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:

RIPK1 gene variants associate with obesity in humans and can be therapeutically silenced to reduce obesity in mice

A Publisher Correction to this article was published on 08 October 2020

This article has been updated

Abstract

Obesity is a major public health burden worldwide and is characterized by chronic low-grade inflammation driven by the cooperation of the innate immune system and dysregulated metabolism in adipose tissue and other metabolic organs. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a central regulator of inflammatory cell function that coordinates inflammation, apoptosis and necroptosis in response to inflammatory stimuli. Here we show that genetic polymorphisms near the human RIPK1 locus associate with increased RIPK1 gene expression and obesity. We show that one of these single nucleotide polymorphisms is within a binding site for E4BP4 and increases RIPK1 promoter activity and RIPK1 gene expression in adipose tissue. Therapeutic silencing of RIPK1 in vivo in a mouse model of diet-induced obesity dramatically reduces fat mass, total body weight and improves insulin sensitivity, while simultaneously reducing macrophage and promoting invariant natural killer T cell accumulation in adipose tissue. These findings demonstrate that RIPK1 is genetically associated with obesity, and reducing RIPK1 expression is a potential therapeutic approach to target obesity and related diseases.

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: Association of human SNPs with RIPK1 in obesity.
Fig. 2: Relationship between E4BP4 and RIPK1 in adipose tissue and regulation by the polymorphism rs5873855.
Fig. 3: Therapeutic knockdown of Ripk1 in a diet-induced obesity mouse model.
Fig. 4: Knockdown of Ripk1 decreases inflammation in liver and adipose tissue.
Fig. 5: Adipose tissue invariant NKT cells are increased in mice treated with Ripk1 ASO.

Similar content being viewed by others

Data availability

Human and mouse scRNA-seq data have been deposited in the GEO database (https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE151889. Other publicly available data used in this study are available at the GTEx database, or under GEO accession numbers GSE36032 and GSE63358 (mouse) and GSE115469 (human liver). All other data that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

Change history

  • 08 October 2020

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Berg, A. H. & Scherer, P. E. Adipose tissue, inflammation and cardiovascular disease. Circ. Res. 96, 939–949 (2005).

    CAS  PubMed  Google Scholar 

  2. Weisberg, S. P. et al. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 112, 1796–1808 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Stunkard, A. J., Foch, T. T. & Hrubec, Z. A twin study of human obesity. JAMA 256, 51–54 (1986).

    CAS  PubMed  Google Scholar 

  4. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Gerken, T. et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science 318, 1469–1472 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Scuteri, A. et al. Genome-wide association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genet. 3, e115 (2007).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  8. 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 (Silver Spring) 16, 1961–1965 (2008).

    CAS  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  10. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. 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).

    CAS  PubMed  Google Scholar 

  12. Nikpay, M., Turner, A. W. & McPherson, R. Partitioning the pleiotropy between coronary artery disease and body mass index reveals the importance of low frequency variants and central nervous system-specific functional elements. Circ. Genom. Precis. Med. 11, e002050 (2018).

    CAS  PubMed  Google Scholar 

  13. Karunakaran, D. et al. Targeting macrophage necroptosis for therapeutic and diagnostic interventions in atherosclerosis. Sci. Adv. 2, e1600224 (2016).

    PubMed  PubMed Central  Google Scholar 

  14. Silke, J., Rickard, J. A. & Gerlic, M. The diverse role of RIP kinases in necroptosis and inflammation. Nat. Immunol. 16, 689–697 (2015).

    CAS  PubMed  Google Scholar 

  15. Kondylis, V., Kumari, S., Vlantis, K. & Pasparakis, M. The interplay of IKK, NFκB and RIPK1 signaling in the regulation of cell death, tissue homeostasis and inflammation. Immunol. Rev. 277, 113–127 (2017).

