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:

The rs1421085 variant within FTO promotes brown fat thermogenesis

Abstract

One lead genetic risk signal of obesity—the rs1421085 T>C variant within the FTO gene—is reported to be functional in vitro but lacks evidence at an organism level. Here we recapitulate the homologous human variant in mice with global and brown adipocyte-specific variant knock-in and reveal that mice carrying the C-allele show increased brown fat thermogenic capacity and resistance to high-fat diet-induced adiposity, whereas the obesity-related phenotypic changes are blunted at thermoneutrality. Both in vivo and in vitro data reveal that the C-allele in brown adipocytes enhances the transcription of the Fto gene, which is associated with stronger chromatin looping linking the enhancer region and Fto promoter. Moreover, FTO knockdown or inhibition effectively eliminates the increased thermogenic ability of brown adipocytes carrying the C-allele. Taken together, these findings identify rs1421085 T>C as a functional variant promoting brown fat thermogenesis.

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: The homologous rs1421085 T>C variant promotes energy expenditure and resists HFD-induced adiposity.
Fig. 2: The homologous rs1421085 T>C variant augments the thermogenic capacity of brown adipocytes.
Fig. 3: Brown adipocyte-specific knock-in of the homologous rs1421085 T>C variant enhances thermogenesis and resists HFD-induced adiposity.
Fig. 4: The homologous rs1421085 T>C variant increases thermogenesis via upregulating Fto expression.
Fig. 5: Thermoneutrality blunts the effects of the homologous rs1421085 T>C variant on energy expenditure and HFD-induced adiposity.

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in this article and its supplementary information files. Abbreviations and corresponding full names used in this study are listed in Supplementary Table 4. Source data are provided with this paper.

References

  1. Claussnitzer, M. et al. A brief history of human disease genetics. Nature 577, 179–189 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Wijmenga, C. & Zhernakova, A. The importance of cohort studies in the post-GWAS era. Nat. Genet. 50, 322–328 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. van der Klaauw, A. A. & Farooqi, I. S. The hunger genes: pathways to obesity. Cell 161, 119–132 (2015).

    Article  PubMed  Google Scholar 

  4. Zhu, F., Nair, R. R., Fisher, E. M. C. & Cunningham, T. J. Humanising the mouse genome piece by piece. Nat. Commun. 10, 1845 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Yengo, L. et al. Meta-analysis of genome-wide association studies for height and body mass index in 700000 individuals of European ancestry. Hum. Mol. Genet. 27, 3641–3649 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Locke, A. E. et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 518, 197–206 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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  PubMed  PubMed Central  Google Scholar 

  8. Dina, C. et al. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat. Genet. 39, 724–726 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Sovio, U. et al. Association between common variation at the FTO locus and changes in body mass index from infancy to late childhood: the complex nature of genetic association through growth and development. PLoS Genet. 7, e1001307 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Almeida, S. M. et al. Association between LEPR, FTO, MC4R, and PPARG-2 polymorphisms with obesity traits and metabolic phenotypes in school-aged children. Endocrine 60, 466–478 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Young, A. I., Wauthier, F. & Donnelly, P. Multiple novel gene-by-environment interactions modify the effect of FTO variants on body mass index. Nat. Commun. 7, 12724 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Smemo, S. et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature 507, 371–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wing, M. R. et al. Analysis of FTO gene variants with obesity and glucose homeostasis measures in the multiethnic Insulin Resistance Atherosclerosis Study cohort. Int. J. Obes. 35, 1173–1182 (2011).

    Article  CAS  Google Scholar 

  14. Adeyemo, A. et al. FTO genetic variation and association with obesity in West Africans and African Americans. Diabetes 59, 1549–1554 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hester, J. M. et al. Implication of European-derived adiposity loci in African Americans. Int. J. Obes. 36, 465–473 (2012).

    Article  CAS  Google Scholar 

  16. Loos, R. J. & Yeo, G. S. The bigger picture of FTO: the first GWAS-identified obesity gene. Nat. Rev. Endocrinol. 10, 51–61 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Mei, H. et al. FTO influences on longitudinal BMI over childhood and adulthood and modulation on relationship between birth weight and longitudinal BMI. Hum. Genet. 128, 589–596 (2010).

