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.

  • Review Article
  • Published:

The aetiology and molecular landscape of insulin resistance

Abstract

Insulin resistance, defined as a defect in insulin-mediated control of glucose metabolism in tissues — prominently in muscle, fat and liver — is one of the earliest manifestations of a constellation of human diseases that includes type 2 diabetes and cardiovascular disease. These diseases are typically associated with intertwined metabolic abnormalities, including obesity, hyperinsulinaemia, hyperglycaemia and hyperlipidaemia. Insulin resistance is caused by a combination of genetic and environmental factors. Recent genetic and biochemical studies suggest a key role for adipose tissue in the development of insulin resistance, potentially by releasing lipids and other circulating factors that promote insulin resistance in other organs. These extracellular factors perturb the intracellular concentration of a range of intermediates, including ceramide and other lipids, leading to defects in responsiveness of cells to insulin. Such intermediates may cause insulin resistance by inhibiting one or more of the proximal components in the signalling cascade downstream of insulin (insulin receptor, insulin receptor substrate (IRS) proteins or AKT). However, there is now evidence to support the view that insulin resistance is a heterogeneous disorder that may variably arise in a range of metabolic tissues and that the mechanism for this effect likely involves a unified insulin resistance pathway that affects a distal step in the insulin action pathway that is more closely linked to the terminal biological response. Identifying these targets is of major importance, as it will reveal potential new targets for treatments of diseases associated with insulin resistance.

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: Tissue-specific regulation of metabolism during fasting, feeding and insulin resistance.
Fig. 2: Progression from insulin resistance to type 2 diabetes.
Fig. 3: Dose–response characteristics of insulin action.
Fig. 4: Putative factors that contribute to insulin resistance.
Fig. 5: Putative intracellular drivers of insulin resistance.

Similar content being viewed by others

References

  1. Root, H. F. Insulin resistance and bronze diabetes. N. Engl. J. Med. 201, 201–206 (1929).

    Article  Google Scholar 

  2. Himsworth, H. P. Diabetes mellitus: its differentiation into insulin-sensitive and insulin-insensitive types. 1936. Int. J. Epidemiol. 42, 1594–1598 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Petersen, M. C. & Shulman, G. I. Mechanisms of insulin action and insulin resistance. Physiol. Rev. 98, 2133–2223 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Barber, T. M., Kyrou, I., Randeva, H. S. & Weickert, M. O. Mechanisms of insulin resistance at the crossroad of obesity with associated metabolic abnormalities and cognitive dysfunction. Int. J. Mol. Sci. 22, 546 (2021).

    Article  CAS  PubMed Central  Google Scholar 

  5. Philipson, L. H. Harnessing heterogeneity in type 2 diabetes mellitus. Nat. Rev. Endocrinol. 16, 79–80 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ahlqvist, E. et al. Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 6, 361–369 (2018).

    Article  PubMed  Google Scholar 

  7. Erion, K. A. & Corkey, B. E. Hyperinsulinemia: a cause of obesity? Curr. Obes. Rep. 6, 178–186 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Clemmensen, C. et al. Gut-brain cross-talk in metabolic control. Cell 168, 758–774 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rizza, R. A., Mandarino, L. J., Genest, J., Baker, B. A. & Gerich, J. E. Production of insulin resistance by hyperinsulinaemia in man. Diabetologia 28, 70–75 (1985).

    Article  CAS  PubMed  Google Scholar 

  10. Ferrannini, E. et al. Influence of ethnicity and familial diabetes on glucose tolerance and insulin action: a physiological analysis. J. Clin. Endocrinol. Metab. 88, 3251–3257 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Kashyap, S. R. et al. Discordant effects of a chronic physiological increase in plasma FFA on insulin signaling in healthy subjects with or without a family history of type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 287, E537–E546 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Lillioja, S. et al. Insulin resistance and insulin secretory dysfunction as precursors of non-insulin-dependent diabetes mellitus. Prospective studies of Pima Indians. N. Engl. J. Med. 329, 1988–1992 (1993).

    Article  CAS  PubMed  Google Scholar 

  13. Vaag, A., Henriksen, J. E. & Beck-Nielsen, H. Decreased insulin activation of glycogen synthase in skeletal muscles in young nonobese Caucasian first-degree relatives of patients with non-insulin-dependent diabetes mellitus. J. Clin. Invest. 89, 782–788 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hollenbeck, C. & Reaven, G. M. Variations in insulin-stimulated glucose uptake in healthy individuals with normal glucose tolerance. J. Clin. Endocrinol. Metab. 64, 1169–1173 (1987).

    Article  CAS  PubMed  Google Scholar 

  15. Ghaben, A. L. & Scherer, P. E. Adipogenesis and metabolic health. Nat. Rev. Mol. Cell Biol. 20, 242–258 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Jacob, S. et al. Association of increased intramyocellular lipid content with insulin resistance in lean nondiabetic offspring of type 2 diabetic subjects. Diabetes 48, 1113–1119 (1999).

    Article  CAS  PubMed  Google Scholar 

  17. Lim, K., Haider, A., Adams, C., Sleigh, A. & Savage, D. Lipodystrophy: a paradigm for understanding the consequences of ‘overloading’ adipose tissue. Physiol. Rev. https://doi.org/10.1152/physrev.00032.2020 (2020).

