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:

Loss-of-function mutations in the melanocortin 4 receptor in a UK birth cohort

Abstract

Mutations in the melanocortin 4 receptor gene (MC4R) are associated with obesity but little is known about the prevalence and impact of such mutations throughout human growth and development. We examined the MC4R coding sequence in 5,724 participants from the Avon Longitudinal Study of Parents and Children, functionally characterized all nonsynonymous MC4R variants and examined their association with anthropometric phenotypes from childhood to early adulthood. The frequency of heterozygous loss-of-function (LoF) mutations in MC4R was ~1 in 337 (0.30%), considerably higher than previous estimates. At age 18 years, mean differences in body weight, body mass index and fat mass between carriers and noncarriers of LoF mutations were 17.76 kg (95% CI 9.41, 26.10), 4.84 kg m−2 (95% CI 2.19, 7.49) and 14.78 kg (95% CI 8.56, 20.99), respectively. MC4R LoF mutations may be more common than previously reported and carriers of such variants may enter adult life with a substantial burden of excess adiposity.

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: Ligand-activated cAMP accumulation of previously uncharacterized MC4R mutations.
Fig. 2: BMI at different ages with MC4R LoF of cAMP accumulation.
Fig. 3: Association between MC4R LoF of cAMP accumulation with BMI trajectory between the ages of 18 months and 18 years using linear-spline multilevel models.
Fig. 4: Comparison of age-specific and longitudinal associations of MC4R LoF of cAMP accumulation and a weighted genome-wide PRS with BMI.

Similar content being viewed by others

Data availability

Full details of the cohort and study design have been described previously and are available at http://www.alspac.bris.ac.uk. Please note that the study website contains details of all the data that are available through a fully searchable data dictionary and variable search tool (http://www.bristol.ac.uk/alspac/researchers/our-data/). ALSPAC data are available through a system of managed open access. Data for this project were accessed under the project number B2891. The application steps for ALSPAC data access are below.

(1) Please read the ALSPAC access policy, which describes the process of accessing the data in detail and outlines the costs associated with doing so.

(2) You may also find it useful to browse the fully searchable research proposals database, which lists all research projects that have been approved since April 2011.

(3) Please submit your research proposal for consideration by the ALSPAC Executive Committee.

You will receive a response within 10 working days to advise you whether your proposal has been approved. If you have any questions about accessing data, please email alspac-data@bristol.ac.uk.

Code availability

Code for data analyses is freely available on request.

References

  1. Zhang, Y. et al. Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–432 (1994).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  3. Montague, C. T. et al. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387, 903–908 (1997).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Clément, K. et al. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392, 398–401 (1998).

    Article  PubMed  Google Scholar 

  6. Gantz, I. et al. Molecular cloning, expression, and gene localization of a fourth melanocortin receptor. J. Biol. Chem. 268, 15174–15179 (1993).

    Article  CAS  PubMed  Google Scholar 

  7. Mountjoy, K. G., Mortrud, M. T., Low, M. J., Simerly, R. B. & Cone, R. D. Localization of the melanocortin-4 receptor (MC4-R) in neuroendocrine and autonomic control circuits in the brain. Mol. Endocrinol. 8, 1298–1308 (1994).

    CAS  PubMed  Google Scholar 

  8. Cowley, M. A. et al. Integration of NPY, AGRP, and melanocortin signals in the hypothalamic paraventricular nucleus: evidence of a cellular basis for the adipostat. Neuron 24, 155–163 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Cowley, M. A. et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411, 480–484 (2001).

    Article  CAS  PubMed  Google Scholar 

  10. Haynes, W. G., Morgan, D. A., Djalali, A., Sivitz, W. I. & Mark, A. L. Interactions between the melanocortin system and leptin in control of sympathetic nerve traffic. Hypertension 33, 542–547 (1999).

    Article  CAS  PubMed  Google Scholar 

  11. 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  PubMed  Google Scholar 

  12. 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  PubMed  Google Scholar 

  13. Farooqi, I. S. et al. Dominant and recessive inheritance of morbid obesity associated with melanocortin 4 receptor deficiency. J. Clin. Invest. 106, 271–279 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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  PubMed  PubMed Central  Google Scholar 