    Article  PubMed  Google Scholar 

  18. Couto Alves, A. et al. GWAS on longitudinal growth traits reveals different genetic factors influencing infant, child, and adult BMI. Sci. Adv. 5, eaaw3095 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Boissel, S. et al. Loss-of-function mutation in the dioxygenase-encoding FTO gene causes severe growth retardation and multiple malformations. Am. J. Hum. Genet. 85, 106–111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fischer, J. et al. Inactivation of the Fto gene protects from obesity. Nature 458, 894–898 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Wang, C. Y. et al. Loss of FTO in adipose tissue decreases Angptl4 translation and alters triglyceride metabolism. Sci. Signal 8, ra127 (2015).

    Article  PubMed  Google Scholar 

  22. McMurray, F. et al. Adult onset global loss of the fto gene alters body composition and metabolism in the mouse. PLoS Genet. 9, e1003166 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Claussnitzer, M. et al. FTO obesity variant circuitry and adipocyte browning in humans. N. Engl. J. Med. 373, 895–907 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sobreira, D. R. et al. Extensive pleiotropism and allelic heterogeneity mediate metabolic effects of IRX3 and IRX5. Science 372, 1085–1091 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Laber, S. et al. Linking the FTO obesity rs1421085 variant circuitry to cellular, metabolic, and organismal phenotypes in vivo. Sci. Adv. 7, eabg0108 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cohen, P. & Kajimura, S. The cellular and functional complexity of thermogenic fat. Nat. Rev. Mol. Cell Biol. 22, 393–409 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, J. et al. Ablation of LGR4 promotes energy expenditure by driving white-to-brown fat switch. Nat. Cell Biol. 15, 1455–1463 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Stratigopoulos, G., LeDuc, C. A., Cremona, M. L., Chung, W. K. & Leibel, R. L. Cut-like homeobox 1 (CUX1) regulates expression of the fat mass and obesity-associated and retinitis pigmentosa GTPase regulator-interacting protein-1-like (RPGRIP1L) genes and coordinates leptin receptor signaling. J. Biol. Chem. 286, 2155–2170 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Stratigopoulos, G. et al. Regulation of Fto/Ftm gene expression in mice and humans. Am. J. Physiol. Regul. Integr. Comp. Physiol. 294, R1185–R1196 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Jerkovic, I. & Cavalli, G. Understanding 3D genome organization by multidisciplinary methods. Nat. Rev. Mol. Cell Biol. 22, 511–528 (2021).

    Article  CAS  PubMed  Google Scholar 

  31. Peng, S. et al. Identification of entacapone as a chemical inhibitor of FTO mediating metabolic regulation through FOXO1. Sci. Transl. Med. 11, eaau7116 (2019).

    Article  PubMed  Google Scholar 

  32. Ohno, H., Shinoda, K., Ohyama, K., Sharp, L. Z. & Kajimura, S. EHMT1 controls brown adipose cell fate and thermogenesis through the PRDM16 complex. Nature 504, 163–167 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Su, R. et al. R-2HG exhibits anti-tumor activity by targeting FTO/m(6)A/MYC/CEBPA signaling. Cell 172, 90–105 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Symonds, M. E., Mostyn, A., Pearce, S., Budge, H. & Stephenson, T. Endocrine and nutritional regulation of fetal adipose tissue development. J. Endocrinol. 179, 293–299 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Cypess, A. M. et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 360, 1509–1517 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ganeshan, K. & Chawla, A. Warming the mouse to model human diseases. Nat. Rev. Endocrinol. 13, 458–465 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Velickovic, K. et al. Expression and subcellular localization of estrogen receptors α and β in human fetal brown adipose tissue. J. Clin. Endocrinol. Metab. 99, 151–159 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Jastroch, M., Oelkrug, R. & Keipert, S. Insights into brown adipose tissue evolution and function from non-model organisms. J. Exp. Biol. 221, jeb169425 (2018).

    Article  PubMed  Google Scholar 

  39. Ravussin, E. & Galgani, J. E. The implication of brown adipose tissue for humans. Annu. Rev. Nutr. 31, 33–47 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Herman, M. A. & Rosen, E. D. Making biological sense of GWAS data: lessons from the FTO locus. Cell Metab. 22, 538–539 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. O'Rahilly, S., Coll, A. P. & Yeo, G. S. FTO obesity variant and adipocyte browning in humans. N. Engl. J. Med. 374, 191 (2016).