    Article  PubMed  Google Scholar 

  18. Blüher, M. Metabolically healthy obesity. Endocr. Rev. 41, 405–420 (2020).

    Article  Google Scholar 

  19. Pontiroli, A. E., Alberetto, M., Capra, F. & Pozza, G. The glucose clamp technique for the study of patients with hypoglycemia: insulin resistance as a feature of insulinoma. J. Endocrinol. Invest. 13, 241–245 (1990).

    Article  CAS  PubMed  Google Scholar 

  20. Marbán, S. L. & Roth, J. Transgenic hyperinsulinemia: a mouse model of insulin resistance and glucose intolerance without obesity. in Lessons from Animal Diabetes VI: 75th Anniversary of the Insulin Discovery (ed. Shafrir, E.) 201–224 (Birkhäuser, 1996).

  21. Gray, S. L., Donald, C., Jetha, A., Covey, S. D. & Kieffer, T. J. Hyperinsulinemia precedes insulin resistance in mice lacking pancreatic beta-cell leptin signaling. Endocrinology 151, 4178–4186 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Alemzadeh, R., Slonim, A. E., Zdanowicz, M. M. & Maturo, J. Modification of insulin resistance by diazoxide in obese Zucker rats. Endocrinology 133, 705–712 (1993).

    Article  CAS  PubMed  Google Scholar 

  23. Pedersen, D. J. et al. A major role of insulin in promoting obesity-associated adipose tissue inflammation. Mol. Metab. 4, 507–518 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Templeman, N. M. et al. Reduced circulating insulin enhances insulin sensitivity in old mice and extends lifespan. Cell Rep. 20, 451–463 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Czech, M. P. Insulin action and resistance in obesity and type 2 diabetes. Nat. Med. 23, 804–814 (2017). Recent state-of-the-art review of IR, obesity and T2D.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tan, S.-X. et al. Selective insulin resistance in adipocytes. J. Biol. Chem. 290, 11337–11348 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tonks, K. T. et al. Impaired Akt phosphorylation in insulin-resistant human muscle is accompanied by selective and heterogeneous downstream defects. Diabetologia 56, 875–885 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Brown, M. S. & Goldstein, J. L. Selective versus total insulin resistance: a pathogenic paradox. Cell Metab. 7, 95–96 (2008). Describes the initial formulation of the concept of selective IR, which has become increasingly accepted as critical to understanding T2D.

    Article  CAS  PubMed  Google Scholar 

  29. Hillgartner, F. B., Salati, L. M. & Goodridge, A. G. Physiological and molecular mechanisms involved in nutritional regulation of fatty acid synthesis. Physiol. Rev. 75, 47–76 (1995).

    Article  CAS  PubMed  Google Scholar 

  30. Hellerstein, M. K., Schwarz, J. M. & Neese, R. A. Regulation of hepatic de novo lipogenesis in humans. Annu. Rev. Nutr. 16, 523–557 (1996).

    Article  CAS  PubMed  Google Scholar 

  31. Titchenell, P. M. et al. Direct hepatocyte insulin signaling is required for lipogenesis but is dispensable for the suppression of glucose production. Cell Metab. 23, 1154–1166 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hammarstedt, A., Gogg, S., Hedjazifar, S., Nerstedt, A. & Smith, U. Impaired adipogenesis and dysfunctional adipose tissue in human hypertrophic obesity. Physiol. Rev. 98, 1911–1941 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Kolterman, O. G., Insel, J., Saekow, M. & Olefsky, J. M. Mechanisms of insulin resistance in human obesity: evidence for receptor and postreceptor defects. J. Clin. Invest. 65, 1272–1284 (1980). One of the original articles resolving defects in insulin action into two separate components.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Jia, Q., Morgan-Bathke, M. E. & Jensen, M. D. Adipose tissue macrophage burden, systemic inflammation, and insulin resistance. Am. J. Physiol. Endocrinol. Metab. 319, E254–E264 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Turner, N. et al. Distinct patterns of tissue-specific lipid accumulation during the induction of insulin resistance in mice by high-fat feeding. Diabetologia 56, 1638–1648 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Fazakerley, D. J. et al. Mitochondrial CoQ deficiency is a common driver of mitochondrial oxidants and insulin resistance. eLife 7, e32111 (2018). Comprehensive evidence to show that the levels of CoQ in mitochondria are lower in IR and that supplementation with CoQ reverses IR.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Perry, R. J. et al. Hepatic acetyl CoA links adipose tissue inflammation to hepatic insulin resistance and type 2 diabetes. Cell 160, 745–758 (2015). A series of experiments supporting the idea of non-cell-autonomous regulation of hepatic glucose output through insulin’s regulation of lipolysis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rebrin, K., Steil, G. M., Mittelman, S. D. & Bergman, R. N. Causal linkage between insulin suppression of lipolysis and suppression of liver glucose output in dogs. J. Clin. Invest. 98, 741–749 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Minokoshi, Y., Kahn, C. R. & Kahn, B. B. Tissue-specific ablation of the GLUT4 glucose transporter or the insulin receptor challenges assumptions about insulin action and glucose homeostasis. J. Biol. Chem. 278, 33609–33612 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Kim, J. K. et al. Glucose toxicity and the development of diabetes in mice with muscle-specific inactivation of GLUT4. J. Clin. Invest. 108, 153–160 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gancheva, S., Jelenik, T., Álvarez-Hernández, E. & Roden, M. Interorgan metabolic crosstalk in human insulin resistance. Physiol. Rev. 98, 1371–1415 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Severinsen, M. C. K. & Pedersen, B. K. Muscle-organ crosstalk: the emerging roles of myokines. Endocr. Rev. 41, 594–609 (2020).