    CAS  PubMed  Google Scholar 

  16. Civelek, M. et al. Genetic regulation of adipose gene expression and cardio-metabolic traits. Am. J. Hum. Genet. 100, 428–443 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Laakso, M. et al. The Metabolic Syndrome in Men study: a resource for studies of metabolic and cardiovascular diseases. J. Lipid. Res. 58, 481–493 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. The GTEx Consortium. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

    Google Scholar 

  19. Davies, R. W. et al. A 680-kb duplication at the FTO locus in a kindred with obesity and a distinct body fat distribution. Eur. J. Hum. Genet. 21, 1417–1422 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. van Rheenen, W., Peyrot, W. J., Schork, A. J., Lee, S. H. & Wray, N. R. Genetic correlations of polygenic disease traits: from theory to practice. Nat. Rev. Genet. 20, 567–581 (2019).

  21. Pingault, J. B. et al. Using genetic data to strengthen causal inference in observational research. Nat. Rev. Genet. 19, 566–580 (2018).

    CAS  PubMed  Google Scholar 

  22. Zhu, Z. et al. Causal associations between risk factors and common diseases inferred from GWAS summary data. Nat. Commun. 9, 224 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Westra, H. J. et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat. Genet. 45, 1238–1243 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. van Nas, A. et al. The systems genetics resource: a web application to mine global data for complex disease traits. Front. Genet. 4, 84 (2013).

    PubMed  PubMed Central  Google Scholar 

  25. Ward, L. D. & Kellis, M. HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res. 40, D930–D934 (2011).

    PubMed  PubMed Central  Google Scholar 

  26. Cowell, I. G. E4BP4/NFIL3, a PAR-related bZIP factor with many roles. Bioessays 24, 1023–1029 (2002).

    CAS  PubMed  Google Scholar 

  27. Lynch, L. et al. Regulatory iNKT cells lack expression of the transcription factor PLZF and control the homeostasis of Treg cells and macrophages in adipose tissue. Nat. Immunol. 16, 85–95 (2015).

    CAS  PubMed  Google Scholar 

  28. Roy, S. et al. Redefining the transcriptional regulatory dynamics of classically and alternatively activated macrophages by deepCAGE transcriptomics. Nucleic Acids Res. 43, 6969–6982 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Takahashi, S. et al. A promoter in the novel exon of hPPARγ directs the circadian expression of PPARγ. J. Atheroscler. Thromb. 17, 73–83 (2010).

    CAS  PubMed  Google Scholar 

  30. Lynch, L. et al. Adipose tissue iNKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity 37, 574–587 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ji, Y. et al. Activation of natural killer T cells promotes M2 macrophage polarization in adipose tissue and improves systemic glucose tolerance via IL-4/STAT6 protein signaling axis in obesity. J. Biol. Chem. 287, 13561–13571 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Jaitin, D. A. et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell 178, 686–698(2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. MacParland, S. A. et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat. Commun. 9, 4383 (2018).

    PubMed  PubMed Central  Google Scholar 

  34. Kenna, T. et al. NKT cells from normal and tumor-bearing human livers are phenotypically and functionally distinct from murine NKT cells. J. Immunol. 171, 1775–1779 (2003).

    CAS  PubMed  Google Scholar 

  35. Fengler, V. H. et al. Susceptibility of different mouse wild-type strains to develop diet-induced NAFLD/AFLD-associated liver disease. PLoS ONE 11, e0155163 (2016).

    PubMed  PubMed Central  Google Scholar 

  36. Berger, S. B. et al. Cutting edge: RIP1 kinase activity is dispensable for normal development but is a key regulator of inflammation in SHARPIN-deficient mice. J. Immunol. 192, 5476–5480 (2014).

    CAS  PubMed  Google Scholar 

  37. Woo, Y. D. C., Kim, H. Y. & Chung, D. H. Invariant NKT cells recruit CD103+ dendritic cells via XCL1–XCR1 axis, thereby promoting allergic airway resistance. J. Immunol. 198, 1 (2017).

    Google Scholar 

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

    PubMed  Google Scholar 

  39. Akiyama, M. et al. Genome-wide association study identifies 112 new loci for body mass index in the Japanese population. Nat. Genet. 49, 1458–1467 (2017).

