis of Ogn; T.W.K., V.K. and M.P. provided telemetric blood pressure data; P.P.P. provided human tissues for protein studies; S.K.P., D.J.P. and C.K. provided the human cardiac MRI data; E.P. designed, interpreted and supervised all statistical analyses; E.P., I.G. and R.S. performed statistical and bioinformatic analyses and were aided by J.M.; and E.P. and S.A.C. wrote the manuscript.

Corresponding authors

Correspondence to Timothy J Aitman or Stuart A Cook.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8, Supplementary Tables 1–10, Supplementary Methods (PDF 755 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Petretto, E., Sarwar, R., Grieve, I. et al. Integrated genomic approaches implicate osteoglycin (Ogn) in the regulation of left ventricular mass. Nat Genet 40, 546–552 (2008). https://doi.org/10.1038/ng.134

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.134

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