  15. Hinney, A. et al. Prevalence, spectrum, and functional characterization of melanocortin-4 receptor gene mutations in a representative population-based sample and obese adults from Germany. J. Clin. Endocrinol. Metab. 91, 1761–1769 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Dempfle, A. et al. Large quantitative effect of melanocortin-4 receptor gene mutations on body mass index. J. Med. Genet. 41, 795–800 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Farooqi, I. S. et al. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N. Engl. J. Med. 348, 1085–1095 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Collet, T.-H. et al. Evaluation of a melanocortin-4 receptor (MC4R) agonist (Setmelanotide) in MC4R deficiency. Mol. Metab. 6, 1321–1329 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Stutzmann, F. et al. Prevalence of melanocortin-4 receptor deficiency in Europeans and their age-dependent penetrance in multigenerational pedigrees. Diabetes 57, 2511–2518 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Turcot, V. et al. Protein-altering variants associated with body mass index implicate pathways that control energy intake and expenditure in obesity. Nat. Genet. 50, 26–41 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Gonçalves, J. P. L., Palmer, D. & Meldal, M. MC4R agonists: structural overview on antiobesity therapeutics. Trends Pharmacol. Sci. 39, 402–423 (2018).

    Article  PubMed  Google Scholar 

  22. Apovian, C. M. The obesity epidemic–understanding the disease and the treatment. N. Engl. J. Med. 374, 177–179 (2016).

    Article  PubMed  Google Scholar 

  23. Boyd, A. et al. Cohort profile: the ‘children of the 90s’–the index offspring of the Avon Longitudinal Study of Parents and Children. Int. J. Epidemiol. 42, 111–127 (2013).

    Article  PubMed  Google Scholar 

  24. Fraser, A. et al. Cohort profile: the Avon Longitudinal Study of Parents and Children: ALSPAC mothers cohort. Int. J. Epidemiol. 42, 97–110 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Cornish, R. P., Macleod, J., Boyd, A. & Tilling, K. Factors associated with participation over time in the Avon Longitudinal Study of Parents and Children: a study using linked education and primary care data. Int. J. Epidemiol. 50, 293–302 (2020).

    Article  PubMed Central  Google Scholar 

  26. Lotta, L. A. et al. Human gain-of-function MC4R variants show signaling bias and protect against obesity. Cell 177, 597–607 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Greenfield, J. R. et al. Modulation of blood pressure by central melanocortinergic pathways. N. Engl. J. Med. 360, 44–52 (2009).

    Article  CAS  PubMed  Google Scholar 

  28. Khera, A. V. et al. Polygenic prediction of weight and obesity trajectories from birth to adulthood. Cell 177, 587–596 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Krakoff, J. et al. Lower metabolic rate in individuals heterozygous for either a frameshift or a functional missense MC4R variant. Diabetes 57, 3267–3272 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Thearle, M. S. et al. Greater impact of melanocortin-4 receptor deficiency on rates of growth and risk of type 2 diabetes during childhood compared with adulthood in Pima Indians. Diabetes 61, 250–257 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Tunç, S. et al. Melanocortin-4 receptor gene mutations in a group of Turkish obese children and adolescents. J. Clin. Res. Pediatr. Endocrinol. 9, 216–221 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Martinelli, C. E. et al. Obesity due to melanocortin 4 receptor (MC4R) deficiency is associated with increased linear growth and final height, fasting hyperinsulinemia, and incompletely suppressed growth hormone secretion. J. Clin. Endocrinol. Metab. 96, E181–E188 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Fry, A. et al. Comparison of sociodemographic and health-related characteristics of UK Biobank participants with those of the general population. Am. J. Epidemiol. 186, 1026–1034 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Majithia, A. R. et al. Prospective functional classification of all possible missense variants in PPARG. Nat. Genet. 48, 1570–1575 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chami, N., Preuss, M., Walker, R. W., Moscati, A. & Loos, R. J. F. The role of polygenic susceptibility to obesity among carriers of pathogenic mutations in MC4R in the UK Biobank population. PLoS Med. 17, e1003196 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Baker, J. L., Olsen, L. W. & Sørensen, T. I. A. Childhood body-mass index and the risk of coronary heart disease in adulthood. N. Engl. J. Med. 357, 2329–2337 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bjerregaard, L. G. et al. Change in overweight from childhood to early adulthood and risk of type 2 diabetes. N. Engl. J. Med. 378, 1302–1312 (2018).