    Article  PubMed Central  Google Scholar 

  43. Burchfield, J. G. et al. High dietary fat and sucrose results in an extensive and time-dependent deterioration in health of multiple physiological systems in mice. J. Biol. Chem. 293, 5731–5745 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hoehn, K. L. et al. IRS1-independent defects define major nodes of insulin resistance. Cell Metab. 7, 421–433 (2008). An important study challenging the view that IRS1 is the critical biochemical site for IR.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Copps, K. D. & White, M. F. Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2. Diabetologia 55, 2565–2582 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kahn, C. R. et al. The syndromes of insulin resistance and acanthosis nigricans. Insulin-receptor disorders in man. N. Engl. J. Med. 294, 739–745 (1976).

    Article  CAS  PubMed  Google Scholar 

  47. Kono, T. & Barham, F. W. The relationship between the insulin-binding capacity of fat cells and the cellular response to insulin. Studies with intact and trypsin-treated fat cells. J. Biol. Chem. 246, 6210–6216 (1971). One of the first studies to highlight the spareness in insulin receptors in adipocytes giving rise to the notion that defects in insulin receptors are unlikely to cause IR in humans.

    Article  CAS  PubMed  Google Scholar 

  48. Kahn, C. R. Insulin resistance, insulin insensitivity, and insulin unresponsiveness: a necessary distinction. Metabolism 27, 1893–1902 (1978).

    Article  CAS  PubMed  Google Scholar 

  49. Olefsky, J. M., Kolterman, O. G. & Scarlett, J. A. Insulin action and resistance in obesity and noninsulin-dependent type II diabetes mellitus. Am. J. Physiol. 243, E15–E30 (1982).

    CAS  PubMed  Google Scholar 

  50. Camps, M. et al. Evidence for the lack of spare high-affinity insulin receptors in skeletal muscle. Biochem. J. 285, 993–999 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gumà, A. et al. Effect of benzyl succinate on insulin receptor function and insulin action in skeletal muscle: further evidence for a lack of spare high-affinity insulin receptors. Mol. Cell. Endocrinol. 91, 29–33 (1993).

    Article  PubMed  Google Scholar 

  52. Fehlmann, M., Morin, O., Kitabgi, P. & Freychet, P. Insulin and glucagon receptors of isolated rat hepatocytes: comparison between hormone binding and amino acid transport stimulation. Endocrinology 109, 253–261 (1981).

    Article  CAS  PubMed  Google Scholar 

  53. Merry, T. L. et al. Impairment of insulin signalling in peripheral tissue fails to extend murine lifespan. Aging Cell 16, 761–772 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Czech, M. P. Cellular basis of insulin insensitivity in large rat adipocytes. J. Clin. Invest. 57, 1523–1532 (1976).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Melvin, A., O’Rahilly, S. & Savage, D. B. Genetic syndromes of severe insulin resistance. Curr. Opin. Genet. Dev. 50, 60–67 (2018).

    Article  CAS  PubMed  Google Scholar 

  56. Crouthamel, M.-C. et al. Mechanism and management of AKT inhibitor-induced hyperglycemia. Clin. Cancer Res. 15, 217–225 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Nandi, A., Kitamura, Y., Kahn, C. R. & Accili, D. Mouse models of insulin resistance. Physiol. Rev. 84, 623–647 (2004).

    Article  CAS  PubMed  Google Scholar 

  58. Jaiswal, N. et al. The role of skeletal muscle Akt in the regulation of muscle mass and glucose homeostasis. Mol. Metab. 28, 1–13 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lu, M. et al. Insulin regulates liver metabolism in vivo in the absence of hepatic Akt and Foxo1. Nat. Med. 18, 388–395 (2012). Mouse genetic experiments demonstrating non-cell-autonomous regulation of hepatic metabolism.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Tan, S.-X. et al. Amplification and demultiplexing in insulin-regulated Akt protein kinase pathway in adipocytes. J. Biol. Chem. 287, 6128–6138 (2012).

    Article  CAS  PubMed  Google Scholar 

  61. Ng, Y. et al. Cluster analysis of insulin action in adipocytes reveals a key role for Akt at the plasma membrane. J. Biol. Chem. 285, 2245–2257 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Larance, M. et al. Global phosphoproteomics identifies a major role for AKT and 14-3-3 in regulating EDC3. Mol. Cell. Proteomics 9, 682–694 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Trefely, S. et al. Kinome screen identifies PFKFB3 and glucose metabolism as important regulators of the insulin/insulin-like growth factor (IGF)-1 signaling pathway. J. Biol. Chem. 290, 25834–25846 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ricort, J. M., Tanti, J. F., Van Obberghen, E. & Le Marchand-Brustel, Y. Alterations in insulin signalling pathway induced by prolonged insulin treatment of 3T3-L1 adipocytes. Diabetologia 38, 1148–1156 (1995).

    Article  CAS  PubMed  Google Scholar 

  65. Kurowski, T. G. et al. Hyperglycemia inhibits insulin activation of Akt/protein kinase B but not phosphatidylinositol 3-kinase in rat skeletal muscle. Diabetes 48, 658–663 (1999).

    Article  CAS  PubMed  Google Scholar 

  66. Oku, A. et al. Inhibitory effect of hyperglycemia on insulin-induced Akt/protein kinase B activation in skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 280, E816–E824 (2001).

