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:

Subcellular localization of MC4R with ADCY3 at neuronal primary cilia underlies a common pathway for genetic predisposition to obesity

Abstract

Most monogenic cases of obesity in humans have been linked to mutations in genes encoding members of the leptin–melanocortin pathway. Specifically, mutations in MC4R, the melanocortin-4 receptor gene, account for 3–5% of all severe obesity cases in humans1,2,3. Recently, ADCY3 (adenylyl cyclase 3) gene mutations have been implicated in obesity4,5. ADCY3 localizes to the primary cilia of neurons6, organelles that function as hubs for select signaling pathways. Mutations that disrupt the functions of primary cilia cause ciliopathies, rare recessive pleiotropic diseases in which obesity is a cardinal manifestation7. We demonstrate that MC4R colocalizes with ADCY3 at the primary cilia of a subset of hypothalamic neurons, that obesity-associated MC4R mutations impair ciliary localization and that inhibition of adenylyl cyclase signaling at the primary cilia of these neurons increases body weight. These data suggest that impaired signaling from the primary cilia of MC4R neurons is a common pathway underlying genetic causes of obesity in humans.

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

Fig. 1: MC4R localizes to the primary cilia in heterologous cells.
Fig. 2: MC4R localizes specifically to the primary cilia of a subset of paraventricular nucleus (PVN) neurons.
Fig. 3: A subset of human obesity-associated mutations selectively impairs ciliary localization of MC4R.
Fig. 4: Inhibition of adenylyl cyclase at the primary cilia of Sim1 paraventricular nucleus (PNV) neurons is sufficient to cause weight gain.

Similar content being viewed by others

References

  1. Lubrano-Berthelier, C. et al. Melanocortin 4 receptor mutations in a large cohort of severely obese adults: prevalence, functional classification, genotype-phenotype relationship, and lack of association with binge eating. J. Clin. Endocrinol. Metab. 91, 1811–1818 (2006).

    Article  CAS  Google Scholar 

  2. Vaisse, C. et al. Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J. Clin. Invest. 106, 253–262 (2000).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Stergiakouli, E. et al. Genome-wide association study of height-adjusted BMI in childhood identifies functional variant in ADCY3. Obesity (Silver Spring) 22, 2252–2259 (2014).

    Article  CAS  Google Scholar 

  5. Wang, Z. et al. Adult type 3 adenylyl cyclase-deficient mice are obese. PLoS One 4, e6979 (2009).

    Article  Google Scholar 

  6. Bishop, G. A., Berbari, N. F., Lewis, J. & Mykytyn, K. Type III adenylyl cyclase localizes to primary cilia throughout the adult mouse brain. J. Comp. Neurol. 505, 562–571 (2007).

    Article  Google Scholar 

  7. Reiter, J. F. & Leroux, M. R. Genes and molecular pathways underpinning ciliopathies. Nat. Rev. Mol. Cell Biol. 18, 533–547 (2017).

    Article  CAS  Google Scholar 

  8. Goetz, S. C. & Anderson, K. V. The primary cilium: a signalling centre during vertebrate development. Nat. Rev. Genet. 11, 331–344 (2010).

    Article  CAS  Google Scholar 

  9. Green, J. A. & Mykytyn, K. Neuronal ciliary signaling in homeostasis and disease. Cell. Mol. Life Sci. 67, 3287–3297 (2010).

    Article  CAS  Google Scholar 

  10. Hildebrandt, F., Benzing, T. & Katsanis, N. Ciliopathies. N. Engl. J. Med. 364, 1533–1543 (2011).

    Article  CAS  Google Scholar 

  11. Vaisse, C., Reiter, J. F. & Berbari, N. F. Cilia and obesity. Cold Spring Harb. Perspect. Biol. 9, a028217 (2017).

    Article  Google Scholar 

  12. Shalata, A. et al. Morbid obesity resulting from inactivation of the ciliary protein CEP19 in humans and mice. Am. J. Hum. Genet. 93, 1061–1071 (2013).

    Article  CAS  Google Scholar 

  13. Acs, P. et al. A novel form of ciliopathy underlies hyperphagia and obesity in Ankrd26 knockout mice. Brain Struct. Funct. 220, 1511–1528 (2015).

    Article  CAS  Google Scholar 

  14. Davenport, J. R. et al. Disruption of intraflagellar transport in adult mice leads to obesity and slow-onset cystic kidney disease. Curr. Biol. 17, 1586–1594 (2007).

    Article  CAS  Google Scholar 

  15. Wu, L., Shen, C., Seed Ahmed, M., Östenson, C. G. & Gu, H. F. Adenylate cyclase 3: a new target for anti-obesity drug development. Obes. Rev. 17, 907–914 (2016).

    Article  CAS  Google Scholar 

  16. Krashes, M. J., Lowell, B. B. & Garfield, A. S. Melanocortin-4 receptor-regulated energy homeostasis. Nat. Neurosci. 19, 206–219 (2016).

    Article  CAS  Google Scholar 

  17. 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 

  18. Morton, G. J., Meek, T. H. & Schwartz, M. W. Neurobiology of food intake in health and disease. Nat. Rev. Neurosci. 15, 367–378 (2014).

    Article  CAS  Google Scholar 

  19. Lubrano-Berthelier, C. et al. Intracellular retention is a common characteristic of childhood obesity-associated MC4R mutations. Hum. Mol. Genet. 12, 145–153 (2003).

    Article  CAS  Google Scholar 

  20. Aanstad, P. et al. The extracellular domain of Smoothened regulates ciliary localization and is required for high-level Hh signaling. Curr. Biol. 19, 1034–1039 (2009).