    Article  PubMed  Google Scholar 

  38. Richardson, T. G., Sanderson, E., Elsworth, B., Tilling, K. & Davey Smith, G. Use of genetic variation to separate the effects of early and later life adiposity on disease risk: Mendelian randomisation study. BMJ 369, m1203 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Northstone, K. et al. The Avon Longitudinal Study of Parents and Children (ALSPAC): an update on the enrolled sample of index children in 2019. Wellcome Open Res. 4, 51 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Howe, L. D. et al. Changes in ponderal index and body mass index across childhood and their associations with fat mass and cardiovascular risk factors at age 15. PLoS ONE 5, e15186 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Wade, K. H. et al. Assessing the causal role of body mass index on cardiovascular health in young adults. Circulation 138, 2187–2201 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Lang, R. M. et al. Recommendations for chamber quantification: a report from the American Society of Echocardiography’s Guidelines and Standards Committee and the Chamber Quantification Writing Group, developed in conjunction with the European Association of Echocardiography, a branch of the European Society of Cardiology. J. Am. Soc. Echocardiogr. 18, 1440–1463 (2005).

    Article  PubMed  Google Scholar 

  43. Leckie, G. & Charlton, C. runmlwin: a program to run the MLwiN multilevel modeling software from within Stata. J. Stat. Softw. 52, 1–40 (2013).

    Google Scholar 

Download references

Acknowledgements

We are extremely grateful to all the families who took part in this study, the midwives for their help in recruiting them and the whole ALSPAC team, which includes interviewers, computer and laboratory technicians, clerical workers, research scientists, volunteers, managers, receptionists and nurses. We acknowledge the technical support of Tolulope Osunnuyi and the NIHR BRC-MRC BioRepository at Cambridge Biomedical Research Centre. We also thank staff members of the Wellcome Trust-MRC Institute of Metabolic Science Genomics and Transcriptomic core and the Genomics Core at the CRUK Cambridge Institute for their experimental support for next-generation sequencing. WES was obtained from ALSPAC (under proposal no. B2680) for comparison to the current study and we thank E. Robinson and B. Neale from the BROAD Institute for their contribution to exome sequencing. The UK MRC and Wellcome Trust (grant ref. 102215/2/13/2) and the University of Bristol provide core support for ALSPAC. Genome-wide association data were generated by Sample Logistics and Genotyping Facilities at Wellcome Sanger Institute and LabCorp (Laboratory Corporation of America) using support from 23andMe. Mutational screening, sequencing and functional analyses were supported by MRC Metabolic Diseases Unit funding (MC_UU_00014/1). This publication is the work of all authors and K.H.W., N.J.T. and S.O. serve as guarantors for the contents of this paper. K.H.W. was supported by the Elizabeth Blackwell Institute for Health Research, University of Bristol and the Wellcome Trust Institutional Strategic Support Fund (204813/Z/16/Z). N.J.T. is a Wellcome Trust Investigator (202802/Z/16/Z), a workpackage lead in the Integrative Cancer Epidemiology Programme that is supported by a Cancer Research UK programme grant (C18281/A19169) and works within the University of Bristol National Institute for Health Research Biomedical Research Centre. D.A.H. and L.J.C. are supported by N.J.T.’s Wellcome Trust Investigator grant (202802/Z/16/Z) and work within the MRC Integrative Epidemiology Unit (MC_UU_00011). B.Y.H.L. is supported by a BBSRC Project Grant (BB/S017593/1). A. Melvin holds a PhD studentship supported jointly by the University of Cambridge Experimental Medicine Training Initiative programme in partnership with AstraZeneca. I.S.F. was supported by the Wellcome Trust (098497/Z/12/Z), the NIHR Cambridge Biomedical Research Centre, the Botnar Fondation and the Bernard Wolfe Health Neuroscience Endowment and a Wellcome Developing Concept Fund award (with J.M.). S.O.R. and G.S.H.Y. is supported by the MRC Metabolic Disease Unit (MC_UU_00014/1) and S.O.R. by a Wellcome Trust Investigator award (WT 095515/Z/11/Z) and National Institute for Health Research Cambridge Biomedical Research Centre. The Wellcome-MRC Institute of Metabolic Science Genomics and transcriptomics core facility is supported by the Medical Research Council (MC_UU_00014/5) and the Wellcome Trust (208363/Z/17/Z).

Author information

Authors and Affiliations

Authors

Contributions

K.H.W., B.Y.H.L., A. Melvin, G.S.H.Y., N.J.T. and S.O.R. designed the study. C.Z., K.R. and K.D. conducted the genomic sequencing. J.M. and I.S.F. contributed to the design of in vitro assays. W.P., J.H.C., K.L., K.D., J.M. and A.W. planned and performed the in vitro studies. B.Y.H.L., A. Melvin and A. Mörseburg conducted bioinformatic analysis and analyzed the data from in vitro experiments. K.H.W. conducted the analysis of phenotype data in the ALSPAC cohort. L.J.C. critically reviewed the analysis in ALSPAC and the manuscript. D.A.H., K.N. and S.N. were involved in early conversations on the project and provided access to phenotypic, genotypic and exome data. K.H.W., B.Y.H.L., A. Melvin, G.S.H.Y., N.J.T. and S.O.R. wrote the manuscript and it was reviewed by all authors.