    Article  CAS  PubMed  Google Scholar 

  67. Ronnett, G. V., Knutson, V. P. & Lane, M. D. Insulin-induced down-regulation of insulin receptors in 3T3-L1 adipocytes. Altered rate of receptor inactivation. J. Biol. Chem. 257, 4285–4291 (1982).

    Article  CAS  PubMed  Google Scholar 

  68. Draznin, B. Molecular mechanisms of insulin resistance: serine phosphorylation of insulin receptor substrate-1 and increased expression of p85alpha: the two sides of a coin. Diabetes 55, 2392–2397 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Copps, K. D. et al. Irs1 serine 307 promotes insulin sensitivity in mice. Cell Metab. 11, 84–92 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Brandon, A. E. et al. Protein kinase C epsilon deletion in adipose tissue, but not in liver, improves glucose tolerance. Cell Metab. 29, 183–191.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  71. Deblon, N. et al. Chronic mTOR inhibition by rapamycin induces muscle insulin resistance despite weight loss in rats. Br. J. Pharmacol. 165, 2325–2340 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lamming, D. W. et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science 335, 1638–1643 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Friedman, J. E., Caro, J. F., Pories, W. J., Azevedo, J. L. Jr & Dohm, G. L. Glucose metabolism in incubated human muscle: effect of obesity and non-insulin-dependent diabetes mellitus. Metabolism 43, 1047–1054 (1994).

    Article  CAS  PubMed  Google Scholar 

  74. Shulman, G. I. et al. Quantitation of muscle glycogen synthesis in normal subjects and subjects with non-insulin-dependent diabetes by 13C nuclear magnetic resonance spectroscopy. N. Engl. J. Med. 322, 223–228 (1990). Demonstration of muscle IR in humans with T2D.

    Article  CAS  PubMed  Google Scholar 

  75. Baron, A. D., Laakso, M., Brechtel, G. & Edelman, S. V. Reduced capacity and affinity of skeletal muscle for insulin-mediated glucose uptake in noninsulin-dependent diabetic subjects. Effects of insulin therapy. J. Clin. Invest. 87, 1186–1194 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Friedman, J. E. et al. Restoration of insulin responsiveness in skeletal muscle of morbidly obese patients after weight loss. Effect on muscle glucose transport and glucose transporter GLUT4. J. Clin. Invest. 89, 701–705 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Meyer, M. M., Levin, K., Grimmsmann, T., Beck-Nielsen, H. & Klein, H. H. Insulin signalling in skeletal muscle of subjects with or without type II-diabetes and first degree relatives of patients with the disease. Diabetologia 45, 813–822 (2002).

    Article  CAS  PubMed  Google Scholar 

  78. Kim, Y. B., Nikoulina, S. E., Ciaraldi, T. P., Henry, R. R. & Kahn, B. B. Normal insulin-dependent activation of Akt/protein kinase B, with diminished activation of phosphoinositide 3-kinase, in muscle in type 2 diabetes. J. Clin. Invest. 104, 733–741 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Ramos, P. A. et al. Insulin-stimulated muscle glucose uptake and insulin signaling in lean and obese humans. J. Clin. Endocrinol. Metab. 106, e1631–e1646 (2020).

    Article  Google Scholar 

  80. Vind, B. F. et al. Impaired insulin-induced site-specific phosphorylation of TBC1 domain family, member 4 (TBC1D4) in skeletal muscle of type 2 diabetes patients is restored by endurance exercise-training. Diabetologia 54, 157–167 (2011).

    Article  CAS  PubMed  Google Scholar 

  81. Caro, J. F. et al. Insulin receptor kinase in human skeletal muscle from obese subjects with and without noninsulin dependent diabetes. J. Clin. Invest. 79, 1330–1337 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Garvey, W. T., Maianu, L., Zhu, J. H., Hancock, J. A. & Golichowski, A. M. Multiple defects in the adipocyte glucose transport system cause cellular insulin resistance in gestational diabetes. Heterogeneity in the number and a novel abnormality in subcellular localization of GLUT4 glucose transporters. Diabetes 42, 1773–1785 (1993).

    Article  CAS  PubMed  Google Scholar 

  83. Ryder, J. W. et al. Use of a novel impermeable biotinylated photolabeling reagent to assess insulin- and hypoxia-stimulated cell surface GLUT4 content in skeletal muscle from type 2 diabetic patients. Diabetes 49, 647–654 (2000).

    Article  CAS  PubMed  Google Scholar 

  84. Garvey, W. T. et al. Evidence for defects in the trafficking and translocation of GLUT4 glucose transporters in skeletal muscle as a cause of human insulin resistance. J. Clin. Invest. 101, 2377–2386 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Gumà, A., Zierath, J. R., Wallberg-Henriksson, H. & Klip, A. Insulin induces translocation of GLUT-4 glucose transporters in human skeletal muscle. Am. J. Physiol. 268, E613–E622 (1995).

    PubMed  Google Scholar 

  86. Dills, W. L. Jr, McDonough, G. M. & Kingsley, P. B. Glucose-stimulated protein synthesis in rat testis slices: substrate specificity and effects of insulin and substrate analogs. Biol. Reprod. 25, 466–474 (1981).