    PubMed  Google Scholar 

  42. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sun, Y. et al. Human RSPO1 mutation represses beige adipocyte thermogenesis and contributes to diet-induced adiposity. Adv. Sci. 10, e2207152 (2023).

    Article  Google Scholar 

  44. Cannon, B. & Nedergaard, J. Nonshivering thermogenesis and its adequate measurement in metabolic studies. J. Exp. Biol. 214, 242–253 (2011).

    Article  PubMed  Google Scholar 

  45. Lean, M. E., James, W. P., Jennings, G. & Trayhurn, P. Brown adipose tissue uncoupling protein content in human infants, children and adults. Clin. Sci. 71, 291–297 (1986).

    Article  CAS  Google Scholar 

  46. Descamps, O. S., Tarantino, E. & Guilmot, P. F. Does FTO have a paradoxical effect in fetal life? BMC Genet. 15, 145 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Monda, K. L. et al. A meta-analysis identifies new loci associated with body mass index in individuals of African ancestry. Nat. Genet. 45, 690–696 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Peters, U. et al. A systematic mapping approach of 16q12.2/FTO and BMI in more than 20,000 African Americans narrows in on the underlying functional variation: results from the Population Architecture using Genomics and Epidemiology (PAGE) study. PLoS Genet. 9, e1003171 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cannon, B. & Nedergaard, J. Brown adipose tissue: function and physiological significance. Physiol. Rev. 84, 277–359 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Clemente, F. J. et al. A selective sweep on a deleterious mutation in CPT1A in arctic populations. Am. J. Hum. Genet. 95, 584–589 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kamberov, Y. G. et al. Modeling recent human evolution in mice by expression of a selected EDAR variant. Cell 152, 691–702 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Huerta-Sánchez, E. et al. Altitude adaptation in Tibetans caused by introgression of Denisovan-like DNA. Nature 512, 194–197 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Zhang, Z. et al. The rs1421085 variant within FTO promotes but not inhibits thermogenesis and is potentially associated with human migration. Preprint at bioRxiv https://doi.org/10.1101/2021.08.13.456245 (2021).

  54. Cecil, J. E., Tavendale, R., Watt, P., Hetherington, M. M. & Palmer, C. N. An obesity-associated FTO gene variant and increased energy intake in children. N. Engl. J. Med. 359, 2558–2566 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Speakman, J. R., Rance, K. A. & Johnstone, A. M. Polymorphisms of the FTO gene are associated with variation in energy intake, but not energy expenditure. Obesity 16, 1961–1965 (2008).

    Article  CAS  PubMed  Google Scholar 

  56. McCaffery, J. M. et al. Obesity susceptibility loci and dietary intake in the Look AHEAD Trial. Am. J. Clin. Nutr. 95, 1477–1486 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ong, K. K., Ahmed, M. L., Emmett, P. M., Preece, M. A. & Dunger, D. B. Association between postnatal catch-up growth and obesity in childhood: prospective cohort study. Brit. Med. J. 320, 967–971 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zou, Y. et al. IRX3 promotes the browning of white adipocytes and its rare variants are associated with human obesity risk. eBioMedicine 24, 64–75 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Zhang, Z. et al. IRX3 overexpression enhances Ucp1 expression in vivo. Front. Endocrinol. 12, 634191 (2021).

    Article  Google Scholar 

  60. Hozumi, A. et al. Enhancer activity sensitive to the orientation of the gene it regulates in the chordate genome. Dev. Biol. 375, 79–91 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Mouri, K. et al. Enhancer adoption caused by genomic insertion elicits interdigital Shh expression and syndactyly in mouse. Proc. Natl Acad. Sci. USA 115, 1021–1026 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Manghwar, H. et al. CRISPR/Cas systems in genome editing: methodologies and tools for sgRNA design, off-target evaluation, and strategies to mitigate off-target effects. Adv. Sci. 7, 1902312 (2020).

    Article  CAS  Google Scholar 

  63. Tschöp, M. H. et al. A guide to analysis of mouse energy metabolism. Nat. Methods 9, 57–63 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Rahbani, J. F. et al. Creatine kinase B controls futile creatine cycling in thermogenic fat. Nature 590, 480–485 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhang, K. X. et al. Violet-light suppression of thermogenesis by opsin 5 hypothalamic neurons. Nature 585, 420–425 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhi, X. et al. AdipoCount: a new software for automatic adipocyte counting. Front. Physiol. 9, 85 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Hagège, H. et al. Quantitative analysis of chromosome conformation capture assays (3C-qPCR). Nat. Protoc. 2, 1722–1733 (2007).