Corresponding authors

Correspondence to Nicholas J. Timpson or Stephen O’Rahilly.

Ethics declarations

Competing interests

S.O.R. has undertaken remunerated consultancy work for Pfizer, AstraZeneca, GSK and ERX Pharmaceuticals. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Medicine thanks Anke Hinney and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Jennifer Sargent was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Differences in body mass index in those sequenced for MC4R LoF mutations and those not sequenced from the rest of ALSPAC.

Estimates reflect the mean difference (95% CI) in BMI (kg/m2) comparing those sequenced for MC4R LoF mutations to those not sequenced, obtained from two-sided t-tests (p-values were not corrected for multiple comparisons). ALSPAC = Avon Longitudinal Study of Parents and Children; BMI = body mass index; CI = confidence interval; LoF = loss of function.

Extended Data Fig. 2 β-arrestin-2 functional classification of MC4R variants.

Colors represent cLoF (purple), pLoF (light brown), WT-like (green) and GoF (black). β-arrestin-2 coupling activity of MC4R mutations were characterised using a protein-protein interaction assay. a, Dose response curves of MC4R mutants upon activation by NDP-MSH grouped by cLoF, pLoF, WT-like and GoF compared wild-type MC4R. Means + /- S.E.M. are shown (N = 3–8 independent experiments); and b, The relative EC50 (fold WT response, left panel) and Emax (% WT response right panel) were determined for each mutants and are presented in mean and 95% CI. * indicates p < 0.05 by paired t-test (p-values were two-sided and uncorrected for multiple comparisons). CI = confidence interval; cLoF = complete loss of function; GoF = gain of function; N.D. = not determined; NDP-αMSH = [Nle4,D-Phe7]-α-melanocyte-stimulating hormone; pLoF = partial loss of function; WT-like = wild-type like.

Extended Data Fig. 3 Mean weight across time with MC4R LoF of cAMP accumulation.

Mean weight +/− 95% CI at different ages (sample sizes across all ages presented in Supplementary Table 4) with MC4R LoF of cAMP (carriers of pLoF and cLoF) and the reference group (that is, non-LoF carriers – combining individuals with synonymous, common variations or no LoF mutations and individuals with WT-like and GoF mutations). Figures only show results where all mutational groups (that is, WT-like, GoF, pLoF and cLoF mutations) were represented by at least one individual at all time points between birth and 24 years. CI = confidence interval; cLoF = complete loss of function; GoF = gain of function; LoF = loss of function; pLoF = partial loss of function; WT = wild-type.

Extended Data Fig. 4 Mean height across time with MC4R LoF of cAMP accumulation.

Mean height +/− 95% CI at different ages (sample sizes across ages presented in Supplementary Table 4) with MC4R LoF of cAMP (carriers of pLoF and cLoF) and the reference group (that is, non-LoF carriers – combining individuals with synonymous, common variations or no LoF mutations and individuals with WT-like and GoF mutations). Figures only show results where all mutational groups (that is, WT-like, GoF, pLoF and cLoF mutations) were represented by at least one individual at all time points between birth and 24 years. CI = confidence interval; cLoF = complete loss of function; GoF = gain of function; LoF = loss of function; pLoF = partial loss of function; WT = wild-type.

Extended Data Fig. 5 Association between MC4R LoF of cAMP accumulation and arterial BP across age in a model adjusting only for sex and a model adjusted for sex and BMI at the same age.

Estimates represent the change (mmHg) in SBP or DBP (top and bottom panel, respectively) in carriers (that is, pLoF or cLoF) vs. non-LoF carriers (that is, individuals with synonymous, common variations or no LoF mutations and individuals with WT-like and GoF mutations) of MC4R LoF mutations, obtained from linear regression (p-values presented are two-sided and not corrected for multiple comparisons). BMI = body mass index; BP = blood pressure; CI = confidence interval; cLoF = complete loss of function; DBP = diastolic blood pressure; GoF = gain of function; LoF = loss of function; pLoF = partial loss of function; SBP = systolic blood pressure; WT-like = wild-type like.

Extended Data Fig. 6 Association of MC4R LoF of cAMP accumulation with both central BP and LVMI at age 18 years in a model adjusting only for sex and a model adjusted for sex and BMI at the same age.

