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:

Inactivation of the mouse melanocortin-3 receptor results in increased fat mass and reduced lean body mass

Abstract

Genetic1,2,3,4,5,6,7,8 and pharmacological6,9,10,11,12 studies have defined a role for the melanocortin-4 receptor (Mc4r) in the regulation of energy homeostasis. The physiological function of Mc3r, a melanocortin receptor expressed at high levels in the hypothalamus13, has remained unknown. We evaluated the potential role of Mc3r in energy homeostasis by studying Mc3r-deficient (Mc3r−/−) mice and compared the functions of Mc3r and Mc4r in mice deficient for both genes. The 4–6-month Mc3r−/− mice have increased fat mass, reduced lean mass and higher feed efficiency than wild-type littermates, despite being hypophagic and maintaining normal metabolic rates. (Feed efficiency is the ratio of weight gain to food intake.) Consistent with increased fat mass, Mc3r−/− mice are hyperleptinaemic and male Mc3r−/− mice develop mild hyperinsulinaemia. Mc3r−/− mice did not have significantly altered corticosterone or total thyroxine (T4) levels. Mice lacking both Mc3r and Mc4r become significantly heavier than Mc4r−/− mice. We conclude that Mc3r and Mc4r serve non-redundant roles in the regulation of energy homeostasis.

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: Targeted disruption of the mouse Mc3r locus.
Figure 2: Growth curves (a,b) and body composition (c, d) of wild-type (+/+), Mc3r+/− (+/−) and Mc3r−/− (−/−) littermate mice.
Figure 3: Adipose tissue mass in wild-type (+/+), Mc3r+/− (+/−) and Mc3r−/− (−/−) littermates (a,b,c,d).
Figure 4: Plasma leptin (a), insulin (b), glucose (c), corticosterone (d) and total T4 (e) levels of wild-type (+/+), Mc3r+/− (+/−) and Mc3r−/− (−/−) littermates.
Figure 5: Food intake, weight gain and feed efficiency of individually housed male wild-type (+/+, n=11), Mc3r+/− (+/−, n=20) and Mc3r−/− (−/−, n=16) littermates were maintained on a regular chow diet.
Figure 6: Metabolic rate (a), locomotor activity (b,d) and fine movements (c,e) of individually housed Mc3r −/− (−/−) and wild-type (+/+) littermates.

Similar content being viewed by others

References

  1. Krude, H. et al. Severe early onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nature Genet. 19, 155–157 ( 1998).

    Article  CAS  Google Scholar 

  2. Chagnon, Y.C. et al. Linkage and association studies between the melanocortin receptors 4 and 5 genes and obesity-related phenotypes in the quebec family study. Mol. Med. 3, 663–673 ( 1997).

    Article  CAS  Google Scholar 

  3. Yeo, G.S.H. et al. A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nature Genet. 20, 111–112 (1998).

    Article  CAS  Google Scholar 

  4. Vaisse, C., Clement, K., Guy-Grand, B. & Froguel, P. A frameshift mutation in human MC4R is associated with a dominant form of obesity. Nature Genet. 20, 113– 114 (1998).

    Article  CAS  Google Scholar 

  5. Michaud, E.J. et al. A molecular model for the genetic and phenotypic characteristics of the mouse lethal yellow (Ay) mutation. Proc. Natl Acad. Sci. USA 91, 2562–2566 (1994).

    Article  CAS  Google Scholar 

  6. Ollmann, M.M. et al. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278, 135–138 (1997).

    Article  CAS  Google Scholar 

  7. Graham, M., Shutter, J.R., Sarmiento, U., Sarosi, I. & Stark, K.L. Overexpression of Agrt leads to obesity in transgenic mice. Nature Genet. 17, 273–274 (1997).

    Article  CAS  Google Scholar 

  8. Huszar, D. et al. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 88, 131– 141 (1997).

    Article  CAS  Google Scholar 

  9. Fan, W., Boston, B.A., Kesterson, R.A., Hruby, V.J. & Cone, R.D. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385, 165–168 (1997).

    Article  CAS  Google Scholar 

  10. Lu, D. et al. Agouti protein is an antagonist of the melanocyte-stimulating-hormone receptor. Nature 371, 799– 802 (1994).

    Article  CAS  Google Scholar 

  11. Marsh, D.J. et al. Response of melanocortin-4 receptor-deficient mice to anorectic and orexigenic peptides. Nature Genet. 21, 119–122 (1999).

    Article  CAS  Google Scholar 

  12. Chen, A.S. et al. Role of the melanocortin-4 receptor in metabolic rate and food intake in mice. Transgenic Res. (in press).

  13. Roselli-Rehfuss, L. et al. Identification of a receptor for γ melanotropin and other proopiomelanocortin peptides in the hypothalamus and limbic system. Proc. Natl Acad. Sci. USA 90, 8856– 8860 (1993).