    Article  CAS  Google Scholar 

  21. Corbit, K. C. et al. Vertebrate Smoothened functions at the primary cilium. Nature 437, 1018–1021 (2005).

    Article  CAS  Google Scholar 

  22. Balthasar, N. et al. Divergence of melanocortin pathways in the control of food intake and energy expenditure. Cell 123, 493–505 (2005).

    Article  CAS  Google Scholar 

  23. Garfield, A. S. et al. A neural basis for melanocortin-4 receptor-regulated appetite. Nat. Neurosci. 18, 863–871 (2015).

    Article  CAS  Google Scholar 

  24. Bromberg, Y., Overton, J., Vaisse, C., Leibel, R. L. & Rost, B. In silico mutagenesis: a case study of the melanocortin 4 receptor. FASEB J. 23, 3059–3069 (2009).

    Article  CAS  Google Scholar 

  25. Calton, M. et al. Association of functionally significant Melanocortin-4 but not Melanocortin-3 receptor mutations with severe adult obesity in a large North-American case control study. Hum. Mol. Genet. 18, 1140–1147 (2009).

    Article  CAS  Google Scholar 

  26. Ersoy, B. A. et al. Mechanism of N-terminal modulation of activity at the melanocortin-4 receptor GPCR. Nat. Chem. Biol. 8, 725–730 (2012).

    Article  CAS  Google Scholar 

  27. Hinney, A. et al. Melanocortin-4 receptor gene: case-control study and transmission disequilibrium test confirm that functionally relevant mutations are compatible with a major gene effect for extreme obesity. J. Clin. Endocrinol. Metab. 88, 4258–4267 (2003).

    Article  CAS  Google Scholar 

  28. Berbari, N. F., Johnson, A. D., Lewis, J. S., Askwith, C. C. & Mykytyn, K. Identification of ciliary localization sequences within the third intracellular loop of G protein-coupled receptors. Mol. Biol. Cell 19, 1540–1547 (2008).

    Article  CAS  Google Scholar 

  29. Marley, A., Choy, R. W. & von Zastrow, M. GPR88 reveals a discrete function of primary cilia as selective insulators of GPCR cross-talk. PLoS One 8, e70857 (2013).

    Article  CAS  Google Scholar 

  30. Fenselau, H. et al. A rapidly acting glutamatergic ARC→PVH satiety circuit postsynaptically regulated by α-MSH. Nat. Neurosci. 20, 42–51 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported by funds from UCSF DRC NIH P30DK063720 and UCSF NORC NIH P30DK098722; an AMC Graduate School PhD Scholarship to J.E.S.; NIH R01AR05439 and NIH R01GM095941, the Burroughs Wellcome Fund and the David & Lucile F. Packard Foundation to J.F.R.; NIH R01DK60540 to C.V.; NIH RO1DK106404 to C.V. and J.F.R.; NIH RO1DA012864 and NIH RO1DA010711 to M.V.Z.; and a New Frontier Research Award through the UCSF Program for Breakthrough Biomedical Research to C.V., J.F.R. and M.V.Z. We thank K. Deisseroth (Stanford University) for providing plasmids.

Author information

Authors and Affiliations

Authors

Contributions

C.V. and J.F.R. supervised the research. J.E.S., Y.W., C.V. and J.F.R. conceived and designed experiments, performed experiments, performed statistical analysis, analyzed the data and wrote the paper. S.Z. performed experiments. A.A.B. performed experiments and analyzed data relevant to Fig. 4. B.A.E. conceived and performed experiments and analyzed data relevant to Fig. 2. A.M. and M.V.Z. contributed regents and expertise relevant to Fig. 4.

Corresponding author

Correspondence to Christian Vaisse.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Sim1-expressing neurons in the paraventricular nucleus are ciliated

a Hypothalamic PVN section of a Tg(Sim1-EGFP) mouse. Sim1 lineage neurons are in green, the neuronal cilia marker Adcy3 is labeled in red, nuclei in pink. b Increased magnification indicating that all Sim1-expressing neurons possess an Adcy3-positive cilium. c Maximal intensity Z projection of a confocal image of a single EGFP-expressing Sim1 neuron in the PVN. Scale bars represent 10 µm.

Supplementary Figure 2 Serpentine plot of human MC4R

Mutations found in obese patients are indicated by a black arrow from the mutated amino-acid to the colored circle that indicates the amino-acid in the mutant protein. Red arrows indicate frameshift mutations. Most patients were heterozygous for one mutation. Amino-acid changes causing severe effects on MC4R membrane expression are in red. Such mutations are expected to alter membrane and cilia localization as well as ligand response. Mutations in blue lead to decreased agonist activation (blue) or decreased constitutive activity (purple). Whether these mutations also alter cilia localization has not been tested. Mutations tested in Fig. 3a are in yellow. Mutations leading specifically to decreased ciliary expression are indicated by a green arrow.

Supplementary Figure 3 Experimental design and validation of coexpression of GPR88* and mCherry

a, b Design of DIO AAV-expressing Flag-GPR88* or mCherry, stereotaxically injected in experimental a and control b mice. c Coronal section of the PVN of the hypothalamus of Sim1-Cre mice injected with the AAV DIO Flag-GPR88* + AAV DIO mCherry allows for verification of the accuracy and PVN coverage of the injections. d–e Maximal intensity projections of confocal sections through the PVN region indicated by a red square in c show that all mCherry expressing neurons also express Flag-GPR88* (green). White scale bars represent 10 µm.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–3, Supplementary Note and Supplementary Table 1

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Siljee, J.E., Wang, Y., Bernard, A.A. et al. Subcellular localization of MC4R with ADCY3 at neuronal primary cilia underlies a common pathway for genetic predisposition to obesity. Nat Genet 50, 180–185 (2018). https://doi.org/10.1038/s41588-017-0020-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-017-0020-9

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