    CAS  PubMed  Google Scholar 

  40. Welsh, P. et al. Unraveling the directional link between adiposity and inflammation: a bidirectional Mendelian randomization approach. J. Clin. Endocrinol. Metab. 95, 93–99 (2010).

    CAS  PubMed  Google Scholar 

  41. Vucic, D., Dixit, V. M. & Wertz, I. E. Ubiquitylation in apoptosis: a post-translational modification at the edge of life and death. Nat. Rev. Mol. Cell. Biol. 12, 439–452 (2011).

    CAS  PubMed  Google Scholar 

  42. Chan, F. K., Luz, N. F. & Moriwaki, K. Programmed necrosis in the cross talk of cell death and inflammation. Annu. Rev. Immunol. 33, 79–106 (2015).

    CAS  PubMed  Google Scholar 

  43. Peltzer, N., Darding, M. & Walczak, H. Holding RIPK1 on the ubiquitin leash in TNFR1 signaling. Trends Cell Biol. 26, 445–461 (2016).

    CAS  PubMed  Google Scholar 

  44. Roderick, J. E. et al. Hematopoietic RIPK1 deficiency results in bone marrow failure caused by apoptosis and RIPK3-mediated necroptosis. Proc. Natl Acad. Sci. USA 111, 14436–14441 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Gautheron, J. et al. The necroptosis-inducing kinase RIPK3 dampens adipose tissue inflammation and glucose intolerance. Nat. Commun. 7, 11869 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Dannappel, M. et al. RIPK1 maintains epithelial homeostasis by inhibiting apoptosis and necroptosis. Nature 513, 90–94 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Berzins, S. P., Smyth, M. J. & Baxter, A. G. Presumed guilty: natural killer T cell defects and human disease. Nat. Rev. Immunol. 11, 131–142 (2011).

    CAS  PubMed  Google Scholar 

  48. Lynch, L. Adipose invariant natural killer T cells. Immunology 142, 337–346 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Huh, J. Y. et al. Deletion of CD1d in adipocytes aggravates adipose tissue inflammation and insulin resistance in obesity. Diabetes 66, 835–847 (2017).

    CAS  PubMed  Google Scholar 

  50. Vieth, J. A. et al. TCRα–TCRβ pairing controls recognition of CD1d and directs the development of adipose NKT cells. Nat. Immunol. 18, 36–44 (2017).

    CAS  PubMed  Google Scholar 

  51. Mookerjee-Basu, J. & Kappes, D. J. iNKT cells do a fat lot of good. Nat. Immunol. 18, 10–12 (2016).

    PubMed  Google Scholar 

  52. Lynch, L. et al. Invariant NKT cells and CD1d+ cells amass in human omentum and are depleted in patients with cancer and obesity. Eur. J. Immunol. 39, 1893–1901 (2009).

    CAS  PubMed  Google Scholar 

  53. Engel, I. et al. Innate-like functions of natural killer T cell subsets result from highly divergent gene programs. Nat. Immunol. 17, 728–739 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Plenge, R. M. Priority index for human genetics and drug discovery. Nat. Genet. 51, 1073–1075 (2019).

    CAS  PubMed  Google Scholar 

  55. Fang, H. et al. A genetics-led approach defines the drug target landscape of 30 immune-related traits. Nat. Genet. 51, 1082–1091 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Nelson, M. R. et al. The support of human genetic evidence for approved drug indications. Nat. Genet. 47, 856–860 (2015).

    CAS  PubMed  Google Scholar 

  57. Plenge, R. M., Scolnick, E. M. & Altshuler, D. Validating therapeutic targets through human genetics. Nat. Rev. Drug Discov. 12, 581–594 (2013).