Estimates represent the change in central DBP (mmHg), central SBP (mmHg) and LVMI (g/m2.7) in carriers (that is, pLoF or cLoF) vs. non-LoF carriers (that is, individuals with synonymous, common variations or no LoF mutations and individuals with WT-like and GoF mutations) of MC4R LoF mutations, obtained from linear regression (p-values presented are two-sided and not corrected for multiple comparisons). BMI = body mass index; CI = confidence interval; cLoF = complete loss of function; DBP = diastolic blood pressure; GoF = gain of function; LoF = loss of function; LVMI = left ventricular mass index; pLoF = partial loss of function; SBP = systolic blood pressure; WT-like = wild-type like.

Extended Data Fig. 7 Association between MC4R LoF of cAMP accumulation with weight and height trajectories between birth and 18 years using linear spline multi-level models.

Values for the reference group (that is, all individuals with synonymous, common variations or no LoF mutations and individuals with WT-like and GoF mutations) and LoF mutations (that is, combining pLoF and cLoF mutations) are depicted in light and dark blue, respectively (N = 5716). Effect estimates and confidence intervals of these associations are presented in Supplementary Table 11 and Supplementary Table 13 for weight and height, respectively, and were generated using linear spline multi-level models. cLoF = complete loss of function; GoF = gain of function; LoF = loss of function; pLoF = partial loss of function; WT-like = wild-type like.

Extended Data Fig. 8 Mean BMI across time with MC4R LoF of β-arrestin-2 coupling.

Mean BMI +/− 95% CI at different ages (sample sizes across all ages presented in Supplementary Table 14) with MC4R LoF of β-arrestin-2 (carriers of pLoF and cLoF) and the reference group (that is, non-LoF carriers – combining individuals with synonymous, common variations or no LoF mutations and individuals with WT-like and GoF mutations). Figures only show results where all mutational groups (that is, WT-like, GoF, pLoF and cLoF mutations) were represented by at least one individual at all time points between birth and 24 years. CI = confidence interval; cLoF = complete loss of function; GoF = gain of function; LoF = loss of function; pLoF = partial loss of function; WT = wild-type.

Extended Data Fig. 9 Mean weight and height across time with MC4R LoF of β-arrestin-2 coupling.

Mean weight and height +/− 95% CI at different ages (sample sizes across all ages presented in Supplementary Table 14) with MC4R LoF of β-arrestin-2 (carriers of pLoF and cLoF) and the reference group (that is, non-LoF carriers – combining individuals with synonymous, common variations or no LoF mutations and individuals with WT-like and GoF mutations). Figures only show results where all mutational groups (that is, WT-like, GoF, pLoF and cLoF mutations) were represented by at least one individual at all time points between birth and 24 years. CI = confidence interval; cLoF = complete loss of function; GoF = gain of function; LoF = loss of function; pLoF = partial loss of function; WT = wild-type.

Extended Data Fig. 10 Identification of rare MC4R variants by pooled sequencing.

Pooled MC4R sequencing workflow for DNA samples from the ALSPAC cohort; b, Agarose gel showing a 1130 bp PCR product using MC4R exon primers (Supplementary Table 1), PCR products from 2 independent DNA samples are shown; c, A plot showing the sequencing coverage of MC4R coding region from a representative pool. The average per-base sequencing depth for all pools was 43,654 ± 356-fold; d, The percentage of true positive and false positive calls binned by variant allele frequency (VAF) detected in pools; e, Receiver operating curve analysis of VAF and call accuracy; f, Comparison of participants included in the current study and another whole-exome sequencing (WES) study (with ALSPAC project number: B2680); g, Comparison of non-synonymous variant carriages between this study and WES study. Of 40 mutational carriages found in Cambridge, 15 were found in both studies. 25 carriers found in the Cambridge study were not part of the WES study and 4 carriers found only in the WES study were not part of the Cambridge study. *rare = minor allele frequency < 0.01%, both p.V103I and p.I251L were above 0.01% and excluded in the analyses. ALSPAC = Avon Longitudinal Study of Parents and Children; AUC = area under the curve; HTS = high-throughput sequencing; WES = whole-exome sequencing.

Supplementary information

Supplementary Information

Supplementary Tables 1–20.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wade, K.H., Lam, B.Y.H., Melvin, A. et al. Loss-of-function mutations in the melanocortin 4 receptor in a UK birth cohort. Nat Med 27, 1088–1096 (2021). https://doi.org/10.1038/s41591-021-01349-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01349-y

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