    Article  PubMed  Google Scholar 

  68. Chen, M. et al. CTNNB1/β-catenin dysfunction contributes to adiposity by regulating the cross-talk of mature adipocytes and preadipocytes. Sci. Adv. 6, eaax9605 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Williamson, D. F., Parker, R. A. & Kendrick, J. S. The box plot: a simple visual method to interpret data. Ann. Intern. Med. 110, 916–921 (1989).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all the participants for their involvement in this study and we thank J. Shen and Q. Liao from BGI-Shenzhen for their comments on the adaptive evolution of the FTO variant. This work was supported by grants from the National Key Research and Development Program of China (2022YFC2505201 to J.W. and 2021YFA1301103 to G.N.), the National Natural Science Foundation of China (92157204 to R.L., 91957124 to J.W., 82088102 to G.N., 82250901 to R.L. and 81930021 to G.N.), Shanghai Municipal Education Commission-Gaofeng Clinical Medicine Grant Support (20161306 and 20171903 Round 2 to R.L.), innovative research teams of high-level local universities in Shanghai (J.W. and G.N.), the Outstanding Academic Leader Program of Shanghai Municipal Health Commission (2018BR01 to J.W.), Science and Technology Commission of Shanghai Municipality (21JC1404400 to G.N.) and the Program of Shanghai Academic/Technology Research Leader (20XD1403200 to J.W. and 23XD1422400 to R.L.).

Author information

Authors and Affiliations

Authors

Contributions

J.W. designed the experiments and supervised the study. Z.Z., N.C., N.Y. and Y.H. carried out animal and molecular experiments. J.W. and R.L. analyzed all the experimental data. D.L., M.T., A.G., P.L., Y.Z. and H.L. provided support in animal model tests. J.Z. provided support in 3C tests. D.Z. collected fetal BAT samples. W.G., J.H. and W.W. provided support for the genetic analysis. G.N. and L.Q. contributed to text revision and discussion. J.W., R.L., Z.Z., N.C. and N.Y. wrote the paper.

Corresponding author

Correspondence to Jiqiu Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks George Stratigopoulos and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Isabella Samuelson, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 The sequencing of the homologous rs1421085 T>C variant in three knock-in mice.

(A to B) Sanger sequence analysis of KIcas9 (A) and KIES (B) mice, respectively. (C) Loxp (left, top), Cre expression (left, bottom), BAT DNA sequence analysis of KIfl/fl and Ucp1-KIfl/fl mice (right). The data is representative of least two independent experiments.

Extended Data Fig. 2 The homologous rs1421085 T>C variant protests HFD-induced obesity but shows no change in body weight under chow diet in KIcas9 mice.