    CAS  PubMed  Google Scholar 

  58. Paik, J. & Duggan, S. Volanesorsen: first global approval. Drugs 79, 1349–1354 (2019).

    CAS  PubMed  Google Scholar 

  59. Witztum, J. L. et al. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N. Engl. J. Med. 381, 531–542 (2019).

    CAS  PubMed  Google Scholar 

  60. Fitzgerald, K. et al. A highly durable RNAi therapeutic inhibitor of PCSK9. N. Engl. J. Med. 376, 41–51 (2017).

    CAS  PubMed  Google Scholar 

  61. Duell, P. B. et al. Long-term mipomersen treatment is associated with a reduction in cardiovascular events in patients with familial hypercholesterolemia. J. Clin. Lipidol. 10, 1011–1021 (2016).

    PubMed  Google Scholar 

  62. Adams, D. et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N. Engl. J. Med. 379, 11–21 (2018).

    CAS  PubMed  Google Scholar 

  63. Stein, C. A. & Castanotto, D. FDA-approved oligonucleotide therapies in 2017. Mol. Ther. 25, 1069–1075 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Levin, A. A. Treating disease at the RNA level with oligonucleotides. N. Engl. J. Med. 380, 57–70 (2019).

    PubMed  Google Scholar 

  65. Nikpay, M. et al. A comprehensive 1000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nat. Genet. 47, 1121–1130 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Ramkhelawon, B. et al. Netrin-1 promotes adipose tissue macrophage retention and insulin resistance in obesity. Nat. Med. 20, 377–384 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Karunakaran, D. et al. Therapeutic inhibition of miR-33 promotes fatty acid oxidation but does not ameliorate metabolic dysfunction in diet-induced obesity. Arterioscler. Thromb. Vasc. Biol. 35, 2536–2543 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2018).

  71. McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: uniform manifold approximation and projection. J. Open Source Softw. 3, 861 (2018).

  72. Frankish, A. et al. GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res. 47, D766–D773 (2019).

    CAS  PubMed  Google Scholar 

  73. Stuart, T. et al. Comprehensive integration of single cell data. Cell 177, 1888–1902.e21 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Carvalho, B. S. & Irizarry, R. A. A framework for oligonucleotide microarray preprocessing. Bioinformatics 26, 2363–2367 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Ritchie, M. E. et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  76. MacDonald, J. W. mogene11sttranscriptcluster.db: Affymetrix mogene11 annotation data (chip mogene11sttranscriptcluster). R package version 8.7.0. (2017).

  77. Riveros-McKay, F. et al. Genetic architecture of human thinness compared to severe obesity. PLoS Genet. 15, e1007603 (2019).

    PubMed  PubMed Central  Google Scholar 

  78. Lloyd-Jones, L. R. et al. The genetic architecture of gene expression in peripheral blood. Am. J. Hum. Genet. 100, 228–237 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank X. Zhao, K. Sheikheleslamy and G. Palidwor for their technical and statistical support. We thank J. Kaprio and A. Rissanen for their contributions and the individuals who participated in the Finnish Twin study. We also thank the University of Ottawa StemCore Facility and the UNGC Sequencing Core at UCLA for performing RNA sequencing. This study was supported with funding from the following organizations: Canadian Institutes of Health Research (K.J.R., R.M., M.E.H., B.C.V. and D.P.C.), NIH (R01 HL119047 to K.J.R.; DK117850 and HL147883 to A.J.L., HL095056, HL28481 and U01 DK105561 to P.P.), Foundation Leduc (A.J.L.), National Psoriasis Foundation (D.K.), Academy of Finland (M.L.; 272376, 266286, 314383 and 315035 to K.H.P.), European Union (M.L.) and Sigrid Juselius Foundation (M.L. and K.H.P.); Finnish Medical Foundation (K.H.P.) and the Finnish Diabetes Research Foundation, Novo Nordisk Foundation, Gyllenberg Foundation, Helsinki University Hospital Research Funds and University of Helsinki (K.H.P.); University of Ottawa Heart Institute Cardiac Endowment Fellowship (A.C.D.); and HHMI Gilliam Fellowship (M.A.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the article.