    Article  CAS  PubMed  Google Scholar 

  87. Chlouverakis, C. The action of glucose on lipolysis. Metabolism 16, 469–472 (1967).

    Article  CAS  PubMed  Google Scholar 

  88. Li, J., Houseknecht, K. L., Stenbit, A. E., Katz, E. B. & Charron, M. J. Reduced glucose uptake precedes insulin signaling defects in adipocytes from heterozygous GLUT4 knockout mice. FASEB J. 14, 1117–1125 (2000).

    Article  CAS  PubMed  Google Scholar 

  89. Shepherd, P. R. & Kahn, B. B. Glucose transporters and insulin action — implications for insulin resistance and diabetes mellitus. N. Engl. J. Med. 341, 248–257 (1999).

    Article  CAS  PubMed  Google Scholar 

  90. Etgen, G. J. Jr et al. Exercise training reverses insulin resistance in muscle by enhanced recruitment of GLUT-4 to the cell surface. Am. J. Physiol. 272, E864–E869 (1997). Study presenting evidence that the defect in glucose transport in IR is specific to insulin action and does not apply to exercise or contraction.

    CAS  PubMed  Google Scholar 

  91. Klip, A. et al. Recruitment of GLUT-4 glucose transporters by insulin in diabetic rat skeletal muscle. Biochem. Biophys. Res. Commun. 172, 728–736 (1990).

    Article  CAS  PubMed  Google Scholar 

  92. Kennedy, J. W. et al. Acute exercise induces GLUT4 translocation in skeletal muscle of normal human subjects and subjects with type 2 diabetes. Diabetes 48, 1192–1197 (1999).

    Article  CAS  PubMed  Google Scholar 

  93. Ploug, T., van Deurs, B., Ai, H., Cushman, S. W. & Ralston, E. Analysis of GLUT4 distribution in whole skeletal muscle fibers: identification of distinct storage compartments that are recruited by insulin and muscle contractions. J. Cell Biol. 142, 1429–1446 (1998). Study demonstrating that insulin and contraction stimulate recruitment of GLUT4 from distinct intracellular pools.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Stöckli, J. et al. The RabGAP TBC1D1 plays a central role in exercise-regulated glucose metabolism in skeletal muscle. Diabetes 64, 1914–1922 (2015).

    Article  PubMed  CAS  Google Scholar 

  95. Humphrey, S. J. et al. Dynamic adipocyte phosphoproteome reveals that Akt directly regulates mTORC2. Cell Metab. 17, 1009–1020 (2013). Important study demonstrating the complexity of protein phosphorylation changes in response to insulin.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Kjøbsted, R. et al. Intact regulation of the AMPK signaling network in response to exercise and insulin in skeletal muscle of male patients with type 2 diabetes: illumination of AMPK activation in recovery from exercise. Diabetes 65, 1219–1230 (2016).

    Article  PubMed  CAS  Google Scholar 

  97. Eguez, L. et al. Full intracellular retention of GLUT4 requires AS160 Rab GTPase activating protein. Cell Metab. 2, 263–272 (2005).

    Article  CAS  PubMed  Google Scholar 

  98. Batista, T. M. et al. A cell-autonomous signature of dysregulated protein phosphorylation underlies muscle insulin resistance in type 2 diabetes. Cell Metab. 32, 844–859.e5 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Boden, G. et al. Excessive caloric intake acutely causes oxidative stress, GLUT4 carbonylation, and insulin resistance in healthy men. Sci. Transl. Med. 7, 304re7 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Hauck, A. K., Huang, Y., Hertzel, A. V. & Bernlohr, D. A. Adipose oxidative stress and protein carbonylation. J. Biol. Chem. 294, 1083–1088 (2019).

    Article  CAS  PubMed  Google Scholar 

  101. Hammarstedt, A., Graham, T. E. & Kahn, B. B. Adipose tissue dysregulation and reduced insulin sensitivity in non-obese individuals with enlarged abdominal adipose cells. Diabetol. Metab. Syndr. 4, 42 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Weyer, C., Foley, J. E., Bogardus, C., Tataranni, P. A. & Pratley, R. E. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance. Diabetologia 43, 1498–1506 (2000).

    Article  CAS  PubMed  Google Scholar 

  103. Abel, E. D. et al. Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409, 729–733 (2001). One of the first studies demonstrating that IR in adipocytes can propriate to other peripheral organs.

    Article  CAS  PubMed  Google Scholar 

  104. Vazirani, R. P. et al. Disruption of adipose Rab10-dependent insulin signaling causes hepatic insulin resistance. Diabetes 65, 1577–1589 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Poulsen, P. et al. Heritability of insulin secretion, peripheral and hepatic insulin action, and intracellular glucose partitioning in young and old Danish twins. Diabetes 54, 275–283 (2005).

    Article  CAS  PubMed  Google Scholar 

  106. Flannick, J. et al. Exome sequencing of 20,791 cases of type 2 diabetes and 24,440 controls. Nature 570, 71–76 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Dimas, A. S. et al. Impact of type 2 diabetes susceptibility variants on quantitative glycemic traits reveals mechanistic heterogeneity. Diabetes 63, 2158–2171 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Lotta, L. A. et al. Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance. Nat. Genet. 49, 17–26 (2017). An important study identifying 53 genetic loci associated with measures of IR in humans, highlighting a major role for adipogenesis.

    Article  CAS  PubMed  Google Scholar 

  109. Levin, K., Hother-Nielsen, O., Henriksen, J. E. & Beck-Nielsen, H. Effects of troglitazone in young first-degree relatives of patients with type 2 diabetes. Diabetes Care 27, 148–154 (2004).

