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:

Validation of candidate causal genes for obesity that affect shared metabolic pathways and networks

This article has been updated

Abstract

A principal task in dissecting the genetics of complex traits is to identify causal genes for disease phenotypes. We previously developed a method to infer causal relationships among genes through the integration of DNA variation, gene transcription and phenotypic information. Here we have validated our method through the characterization of transgenic and knockout mouse models of genes predicted to be causal for abdominal obesity. Perturbation of eight out of the nine genes, with Gas7, Me1 and Gpx3 being newly confirmed, resulted in significant changes in obesity-related traits. Liver expression signatures revealed alterations in common metabolic pathways and networks contributing to abdominal obesity and overlapped with a macrophage-enriched metabolic network module that is highly associated with metabolic traits in mice and humans. Integration of gene expression in the design and analysis of traditional F2 intercross studies allows high-confidence prediction of causal genes and identification of pathways and networks involved.

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: Adiposity (fat/muscle ratio) or body weight growth curves in the mouse models.
Figure 2: Disruption of metabolic pathways involved in fat pad mass in mouse models of the candidate genes.
Figure 3: A portion of the core subnetwork, derived from the liver transcriptional subnetworks representative of gene expression signatures of the mouse models of the candidate genes.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

Change history

  • 22 March 2009

    NOTE: In the version of this article initially published online, there was an error in one of the genotypes in Table 5, a missing genotype in the third-to-last paragraph on page 7, and two minor typographical errors in the right column on page 6. These errors have been corrected for the print, PDF and HTML versions of this article.

References

  1. Doss, S., Schadt, E.E., Drake, T.A. & Lusis, A.J. Cis-acting expression quantitative trait loci in mice. Genome Res. 15, 681–691 (2005).

    Article  CAS  Google Scholar 

  2. Monks, S.A. et al. Genetic inheritance of gene expression in human cell lines. Am. J. Hum. Genet. 75, 1094–1105 (2004).

    Article  CAS  Google Scholar 

  3. Schadt, E.E. et al. Genetics of gene expression surveyed in maize, mouse and man. Nature 422, 297–302 (2003).

    Article  CAS  Google Scholar 

  4. Schadt, E.E. et al. An integrative genomics approach to infer causal associations between gene expression and disease. Nat. Genet. 37, 710–717 (2005).

    Article  CAS  Google Scholar 

  5. Sieberts, S.K. & Schadt, E.E. Moving toward a system genetics view of disease. Mamm. Genome 18, 389–401 (2007).

    Article  Google Scholar 

  6. Merkel, M. et al. Inactive lipoprotein lipase (LPL) alone increases selective cholesterol ester uptake in vivo, whereas in the presence of active LPL it also increases triglyceride hydrolysis and whole particle lipoprotein uptake. J. Biol. Chem. 277, 7405–7411 (2002).

    Article  Google Scholar 

  7. Mirochnitchenko, O., Palnitkar, U., Philbert, M. & Inouye, M. Thermosensitive phenotype of transgenic mice overproducing human glutathione peroxidases. Proc. Natl. Acad. Sci. USA 92, 8120–8124 (1995).

    Article  CAS  Google Scholar 

  8. Huq, A.H., Lovell, R.S., Ou, C.N., Beaudet, A.L. & Craigen, W.J. X-linked glycerol kinase deficiency in the mouse leads to growth retardation, altered fat metabolism, autonomous glucocorticoid secretion and neonatal death. Hum. Mol. Genet. 6, 1803–1809 (1997).

    Article  CAS  Google Scholar 

  9. Chen, Y. et al. Variations in DNA elucidate molecular networks that cause disease. Nature 452, 429–435 (2008).

    Article  CAS  Google Scholar 

  10. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  Google Scholar 

  11. Mi, H. et al. The PANTHER database of protein families, subfamilies, functions and pathways. Nucleic Acids Res. 33, D284–D288 (2005).

    Article  CAS  Google Scholar 

  12. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

  13. Ghazalpour, A. et al. Genomic analysis of metabolic pathway gene expression in mice. Genome Biol. 6, R59 (2005).

    Article  Google Scholar 

  14. Emilsson, V. et al. Genetics of gene expression and its effect on disease. Nature 452, 423–428 (2008).

    Article  CAS  Google Scholar 

  15. Masuzaki, H. et al. A transgenic model of visceral obesity and the metabolic syndrome. Science 294, 2166–2170 (2001).

    Article  CAS  Google Scholar 

  16. Liu, M. et al. Network-based analysis of affected biological processes in type 2 diabetes models. PLoS Genet. 3, e96 (2007).