(A) Body weight curve of male KIcas9 mice and wild-type (WT) littermates under normal chow diet (NCD). n = 10 (WT) and 15 (KIcas9) mice per group. (B to K) IPGTT (B) and ITT (C), TG (D), TC (E), HDL-c (F), and LDL-c (G), H&E staining of iWAT, eWAT, and liver (H), the ratio of fat mass and lean mass to body weight (I), the tissue mass weight (BAT, iWAT, and eWAT) (J), and their ratio to body weight (K) in male KIcas9 mice and WT littermates under HFD. For B and C, n = 17 per group. For D to G, n = 10 mice per group. For H, images are representative of 4 mice per group; iWAT: n = 180 (WT) and 315 (KIcas9) LDs; eWAT: n = 201 (WT) and 223 (KIcas9) LDs; liver: n = 24 (WT) and 20 (KIcas9) regions; scale bar = 200 μm. For I, n = 11 (WT) and 16 (KIcas9) mice per group. For J, BAT: n = 11 (WT) and 15 (KIcas9); iWAT: n = 11 (WT) and 15 (KIcas9); eWAT: n = 10 (WT) and 14 (KIcas9). For K, BAT: n = 11 (WT) and 15 (KIcas9); iWAT: n = 11 (WT) and 15 (KIcas9); eWAT: n = 8 (WT) and 14 (KIcas9). (L) The validation of diet-induced adiposity phenotype of male KIcas9 mice (F3 generation) under HFD challenge initiated from 11 to 27 weeks old. For HFD week 0 to 13, n = 18 (WT) and 19 (KIcas9) mice per group, for HFD week 14 to 16, n = 17 (WT, one died before HFD week 14) and 19 (KIcas9) mice per group. P= 0.0224, 0.0275, 0.0307, 0.0217, 0.0125, 0.0247, 0.008, 0.0244, 0.0266 between groups, at each week of HFD week 8 to 16 (* P < 0.05). (M) Body weight curve of female KIcas9 and WT littermate mice under NCD. n = 21 (WT) and 20 (KIcas9) mice per group. (N to R) Body weight curve (N), body mass (O), the ratio of lean mass to body weight (P), the tissue weights (BAT, iWAT, and eWAT) (Q), and their ratio to body weight (R) in female KIcas9 mice and WT littermates under HFD. For N, n = 9 (WT) and 8 (KIcas9) mice per group, P = 0.0326, 0.0271, 0.0435, 0.0269, 0.0302, 0.0496, 0.0413, 0.0451, 0.0375, 0.0299 at week 1, 2, 3, 4, 6, 7, 8, 10, 11, 12 of HFD between groups (* P < 0.05). For O to P, n = 8 mice per group. For Q to R, n = 9 (WT) and 8 (KIcas9) mice per group. Statistical significance was assessed by unpaired two-sided Student’s t-test (A to R). Data are mean ± s.e.m. of biologically independent samples. Data are representative of at least two independent experiments (B to G).

Source data

Extended Data Fig. 3 The homologous rs1421085 T>C variant protests HFD-induced obesity but shows no change in body weight under chow diet in KIES mice.

(A) Body weight of male KIES mice and WT littermates aged 8- and 23-week-old under NCD. n = 8 mice per group. (B) Body weight curve of female KIES mice and WT littermates under NCD. n = 8 mice per group. (C) The ratio of fat mass and lean mass to body weight in male KIES and WT mice of HFD. n = 12 (WT) and 13 (KIES) mice per group. (D to F) The tissue weights (BAT, iWAT, and eWAT) (D) and their ratio to body weight (E), H&E staining (iWAT, eWAT, and liver) (F) in male KIES mice and WT littermates under HFD. For D and E, n = 12 (WT) and 13 (KIES) mice per group. For F, images are representative of 4 mice per group; iWAT: n = 1082 (WT) and 1019 (KIES) LDs; eWAT: n = 235 (WT) and 310 (KIES) LDs; liver: n = 16 regions per group; scale bar = 200 μm. (G to K) Body weight curve (G), the fat mass and lean mass (H) and their ratio to body weight (I), the tissue weights (BAT, iWAT, eWAT, and liver) (J) and their ratio to body weight (K) of female KIES and WT littermate mice fed HFD from 10- to 24-week-old. For G, n = 13 (WT) and 10 (KIES) mice per group, P = 0.0005, 0.0116, 0.0433, 0.0159, 0.0220, 0.0197, 0.0296, 0.0302, 0.0074, 0.0361, 0.0339, 0.0191, 0.0165, 0.0303, 0.0335 between groups, at each week of HFD week 0 to 14 (* P < 0.05). For H, J and K, n = 13 (WT) and 10 (KIES) mice per group. For I, fat mass: n = 12 (WT) and 10 (KIES); lean mass: n = 13 (WT) and 10 (KIES) mice per group. (L to M) Respiratory exchange ratio (RER) (L) and total physical activities (M) of male KIES and WT mice. Data were collected after the first week of HFD feeding, n = 8 mice per group. (N to O) H&E staining (BAT, iWAT, eWAT, and liver) (N) and transmission electron microscopy (TEM) morphology (BAT) (O) of female KIES and WT mice fed HFD from 10- to 24-week-old. For N, images are representative of 4 mice per group; BAT: n = 353 (WT) and 287 (KIES) LDs; iWAT: n = 199 (WT) and 199 (KIES) LDs; eWAT: n = 247 (WT) and 274 (KIES) LDs; liver: n = 4 regions per group; scale bar = 200 μm. For O, n = 7 (WT) and 6 (KIES); scale bar = 1 μm. Statistical significance was assessed by unpaired two-sided Student’s t-test (A to O). Data are mean ± s.e.m. of biologically independent samples, representative of at least two independent experiments.