Author information

Authors and Affiliations

Authors

Contributions

K.J.R. and D.K. conceived the project and wrote the manuscript with input from all authors. A.W.T., S.S. and P. Lau performed and analysed the chromatin immunoprecipitation experiments in human adipose tissue, and A.W.T. performed the promoter luciferase experiments. A.C.D. and D.S. conducted the flow cytometry analysis of human adipose tissue and liver. D.K. and A.R. conducted and analysed the mouse obesity experiments. J.W.K., M.G., M.A.N. and H.W. performed biochemical analyses on mouse tissue. M.E.H. provided assistance with the indirect calorimetry experiments in mice. K.J.R., A.R. and D.P.C. conducted and analysed the single-cell sequencing of human and mouse adipose tissue. M.N. performed the MR and analyses of the OBLE cohort. C.P., M.L. and A.J.L. conducted and analysed data associated with the METSIM cohort and the HMDP. L.B. and B.R. collected and analysed qPCR analyses of human adipose tissue for RIPK1 expression. M.A., K.H.P. and P.P. performed the experiments and analyses of the human adipose single-nuclei data. R.L. provided the ASOs used in the mouse studies. S.B.B., P.J.G. and J.B. developed and provided the Ripk1 mutant mice. B.C.V., P. Liu, A.J.L. and R.M. provided supervision and guidance throughout the study.

Corresponding authors

Correspondence to Denuja Karunakaran or Katey J. Rayner.

Ethics declarations

Competing interests

R.L. is an employee of Ionis Pharmaceuticals, S.B.B. is an employee of Janssen Pharmaceuticals, P.J.G. is an employee of Inzen Therapeutics and J.B. is an employee of GlaxoSmithKline. The other authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Pooja Jha.

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

Extended data

Extended Data Fig. 1 SNPs in the RIPK1 locus associate with obesity in humans.

a, eQTL analysis of SNPs rs2272990 and rs2064310 in the RIPK1 locus and levels of RIPK1 mRNA in adipose tissue from the METSIM cohort. b, Regional association plots of eQTLs for RIPK1 in subcutaneous adipose from GTEX (probe ENSG00000137275.9). SNPs are coloured based on their linkage disequilibrium (r2) with the labeled top eQTL (rs2064310), which has the smallest P value in the region. Linkage disequilibrium and recombination rate calculations are based on the European population from 1000 Genomes reference panel (Phase I; release 3). c, d, Individual SNPs from the GTEx database and their association with RIPK1 expression in visceral (c) and subcutaneous (d) adipose tissue. Number of subjects homozygous or heterozygous for the minor (Ref) or major (Alt) alleles are indicated. p-values as calculated by the GTEx Consortium detailed at: https://www.gtexportal.org. Box plot shows ranked normalized gene expression in median, 1st and 3rd quartiles, 1.5 interquartile range (IQR) of 1st and 3rd quartiles. e, Odds ratio for the index SNP rs6907943 and LD SNPs in the Ottawa OBLE cohort and comparison to rs9939609 and LD SNPs in the FTO locus. fi, Mendelian Randomization analysis of SNPs that are independently (linkage disequilibrium r2<0.2) associated with expression of RIPK1 and significantly (P<0.05) associated with obesity metrics. The beta and standard error of the effect sizes of RIPK1 expression (eQTL, x-axis) and the effect sizes on obesity metrics (y-axis) are plotted. f- UKHLS& GTEX (Adipose tissue) g- UKHLS & METSIM (Adipose tissue) h- UK Biobank & GTEX (Adipose tissue), i- UK Biobank & Llyod-Jones, 2017 (Whole Blood).