    Article  Google Scholar 

  17. Zhu, J. et al. Increasing the power to detect causal associations by combining genotypic and expression data in segregating populations. PLoS Comput. Biol. 3, e69 (2007).

    Article  Google Scholar 

  18. Schadt, E.E. et al. Mapping the genetic architecture of gene expression in human liver. PLoS Biol. 6, e107 (2008).

    Article  Google Scholar 

  19. Zhu, J. et al. Integrating large-scale functional genomic data to dissect the complexity of yeast regulatory networks. Nat. Genet. 40, 854–861 (2008).

    Article  CAS  Google Scholar 

  20. Yabe, D., Brown, M.S. & Goldstein, J.L. Insig-2, a second endoplasmic reticulum protein that binds SCAP and blocks export of sterol regulatory element-binding proteins. Proc. Natl. Acad. Sci. USA 99, 12753–12758 (2002).

    Article  CAS  Google Scholar 

  21. Janowski, B.A. The hypocholesterolemic agent LY295427 up-regulates INSIG-1, identifying the INSIG-1 protein as a mediator of cholesterol homeostasis through SREBP. Proc. Natl. Acad. Sci. USA 99, 12675–12680 (2002).

    Article  CAS  Google Scholar 

  22. Chambers, J.C. et al. Common genetic variation near MC4R is associated with waist circumference and insulin resistance. Nat. Genet. 40, 716–718 (2008).

    Article  CAS  Google Scholar 

  23. Frayling, T.M. et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316, 889–894 (2007).

    Article  CAS  Google Scholar 

  24. Loos, R.J. et al. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat. Genet. 40, 768–775 (2008).

    Article  CAS  Google Scholar 

  25. Mutch, D.M. & Clement, K. Unraveling the genetics of human obesity. PLoS Genet. 2, e188 (2006).

    Article  Google Scholar 

  26. Thorleifsson, G. et al. Genome-wide association yields new sequence variants at seven loci that associate with measures of obesity. Nat. Genet. 41, 18–24 (2009).

    Article  CAS  Google Scholar 

  27. Willer, C.J. et al. Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat. Genet. 41, 25–34 (2008).

    Article  Google Scholar 

  28. Herbert, A. et al. A common genetic variant is associated with adult and childhood obesity. Science 312, 279–283 (2006).

    Article  CAS  Google Scholar 

  29. Wallace, C. et al. Genome-wide association study identifies genes for biomarkers of cardiovascular disease: serum urate and dyslipidemia. Am. J. Hum. Genet. 82, 139–149 (2008).

    Article  CAS  Google Scholar 

  30. Willer, C.J. et al. Newly identified loci that influence lipid concentrations and risk of coronary artery disease. Nat. Genet. 40, 161–169 (2008).

    Article  CAS  Google Scholar 

  31. Saxena, R. et al. Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316, 1331–1336 (2007).

    Article  CAS  Google Scholar 

  32. Dhanasekaran, S.M. et al. Molecular profiling of human prostate tissues: insights into gene expression patterns of prostate development during puberty. FASEB J. 19, 243–245 (2005).

    Article  CAS  Google Scholar 

  33. Francis, K. et al. Complement C3a receptors in the pituitary gland: a novel pathway by which an innate immune molecule releases hormones involved in the control of inflammation. FASEB J. 17, 2266–2268 (2003).

    Article  CAS  Google Scholar 

  34. Ju, Y.T. et al. gas7: A gene expressed preferentially in growth-arrested fibroblasts and terminally differentiated Purkinje neurons affects neurite formation. Proc. Natl. Acad. Sci. USA 95, 11423–11428 (1998).

    Article  CAS  Google Scholar 

  35. Moorthy, P.P., Kumar, A.A. & Devaraj, H. Expression of the Gas7 gene and Oct4 in embryonic stem cells of mice. Stem Cells Dev. 14, 664–670 (2005).

    Article  CAS  Google Scholar 

  36. Ishibashi, N. & Mirochnitchenko, O. Chemokine expression in transgenic mice overproducing human glutathione peroxidases. Methods Enzymol. 353, 460–476 (2002).

    Article  CAS  Google Scholar 

  37. McClung, J.P. et al. Development of insulin resistance and obesity in mice overexpressing cellular glutathione peroxidase. Proc. Natl. Acad. Sci. USA 101, 8852–8857 (2004).

    Article  CAS  Google Scholar 

  38. Lee, Y.S. et al. Dysregulation of adipose GPx3 in obesity contributes to local and systemic oxidative stress. Mol. Endocrinol. 22, 2176–2189 (2008).

    Article  CAS  Google Scholar 

  39. MacDonald, M.J. Feasibility of a mitochondrial pyruvate malate shuttle in pancreatic islets. Further implication of cytosolic NADPH in insulin secretion. J. Biol. Chem. 270, 20051–20058 (1995).