Source data

Extended Data Fig. 4 The brown adipocyte-specific rs1421085 T>C knock-in (Ucp1-KIfl/fl) mice resist HFD-induced adiposity.

(A to E) The mRNA expression of Fto (A), Irx3 (B), Irx5 (C), Irx6 (D), and Rpgrip1l (E) in the tissues related to energy homeostasis in 4–5 weeks old male homologous rs1421085fl/fl (KIfl/fl) and WT mice. For A, BAT: n = 11 (WT) and 9 (KIES); iWAT, eWAT, liver, muscle and hypothalamus: n = 11 (WT) and 10 (KIES). For B, BAT and iWAT: n = 11 (WT) and 10 (KIES); eWAT, liver, muscle and hypothalamus: n = 11 per group. For C, BAT: n = 10 (WT) and 11 (KIES); iWAT: n = 10 (WT) and 6 (KIES); eWAT: n = 11 per group; liver: n = 10 (WT) and 11 (KIES); muscle: n = 11 (WT) and 10 (KIES); hypothalamus: n = 11 per group. For D, BAT: n = 11 (WT) and 10 (KIES); iWAT, eWAT, liver, muscle and hypothalamus: n = 11 per group. For E, BAT: n = 11 (WT) and 10 (KIES); iWAT, eWAT, liver, muscle, hypothalamus: n = 11 per group. (F to I) Body weight curve (F), the ratio of fat mass and lean mass to body weight (G), the tissue mass (BAT, iWAT, and eWAT) and their ratio to body weight (H and I) in male Ucp1-KIfl/fl and KIfl/fl mice under NCD. For F, n = 10 mice per group. For G and I, n = 12 (KIfl/fl) and 7 (Ucp1-KIfl/fl) mice per group. For H, n = 11 (KIfl/fl) and 7 (Ucp1-KIfl/fl) mice per group. (J) The representative H&E images of iWAT, eWAT, and liver in male Ucp1-KIfl/fl and KIfl/fl mice under HFD; images are representative of 4 mice per group; iWAT: n = 194 (KIfl/fl) and 221 (Ucp1-KIfl/fl) LDs; eWAT: n= 274 (KIfl/fl) and 187 (Ucp1-KIfl/fl) LDs; liver: n = 16 regions per group; scale bar = 200 μm. (K to M) Cumulative food intake (K), RER (L), and total physical activities (M) of single-housed 10-week-old male KIES and WT mice. For K, n = 9 mice per group. For L and M, n = 10 mice per group. Data were collected after the first week of HFD feeding. Statistical significance was assessed by unpaired two-sided Student’s t-test (A to J and L to M) and two-way ANOVA followed by Bonferroni post tests (K). Data are mean ± s.e.m. of biologically independent samples, representative of at least two independent experiments. Exclusion criteria for outliers please see Methods.

Source data

Extended Data Fig. 5 The mRNA expression of candidate target genes of the rs1421085 T>C variant in the knock-in mouse models.

(A to D) The mRNA expression of Irx3, Irx5, Irx6, and Rpgrip1l in male BAT tissues of three knock-in mouse models and induced mature brown adipocytes. For A, n = 6 (WT) and 7 (KIcas9) mice per group. For B, n = 12 (WT) and 13 (KIES) for Irx3; n = 7 (WT) and 13 (KIES) for Irx5, Irx6 and Rpgrip1l. For C, n = 15 (KIfl/fl) and 7 (Ucp1-KIfl/fl) for Irx3, Irx6 and Rpgrip1l; n = 15 (KIfl/fl) and 6 (Ucp1-KIfl/fl) for Irx5. For D, n = 4 (WT) and 3 (KIES) per group. Statistical significance was assessed by unpaired two-sided Student’s t-test (A to D). Data are mean ± s.e.m. of biologically independent samples, representative of at least two independent experiments.

Source data

Extended Data Fig. 6 The expression and demethylase activity of FTO affects brown adipocyte thermogenesis.