Extended Data Fig. 2 Single nuclei sequencing of human adipose tissue and HMDP data from mice.

a, Single-nuclei RNA sequencing on human adipose tissue: UMAP visual representation of 10 cell types derived from single-nucleus RNA-seq of human abdominal subcutaneous adipose tissue. Clusters are identified using the Seurat pipeline and cell types are assigned by biological function of marker genes. Vertical bar plot of average cell type-specific expression of RIPK1 across the 10 cell types identified in human abdominal subcutaneous adipose tissue. Expression was normalized using SCTransform. b, Data from the Hybrid Mouse Diversity Panel (HMDP) showing correlations of Ripk1 mRNA expression and NMR, visceral fat mass at sacrifice, serum insulin and HOMA-IR index in males from approximately 100 inbred strains of mice19. c, Luciferase reporter activity containing the variant sequence plus the human RIPK1 promoter upstream of the firefly luciferase gene (as in Fig. 2b). Data is shown as mean ± SEM of n=4 independent experiments, df. Single cell RNA sequencing of mouse CD45+ immune cells (from GSE128518) after 6, 12 and 18 weeks high fat diet feeding. d, Horizontal bar plot of cell clusters in normal chow (NC) and high fat diet (HFD). e, UMAP plot of Xcl1 cluster (cluster 9, representing NK/NKT cells) and (f) the corresponding expression of Nfil3 and Ripk1 in cluster 9.

Extended Data Fig. 3 Analysis of flow cytometry and single cell RNA sequencing of human adipose.

a, Mean fluorescence intensity (MFI) of RIPK1 and E4BP4 in iNKT cells from human omental adipose tissue (left) and liver (right) from obese patients (n= 12 patients total). iNKT were identified as CD19-CD3+αGalCer-loaded CD1d-tetramer+ population and based on negative control (isotype IgG) staining. Outliers were removed according to ROUT method with a specified FDR of 1% (adipose, two E4BP4 samples; liver one E4BP4 and one RIPK1 sample). Data is mean ± SD, ***p≤0.001 by paired two-tailed Student’s t-test. b, UMAP embeddings of scRNA-seq data from human omentum samples. The colour maps represent the log2-based gene expression values of various T/NK cell markers (CD3D, XCL1, NKG7, CD8A) and myeloid markers (LYZ, CD14). The bottom panels show RIPK1 and NFIL3 expression across these populations. Data deposited in the GEO database (accession number GSE151889).

Extended Data Fig. 4 Analysis of single cell RNA sequencing of human adipose and liver.

Single-cell RNA sequencing comparison of human adipose tissue (from Supplementary Fig. 3) and human liver (from GSE115469), showing expression of major cluster IDs XCL1 (NK/NKT), CD3D (T-cells), CD14 (monocytes) and genes of interest IL10, RIPK1 and NFIL3.

Extended Data Fig. 5 Metabolic analyses of RIPK1 ASO and cont ASO treated mice.

a, C57BL6 mice were treated with 50mg/kg control ASO, RIPK1 ASO-A or ASO-B over 20 weeks while fed a normal chow diet, and body weight was measured weekly. Data depicts mean ± SEM of n=3 mice per group. Two-way ANOVA using Holm-Sidak’s multiple comparison test. *p<0.05 control ASO vs. RIPK1 ASO-A and ASO-B. b, Western blot of RIPK1 (top) and GAPDH (bottom) from adipose tissue of ASO treated mice after 5 weeks of treatment. Quantification of RIPK1 band density normalized to GAPDH band density was performed using Image J. Data representative of mean ± SD of n=7-8 mice/group, p=0.09 RIPK1 ASO-A vs cont ASO, p=0.02 RIPK1 ASO-B vs. cont ASO by two-tailed Student’s t-test. One outlier was removed according to ROUT method with a specified FDR of 1%. cf, Mice were caged in metabolic cages with indirect calorimetric and activity monitoring. VO2 (c) and respiratory exchange rations (RER) (d) and beam breaks were recorded as a measure of physical activity (e). Total food consumption (f) was measured over a 48h period. Data representative of mean ± SEM of n=8 mice/group, *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001, one-way ANOVA using Tukey’s multiple comparison test. g, Plasma levels of alanine transaminase (ALT) and aspartate transaminase (AST) were determined after 20 weeks of ASO treatment. Data representative of mean ± SEM of n=8 mice/group analyzed by one-way ANOVA using Tukey’s multiple comparison test. *p<0.05 vs. control ASO and RIPK1 ASO-B.

Extended Data Fig. 6 Analysis of diet-induced obesity in RIPK1 kinase dead mice.