    Article  CAS  PubMed  Google Scholar 

  110. Chaurasia, B. & Summers, S. A. Ceramides in metabolism: key lipotoxic players. Annu. Rev. Physiol. 83, 303–330 (2021).

    Article  CAS  PubMed  Google Scholar 

  111. Boden, G. Free fatty acids (FFA), a link between obesity and insulin resistance. Front. Biosci. 3, d169–d175 (1998).

    Article  CAS  PubMed  Google Scholar 

  112. Pereira, S. et al. Resveratrol prevents insulin resistance caused by short-term elevation of free fatty acids in vivo. Appl. Physiol. Nutr. Metab. 40, 1129–1136 (2015).

    Article  CAS  PubMed  Google Scholar 

  113. Felber, J. P. & Vannotti, A. Effects of fat infusion on glucose tolerance and insulin plasma levels. Med. Exp. Int. J. Exp. Med. 10, 153–156 (1964).

    CAS  PubMed  Google Scholar 

  114. Wang, L. et al. Adipocyte Gi signaling is essential for maintaining whole-body glucose homeostasis and insulin sensitivity. Nat. Commun. 11, 2995 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Karpe, F., Dickmann, J. R. & Frayn, K. N. Fatty acids, obesity, and insulin resistance: time for a reevaluation. Diabetes 60, 2441–2449 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Perseghin, G., Ghosh, S., Gerow, K. & Shulman, G. I. Metabolic defects in lean nondiabetic offspring of NIDDM parents: a cross-sectional study. Diabetes 46, 1001–1009 (1997).

    Article  CAS  PubMed  Google Scholar 

  117. McQuaid, S. E. et al. Downregulation of adipose tissue fatty acid trafficking in obesity: a driver for ectopic fat deposition? Diabetes 60, 47–55 (2011).

    Article  CAS  PubMed  Google Scholar 

  118. Jeffery, E., Church, C. D., Holtrup, B., Colman, L. & Rodeheffer, M. S. Rapid depot-specific activation of adipocyte precursor cells at the onset of obesity. Nat. Cell Biol. 17, 376–385 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Jeffery, E. et al. The adipose tissue microenvironment regulates depot-specific adipogenesis in obesity. Cell Metab. 24, 142–150 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Arner, P., Arner, E., Hammarstedt, A. & Smith, U. Genetic predisposition for type 2 diabetes, but not for overweight/obesity, is associated with a restricted adipogenesis. PLoS ONE 6, e18284 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Ye, J. Regulation of PPARgamma function by TNF-alpha. Biochem. Biophys. Res. Commun. 374, 405–408 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Shao, M. et al. De novo adipocyte differentiation from Pdgfrβ preadipocytes protects against pathologic visceral adipose expansion in obesity. Nat. Commun. 9, 890 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Kim, J.-Y. et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J. Clin. Invest. 117, 2621–2637 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Shepherd, P. R. et al. Adipose cell hyperplasia and enhanced glucose disposal in transgenic mice overexpressing GLUT4 selectively in adipose tissue. J. Biol. Chem. 268, 22243–22246 (1993).

    Article  CAS  PubMed  Google Scholar 

  125. Carobbio, S., Pellegrinelli, V. & Vidal-Puig, A. Adipose tissue function and expandability as determinants of lipotoxicity and the metabolic syndrome. Adv. Exp. Med. Biol. 960, 161–196 (2017).

    Article  CAS  PubMed  Google Scholar 

  126. Czech, M. P. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol. Metab. 34, 27–42 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. 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 

  128. Kahn, C. R., Wang, G. & Lee, K. Y. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J. Clin. Invest. 129, 3990–4000 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Friedman, J. M. Leptin and the endocrine control of energy balance. Nat. Metab. 1, 754–764 (2019).

    Article  CAS  PubMed  Google Scholar 

  130. Blüher, M. et al. Circulating adiponectin and expression of adiponectin receptors in human skeletal muscle: associations with metabolic parameters and insulin resistance and regulation by physical training. J. Clin. Endocrinol. Metab. 91, 2310–2316 (2006).

    Article  PubMed  CAS  Google Scholar 

  131. Meyer, L. K., Ciaraldi, T. P., Henry, R. R., Wittgrove, A. C. & Phillips, S. A. Adipose tissue depot and cell size dependency of adiponectin synthesis and secretion in human obesity. Adipocyte 2, 217–226 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Yamauchi, T. et al. Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat. Med. 13, 332–339 (2007).

    Article  CAS  PubMed  Google Scholar 

  133. Chen, Z. et al. Effects of adiponectin on T2DM and glucose homeostasis: a mendelian randomization study. Diabetes Metab. Syndr. Obes. 13, 1771–1784 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Ortega Moreno, L. et al. Evidence of a causal relationship between high serum adiponectin levels and increased cardiovascular mortality rate in patients with type 2 diabetes. Cardiovasc. Diabetol. 15, 17 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Yore, M. M. et al. Discovery of a class of endogenous mammalian lipids with anti-diabetic and anti-inflammatory effects. Cell 159, 318–332 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Agbu, P. & Carthew, R. W. MicroRNA-mediated regulation of glucose and lipid metabolism. Nat. Rev. Mol. Cell Biol. 22, 425–438 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Crewe, C., An, Y. A. & Scherer, P. E. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J. Clin. Invest. 127, 74–82 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Hotamisligil, G. S. Inflammation and metabolic disorders. Nature 444, 860–867 (2006).