(A) The mRNA expression of Fto and thermogenesis-related genes at different time points during brown adipocytes differentiation derived from male WT BAT SVFs, n = 6 (Day 2 to Day 6) or 5 (Day 8). (B) Correlation analysis of the mRNA levels of Fto and Ucp1, n = 24. (C and D) Oil Red O staining (C) and OCR measurement (D) of induced brown adipocytes derived from male WT BAT SVFs after Fto knockdown. For C, images are representative of 3 independent biological samples, scale bar = 100 μm; for D, n = 5 (shNS, shFTO-1 and shFTO-2) or 4 (shFTO-3) per group. shNS: control virus. (E) Oil Red O staining of induced brown adipocytes derived from BAT SVFs with Fto overexpression, images are representative of 3 independent biological samples, scale bar = 100 μm. EV: control virus. (F) The m6A abundance in induced mature brown adipocytes in the presence of Entacapone (ENT, 50 μM) or DMSO, n = 3 per group. (G) The mRNA expression of thermogenesis-related genes in induced mature brown adipocytes in the presence of ENT (50 μM) or DMSO, for DMSO, n = 3 (Fto) or 4 (Ucp1, Pgc-1α, Prdm16, Dio2, Cidea, Cox7a, Cox8b) per group; for ENT, n = 4 per group. (H) The protein levels of FTO and UCP1 in induced mature brown adipocytes in the presence of ENT (50 μM) or DMSO. n = 3 per group. For C to E, the SVFs were infected with lentivirus two days before induction (MOI = 50). Statistical significance was assessed by unpaired two-sided Student’s t-test (D, F to H). Data are mean ± s.e.m. of biologically independent samples, representative of at least two independent experiments.

Source data

Extended Data Fig. 7 The Fto deficiency in BAT leads to HFD-induced adiposity.

(A) The mRNA expression of Fto in indicated adipose tissues in 4-week-old female Ftofl/fl; Myf5-cre (MFKO) mice and Ftofl/fl littermates. n = 3 per group for BAT; n = 3 (Ftofl/fl) and 4 (MFKO) for iWAT; n = 4 per group for eWAT. (B to E) Body weight and body weight gain curve (B), body mass (C) and their ratio to body weight (D), and average food intake (E) of male MFKO mice and littermate controls under HFD challenge initiating from 9 to 19 weeks old. For B, n = 8 (Ftofl/fl) and 12 (MFKO) mice per group. For C and D, n = 8 mice per group. For E, n = 4 (Ftofl/fl) and 3 (MFKO) mice per group. (F to I) Representative 24 h O2 consumption (F) and CO2 production (G), average RER (H), EE with body weight as a covariant (I) of single-housed 21-week-old male MFKO and Ftofl/fl mice under 12 weeks of HFD, n = 8 per group. (J and K) Representative images of H&E staining (J) and TEM images (K) in BAT of male MFKO mice and Ftofl/fl littermates. Images are representative of 4 mice per group, scale bar = 200 μm (J) and 1 μm (K). (L) The mRNA expression of myogenesis-related genes in quadriceps femoris of male MFKO mice and littermate controls under NCD. n = 10 (Ftofl/fl) and 7 (MFKO) for MyoD; n = 9 (Ftofl/fl) and 7 (MFKO) for MyoG . (M) The protein expression of FTO, UCP1, and PGC-1α in induced mature brown adipocytes derived from BAT SVFs of male MFKO and littermate control mice, n = 4 per group. Statistical significance was assessed by unpaired two-sided Student’s t-test (A, C to E, H, L and M), and two-way ANOVA followed by Bonferroni post tests (B, F and G) and two-sided ANCOVA (I). The data were presented as means ± s.e.m. of biologically independent samples, representative of at least two independent experiments.

Source data

Extended Data Fig. 8 FTO protein stabilizes Ucp1 mRNA and increases UCP1 protein expression.