    CAS  PubMed  Google Scholar 

  40. van Schothorst, E.M. et al. Adipose gene expression response of lean and obese mice to short-term dietary restriction. Obesity (Silver Spring) 14, 974–979 (2006).

    Article  CAS  Google Scholar 

  41. Qian, S. et al. Deficiency in cytosolic malic enzyme does not increase acetaminophen-induced hepato-toxicity. Basic Clin. Pharmacol. Toxicol. 103, 36–42 (2008).

    Article  CAS  Google Scholar 

  42. Vidal, O. et al. Malic enzyme 1 genotype is associated with backfat thickness and meat quality traits in pigs. Anim. Genet. 37, 28–32 (2006).

    Article  CAS  Google Scholar 

  43. Rahib, L., MacLennan, N.K., Horvath, S., Liao, J.C. & Dipple, K.M. Glycerol kinase deficiency alters expression of genes involved in lipid metabolism, carbohydrate metabolism, and insulin signaling. Eur. J. Hum. Genet. 15, 646–657 (2007).

    Article  CAS  Google Scholar 

  44. Hibuse, T. et al. Aquaporin 7 deficiency is associated with development of obesity through activation of adipose glycerol kinase. Proc. Natl. Acad. Sci. USA 102, 10993–10998 (2005).

    Article  CAS  Google Scholar 

  45. Kawai, J. et al. Functional annotation of a full-length mouse cDNA collection. Nature 409, 685–690 (2001).

    Article  Google Scholar 

  46. Weng, L. et al. Rosetta error model for gene expression analysis. Bioinformatics 22, 1111–1121 (2006).

    Article  CAS  Google Scholar 

  47. He, Y.D. et al. Microarray standard data set and figures of merit for comparing data processing methods and experiment designs. Bioinformatics 19, 956–965 (2003).

    Article  CAS  Google Scholar 

  48. Storey, J.D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl. Acad. Sci. USA 100, 9440–9445 (2003).

    Article  CAS  Google Scholar 

  49. Yang, X. et al. Tissue-specific expression and regulation of sexually dimorphic genes in mice. Genome Res. 16, 995–1004 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank R. Davis, P. Wen, M. Rosales, X. Wu, K. Ranola and X. Xia for helping with the tissue collection. We would also like to thank L. Ingram-Drake and S. Charugundla for technical support and O. Mirochnitchenko and I. Goldberg for providing mouse models. The study was funded by US National Institutes of Health grants DK072206, HL28481 and HL30568.

Author information

Authors and Affiliations

Authors

Contributions

T.A.D., A.J.L., E.E.S., P.Y.L., X.Y., K.M.D., M.L.R. and D.J.M. designed the study. X.Y., H.Q., G.T., S. Majid, B.F., S.Q., L.W.C., D.E.-S., S. Mumick, S.S.W., A.v.N., A.G., M.M. and C.R.F. performed the experiments. X.Y., J.L.D., J. Zhu, G.T., J.K., K.W, J.R.L., T.X., M.C., J. Zhong, C.Z. and B.Z. participated in data analysis. X.Y., J.L.D., J. Zhu, S.Q. and J. Zhong wrote the manuscript, with advice and editing from T.A.D., A.J.L. and E.E.S. R.R.K. coordinated the study. T.A.D., the corresponding author, certifies that all authors have agreed to all content in the manuscript, including the data as presented.

Corresponding author

Correspondence to Thomas A Drake.

Ethics declarations

Competing interests

X.Y., J. Zhu, S.Q., J. Zhong, R.R.K., S. Mumick, T.X., M.C., C.Z., B.Z., D.J.M., M.L.R., J.L.L., P.Y.L. and E.E.S. are all employees of Merck & Co. Inc.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4, Supplementary Tables 1–14 and Supplementary Methods (PDF 703 kb)

Supplementary Figure 5

(Separate scaleable vector graphic file; open with web browser enabled with svg viewer.) The core subnetwork derived from the liver transcriptional subnetworks representative of gene expression signatures of the mouse models of the candidate genes. The liver transcriptional network is the union of Bayesian networks constructed from three crosses derived from B6, C3H and CAST. This core subnetwork consists of key regulators for fatty acid and lipid metabolism, including Insig1 and Insig2 (in red), and is enriched for genes involved in related GO biological processes. (ZIP 172 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yang, X., Deignan, J., Qi, H. et al. Validation of candidate causal genes for obesity that affect shared metabolic pathways and networks. Nat Genet 41, 415–423 (2009). https://doi.org/10.1038/ng.325

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.325

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