    Article  CAS  PubMed  Google Scholar 

  139. Kammoun, H. L., Kraakman, M. J. & Febbraio, M. A. Adipose tissue inflammation in glucose metabolism. Rev. Endocr. Metab. Disord. 15, 31–44 (2014).

    Article  CAS  PubMed  Google Scholar 

  140. McNelis, J. C. & Olefsky, J. M. Macrophages, immunity, and metabolic disease. Immunity 41, 36–48 (2014).

    Article  CAS  PubMed  Google Scholar 

  141. Mathis, D. Immunological goings-on in visceral adipose tissue. Cell Metab. 17, 851–859 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Hotamisligil, G. S. Inflammation, metaflammation and immunometabolic disorders. Nature 542, 177–185 (2017).

    Article  CAS  PubMed  Google Scholar 

  143. Wu, H. & Ballantyne, C. M. Metabolic inflammation and insulin resistance in obesity. Circ. Res. 126, 1549–1564 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Krogh-Madsen, R., Plomgaard, P., Møller, K., Mittendorfer, B. & Pedersen, B. K. Influence of TNF-alpha and IL-6 infusions on insulin sensitivity and expression of IL-18 in humans. Am. J. Physiol. Endocrinol. Metab. 291, E108–E114 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Kriketos, A. D. et al. Inflammation, insulin resistance, and adiposity: a study of first-degree relatives of type 2 diabetic subjects. Diabetes Care 27, 2033–2040 (2004).

    Article  CAS  PubMed  Google Scholar 

  146. Shimobayashi, M. et al. Insulin resistance causes inflammation in adipose tissue. J. Clin. Invest. 128, 1538–1550 (2018). A study that places IR ‘upstream’ of inflammation rather than the more conventional other way around.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Lee, Y. S. et al. Inflammation is necessary for long-term but not short-term high-fat diet-induced insulin resistance. Diabetes 60, 2474–2483 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Rafiq, S. et al. Gene variants influencing measures of inflammation or predisposing to autoimmune and inflammatory diseases are not associated with the risk of type 2 diabetes. Diabetologia 51, 2205–2213 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Everett, B. M. et al. Anti-inflammatory therapy with canakinumab for the prevention and management of diabetes. J. Am. Coll. Cardiol. 71, 2392–2401 (2018).

    Article  CAS  PubMed  Google Scholar 

  150. Ridker, P. M. et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 377, 1119–1131 (2017).

    Article  CAS  PubMed  Google Scholar 

  151. Smith, U., Li, Q., Rydén, M. & Spalding, K. L. Cellular senescence and its role in white adipose tissue. Int. J. Obes. 45, 934–943 (2021).

    Article  CAS  Google Scholar 

  152. Wernstedt Asterholm, I. et al. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 20, 103–118 (2014).

    Article  CAS  PubMed  Google Scholar 

  153. Cox, A. R., Chernis, N., Masschelin, P. M. & Hartig, S. M. Immune cells gate white adipose tissue expansion. Endocrinology 160, 1645–1658 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Chaurasia, B. & Summers, S. A. Ceramides - lipotoxic inducers of metabolic disorders. Trends Endocrinol. Metab. 26, 538–550 (2015).

    Article  CAS  PubMed  Google Scholar 

  155. Luukkonen, P. K. et al. Hepatic ceramides dissociate steatosis and insulin resistance in patients with non-alcoholic fatty liver disease. J. Hepatol. 64, 1167–1175 (2016).

    Article  CAS  PubMed  Google Scholar 

  156. Kolak, M. et al. Adipose tissue inflammation and increased ceramide content characterize subjects with high liver fat content independent of obesity. Diabetes 56, 1960–1968 (2007).

    Article  CAS  PubMed  Google Scholar 

  157. Coen, P. M. et al. Reduced skeletal muscle oxidative capacity and elevated ceramide but not diacylglycerol content in severe obesity. Obesity 21, 2362–2371 (2013).

    Article  CAS  PubMed  Google Scholar 

  158. Turpin, S. M. et al. Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metab. 20, 678–686 (2014). A study that together with Raichur et al. (2014) and Chaurasia et al. (2019) provides critical evidence implicating ceramide and ceramide metabolism in IR in mice.

    Article  CAS  PubMed  Google Scholar 

  159. Stöckli, J. et al. Metabolomic analysis of insulin resistance across different mouse strains and diets. J. Biol. Chem. 292, 19135–19145 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Raichur, S. et al. CerS2 haploinsufficiency inhibits β-oxidation and confers susceptibility to diet-induced steatohepatitis and insulin resistance. Cell Metab. 20, 687–695 (2014).

    Article  CAS  PubMed  Google Scholar 

  161. Chaurasia, B. et al. Targeting a ceramide double bond improves insulin resistance and hepatic steatosis. Science 365, 386–392 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Chavez, J. A. et al. A role for ceramide, but not diacylglycerol, in the antagonism of insulin signal transduction by saturated fatty acids. J. Biol. Chem. 278, 10297–10303 (2003).

    Article  CAS  PubMed  Google Scholar 

  163. Villa, N. Y. et al. Sphingolipids function as downstream effectors of a fungal PAQR. Mol. Pharmacol. 75, 866–875 (2009).