(A to D) RNA Immunoprecipitation (RIP)-qPCR analysis of the interaction of FTO protein and target mRNA in induced mature brown adipocytes. Representative immunoblots showing the products of IP by FTO antibody, with IgG as a negative control (A), agarose gel electrophoresis (AGE) (B), and qPCR analysis of Ucp1 (C) and C/ebpα (D) mRNA abundance of FTO-IP products, n = 4 per group. The enrichment of Ucp1 and C/ebpα mRNA was normalized to 7.7% input. (E and F) Half-life analysis of Ucp1 (E) and C/ebpα (F) mRNA in induced mature brown adipocytes with Fto knockdown. The male WT BAT SVFs were infected with lentivirus two days before induction (MOI = 75). (G and H) Half-life analysis of Ucp1 (G) and C/ebpα (H) mRNA in induced mature brown adipocytes derived from BAT SVFs of male KIES and WT mice. Actinomycin D (5 μg/ml) was added on the eighth day of induction; total RNA was isolated at indicated time points; calculated half-times t1/2 = In2/Kdecay. For E, n = 4 (0 h, 0.5 h, 2 h) or 3 (1 h) for shNC; n = 3 (0 h, 0.5 h, 2 h) or 4 (1 h) for shFTO. For F to H, n = 4 per group. (I) The protein expression and quantification analysis of FTO and UCP1 in induced mature brown adipocytes derived from BAT SVFs of male KIES and WT littermate mice in the presence of ENT (50 μM) or DMSO, n = 3 per group. Statistical significance was assessed by unpaired two-sided Student’s t-test. Data are mean ± s.e.m. of biologically independent samples, representative of at least two independent experiments.

Source data

Extended Data Fig. 9 Thermoneutrality (30 °C) blunts the effects of the brown adipocyte-specific knock-in of the rs1421085 T>C variant on energy expenditure and HFD-induced adiposity under sub-thermoneutrality (22 °C).

(A to C) Average RER (A), total physical activities (B), and cumulative food intake (C) measured in single-housed 22-week-old male KIES and WT mice after 7-week room temperature (22 °C) or thermoneutrality (30 °C) housing under HFD. For A and B, n = 8 per group at 22 °C; n = 7 (WT) and 8 (KIES) at 30 °C. For C, n = 7 per group at 22 °C; n = 6 (WT) and 7 (KIES) at 30 °C. (D to F) Body weight gain curve (D), tissue weights (E), and the representative images of H&E staining (F) in male Ucp1-KIfl/fl mice and KIfl/fl littermates under HFD at 22 °C and 30 °C. For D, n = 4 (22 °C) and 7 (30 °C) mice per group, P = 0.0469, 0.0485 between groups at week 3 to 4 at 22 °C, respectively (* P < 0.05). For E, n = 4 (22 °C) and 6 (30 °C) mice per group. For F, images are representative of 4 mice per group; BAT: n = 375 (KIfl/fl) and 298 (Ucp1-KIfl/fl) LDs at 22 °C, n = 476 (KIfl/fl) and 452 (Ucp1-KIfl/fl) LDs at 30 °C; iWAT: n = 214 (KIfl/fl) and 477 (Ucp1-KIfl/fl) LDs at 22 °C, n = 495 (KIfl/fl) and 461 (Ucp1-KIfl/fl) LDs at 30 °C; eWAT: n = 123 (KIfl/fl) and 253 (Ucp1-KIfl/fl) LDs at 22 °C, n = 239 (KIfl/fl) and 227 (Ucp1-KIfl/fl) LDs at 30°C; liver: n = 8 regions per group; scale bar = 200 μm. Statistical significance was assessed by unpaired two-sided Student’s t-test (A, B, D to F) and two-way ANOVA followed by Bonferroni post tests (C). Data are mean ± s.e.m. of biologically independent samples, representative of at least two independent experiments.

Source data

Extended Data Fig. 10 FTO and UCP1 proteins are co-localized in human fetal brown adipocytes.

Representative immunofluorescence (IF) images of FTO (green) and UCP1 (red) in human fetal brown adipocytes, in which the nuclei are stained with DAPI (blue) and LDs are marked with PLIN1 (pink). Left to right panels: FTO, UCP1, PLIN1, PLIN1/FTO/UCP1/DAPI merged (scale bar = 100 μm), and PLIN1/FTO/UCP1/DAPI merged (scale bar = 20 μm).

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–4.

Source data

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 2

Unprocessed western blots for Fig. 2.

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 3

Unprocessed western blots for Fig. 3.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 4

Unprocessed western blots for Fig. 4.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Unprocessed western blots for Extended Data Fig. 6.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Unprocessed western blots for Extended Data Fig. 7.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Unprocessed western blots for Extended Data Fig. 8.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Z., Chen, N., Yin, N. et al. The rs1421085 variant within FTO promotes brown fat thermogenesis. Nat Metab 5, 1337–1351 (2023). https://doi.org/10.1038/s42255-023-00847-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-023-00847-2

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