WT or RIP1K45A mice (male, n=8/group) were fed a high fat diet for 20 weeks. a, Body weight. b, Epididymal adipose tissue, liver and spleen weights at termination of study. c, d, Fasting glucose tolerance test (GTT, c) or insulin tolerance test (ITT, d) and area under the curve. Data representative of mean ± SEM and analyzed by Two-way ANOVA (body weight, GTT and ITT) or two-tailed Student’s t-test (tissues weights).

Extended Data Fig. 7 Serum cytokine analysis of cont ASO and RIPK1 ASO treated mice.

Serum cytokines from mice treated with cont ASO or RIPK1 ASO-B for 24 weeks. Cytokine analysis was done using the Bioplex Pro Cytokine 23-plex assay. No significant differences between cytokines IL-1α, TNFα, IL-4 or IL-10 were observed. Data representative of mean ± SEM of n=8 mice/group and was analysed by one-way ANOVA using Dunnett’s multiple comparison test.

Extended Data Fig. 8 Analysis of single cell RNA sequencing of mouse adipose tissue.

a, Clustered UMAP embedding of scRNA-seq data from mouse adipose tissue. The colours and labels represent each of the 13 distinct clusters. b, The same embeddings as in (a), but separately coloring the cells derived from either Control ASO or RIPK1 ASO-B samples. c, A graph showing the percentage of cells from each sample that correspond to each cell type. d, UMAP embedding of scRNA-seq data from T cell populations of mouse adipose tissue. T cells were defined as CD3+ clusters. Plots show the log-transformed gene expression of various markers defining NKT (Xcl1), CD4+ T (Cd4), CD8+ T (Cd8a), Memory CD4+ T (Ccr7), Naive CD4+ T (S100a4), Regulatory T (Foxp3), and Gamma Delta T (Cd163l1). Data deposited in the GEO database (accession number GSE151889).

Extended Data Fig. 9 Flow cytometry of adipose and circulating immune cells from cont ASO and RIPK1 ASO treated mice.

a, Representative flow cytometry plots and NKT % (expressed as % of F4/80- cells that are αGalCer-loaded CD1d-tetramer+CD3e+) in spleen from mice treated for 5 weeks with cont ASO or RIPK1 ASO (n=8/group). Data representative of mean ± SEM of n=8 mice/group, analyzed by two-tailed Student’s t-test. b, c, Flow cytometry analysis of circulating leukocytes from mice treated with control ASO or RIP1 ASO-B at 5 weeks: Ly6Clow monocytes *p<0.05 (p=0.0452) Ly6Chigh monocytes *p<0.05 (p=0.0108), and CD3e+ T cells. (c) Flow cytometry analysis of circulating leukocytes from mice treated with control ASO or RIP1 ASO-B at 22 weeks: Ly6Clow monocytes, Ly6Chigh monocytes and CD3e+ T cells (p=0.0132). Data in b, c representative of mean ± SEM of n=8 mice/group, analyzed by two-tailed Student’s t-test.

Extended Data Fig. 10 Flow cytometry of adipose immune cells from cont ASO and RIPK1 ASO treated mice.

a, Regulatory T-cell (CD25+Foxp3+) and NK (NK1.1+) cells analysis by flow cytometry of isolated SVF of adipose tissue from mice treated with control ASO or RIP1 ASO-B for 5 weeks while fed a high fat diet (n=7-8/group). b, Gating strategy for selecting CD45+, CD3e+, CD4+ cells then those that were positive for CD25 and Foxp3 (to represent T regulatory cells) or NK1.1 (to represent NK cells). Data depicts mean ± SEM of n=8 mice/group.

Supplementary information

Supplementary Information

Supplementary Figs. 1–5 and Table 1

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed western blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Karunakaran, D., Turner, A.W., Duchez, AC. et al. RIPK1 gene variants associate with obesity in humans and can be therapeutically silenced to reduce obesity in mice. Nat Metab 2, 1113–1125 (2020). https://doi.org/10.1038/s42255-020-00279-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00279-2

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