    Article  CAS  Google Scholar 

  14. Johnson, P.R. & Hirsch, J. Cellularity of adipose depots in six strains of genetically obese mice. J. Lipid Res. 13, 2–11 (1972).

    CAS  PubMed  Google Scholar 

  15. Yen, T.T., Allan, J.A., Yu, P.-L., Acton, M.A. & Pearson, D.V. Triacylglycerol contents and in vivo lipogenesis of ob/ob, db/db and Avy/a mice. Biochem. Biophys. Acta 441, 213–220 ( 1976).

    Article  CAS  Google Scholar 

  16. Yen, T.T., Steinmetz, J. & Wolff, G.L. Lipolysis in genetically obese and diabetes-prone mice. Horm. Metab. Res. 2, 200– 203 (1970).

    Article  CAS  Google Scholar 

  17. Jones, B.H. et al. Upregulation of adipocyte metabolism by agouti protein: possible paracrine actions in yellow mouse obesity. Am. J. Physiol. 270, E192–E196 (1996).

    Article  CAS  Google Scholar 

  18. Xue, B., Moustaid-Moussa, N., Wilkison, W.O. & Zemel, M.B. The agouti gene product inhibits lipolysis in human adipocytes via a Ca2+-dependent mechanism. FASEB 12, 1391–1396 (1998).

    Article  CAS  Google Scholar 

  19. Fekete, C. et al. α-Melanocyte-stimulating hormone is contained in nerve terminals innervating thyrotropin-releasing hormone-synthesizing neurons in the hypothalamic paraventricular nucleus and prevents fasting-induced suppression of prothyrotropin-releasing hormone gene expression. J. Neurosci. 20, 1550–1558 ( 2000).

    Article  CAS  Google Scholar 

  20. Kim, M.S. et al. The central melanocortin system affects the hypothalamo-pituitary thyroid axis and may mediate the effect of leptin. J. Clin. Invest. 105, 1005–1011 ( 2000).

    Article  CAS  Google Scholar 

  21. Frigeri, L.G., Wolff, G.L. & Teguh, C. Differential responses of yellow Avy/A and agouti A/a (BALB/c×VY) F1 hybrid mice to the same diets: glucose tolerance, weight gain, and adipocyte cellularity. Int. J. Obes. 12, 305–320 ( 1988).

    CAS  PubMed  Google Scholar 

  22. Shimizu, H., Shargill, N.S., Bray, G.A., Yen, T.T. & Geselichen, P.D. Effects of MSH on food intake, body weight and coat color of the yellow obese mouse. Life Sci. 45, 543–552 ( 1989).

    Article  CAS  Google Scholar 

  23. Hruby, V.J. et al. Cyclic lactam α-melanotropin analogues of Ac-Nle4-cyclo[Asp5, D-Phe7, Lys10] α-melanocyte-stimulating hormone-(4-10)-NH2 with bulky aromatic amino acids at position 7 show high antagonist potency and selectivity at specific melanocortin receptors. J. Med. Chem. 38 , 3454–3461 (1995).

    Article  CAS  Google Scholar 

  24. Bagnol, D. et al. Anatomy of an endogenous antagonist: relationship between agouti-related protein and proopiomelanocortin in brain. J. Neurosci. 19, RC26 (1–7) (1999).

    Article  CAS  Google Scholar 

  25. Gantz, I. et al. Molecular cloning of a novel melanocortin receptor. J. Biol. Chem. 268, 8246–8250 (1993).

    CAS  PubMed  Google Scholar 

  26. Chhajlani, V. Distribution of cDNA for melanocortin receptor subtypes in human tissues. Biochem. Mol. Biol. Int. 38, 73–80 (1996).

    CAS  PubMed  Google Scholar 

  27. Lembertas, A.V. et al. Identification of an obesity quantitative trait locus on mouse chromosome 2 and evidence of linkage to body fat and insulin on the human homologous region 20q. J. Clin. Invest. 100, 1240–1247 (1997).

    Article  CAS  Google Scholar 

  28. Guan, X.-M., Yu, H. & Van der Ploeg, L.H.T. Evidence of altered hypothalamic pro-opiomelanocortin/neuropeptide Y mRNA expression in tubby mice. Mol. Brain Res. 59 , 273–279 (1998).

    Article  CAS  Google Scholar 

  29. Kelly, T.L., Berger, N. & Richardson, T.L. DXA body composition: theory and practice. Appl. Radiat. Isot. 49, 511–513 (1998).

    Article  CAS  Google Scholar 

  30. Ellis, K.J. Human body composition: in vivo methods. Physiol. Rev. 80, 649–680 (2000).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Weinberg for providing rat Mc3r cDNA and J. Ronan for assistance in the histological evaluation of adipose tissues.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lex H.T. Van der Ploeg.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chen, A., Marsh, D., Trumbauer, M. et al. Inactivation of the mouse melanocortin-3 receptor results in increased fat mass and reduced lean body mass. Nat Genet 26, 97–102 (2000). https://doi.org/10.1038/79254

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/79254

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