    Article  CAS  PubMed  Google Scholar 

  164. Mente, A. et al. Causal relationship between adiponectin and metabolic traits: a Mendelian randomization study in a multiethnic population. PLoS ONE 8, e66808 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Nawrocki, A. R. et al. Mice lacking adiponectin show decreased hepatic insulin sensitivity and reduced responsiveness to peroxisome proliferator-activated receptor gamma agonists. J. Biol. Chem. 281, 2654–2660 (2006).

    Article  CAS  PubMed  Google Scholar 

  166. Cazzolli, R., Carpenter, L., Biden, T. J. & Schmitz-Peiffer, C. A role for protein phosphatase 2A-like activity, but not atypical protein kinase Czeta, in the inhibition of protein kinase B/Akt and glycogen synthesis by palmitate. Diabetes 50, 2210–2218 (2001).

    Article  CAS  PubMed  Google Scholar 

  167. Fox, T. E. et al. Ceramide recruits and activates protein kinase C zeta (PKC zeta) within structured membrane microdomains. J. Biol. Chem. 282, 12450–12457 (2007).

    Article  CAS  PubMed  Google Scholar 

  168. Chaurasia, B., Talbot, C. L. & Summers, S. A. Adipocyte ceramides-the nexus of inflammation and metabolic disease. Front. Immunol. 11, 576347 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Lyu, K. et al. A membrane-bound diacylglycerol species induces PKCε-mediated hepatic insulin resistance. Cell Metab. 32, 654–664.e5 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Lyu, K. et al. Short-term overnutrition induces white adipose tissue insulin resistance through sn-1,2-diacylglycerol/PKCε/insulin receptor Thr1160 phosphorylation. JCI Insight 6, e139946 (2021).

    PubMed Central  Google Scholar 

  171. Gassaway, B. M. et al. PKCε contributes to lipid-induced insulin resistance through cross talk with p70S6K and through previously unknown regulators of insulin signaling. Proc. Natl Acad. Sci. USA 115, E8996–E9005 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Perreault, L. et al. Intracellular localization of diacylglycerols and sphingolipids influences insulin sensitivity and mitochondrial function in human skeletal muscle. JCI Insight 3, e96805 (2018).

    Article  PubMed Central  Google Scholar 

  173. Gonzalez-Franquesa, A. & Patti, M.-E. Insulin resistance and mitochondrial dysfunction. Adv. Exp. Med. Biol. 982, 465–520 (2017).

    Article  CAS  PubMed  Google Scholar 

  174. Sangwung, P., Petersen, K. F., Shulman, G. I. & Knowles, J. W. Mitochondrial dysfunction, insulin resistance, and potential genetic implications. Endocrinology 161, bqaa017 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Houstis, N., Rosen, E. D. & Lander, E. S. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 440, 944–948 (2006).

    Article  CAS  PubMed  Google Scholar 

  176. Fisher-Wellman, K. H. & Neufer, P. D. Linking mitochondrial bioenergetics to insulin resistance via redox biology. Trends Endocrinol. Metab. 23, 142–153 (2012). An excellent review of the role of mitochondrial bioenergetics and ROS in IR.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Hoehn, K. L. et al. Insulin resistance is a cellular antioxidant defense mechanism. Proc. Natl Acad. Sci. USA 106, 17787–17792 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Anderson, E. J. et al. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J. Clin. Invest. 119, 573–581 (2009). One of the more compelling studies implicating mitochondrial H2O2 as a possible cause of IR.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Ingram, K. H. et al. Skeletal muscle lipid peroxidation and insulin resistance in humans. J. Clin. Endocrinol. Metab. 97, E1182–E1186 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Duplain, H. et al. Stimulation of peroxynitrite catalysis improves insulin sensitivity in high fat diet-fed mice. J. Physiol. 586, 4011–4016 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Fazakerley, D. J. et al. Mitochondrial oxidative stress causes insulin resistance without disrupting oxidative phosphorylation. J. Biol. Chem. 293, 7315–7328 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Boveris, A., Oshino, N. & Chance, B. The cellular production of hydrogen peroxide. Biochem. J. 128, 617–630 (1972).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Rees-Milton, K. J. et al. Statin use is associated with insulin resistance in participants of the Canadian Multicentre Osteoporosis Study. J. Endocr. Soc. 4, bvaa057 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. Koves, T. R. et al. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 7, 45–56 (2008).

    Article  CAS  PubMed  Google Scholar 

  185. Di Paola, M., Cocco, T. & Lorusso, M. Ceramide interaction with the respiratory chain of heart mitochondria. Biochemistry 39, 6660–6668 (2000).

    Article  PubMed  CAS  Google Scholar 

  186. Smith, M. E. et al. Mitochondrial fission mediates ceramide-induced metabolic disruption in skeletal muscle. Biochem. J. 456, 427–439 (2013).

    Article  CAS  PubMed  Google Scholar 

  187. Novgorodov, S. A. et al. Novel pathway of ceramide production in mitochondria: thioesterase and neutral ceramidase produce ceramide from sphingosine and acyl-CoA. J. Biol. Chem. 286, 25352–25362 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. von Haefen, C. et al. Ceramide induces mitochondrial activation and apoptosis via a Bax-dependent pathway in human carcinoma cells. Oncogene 21, 4009–4019 (2002).

    Article  CAS  Google Scholar 

  189. Ye, R., Onodera, T. & Scherer, P. E. Lipotoxicity and cell maintenance in obesity and type 2 diabetes. J. Endocr. Soc. 3, 617–631 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Turpin, S. M. et al. Examination of ‘lipotoxicity’ in skeletal muscle of high-fat fed and ob/ob mice. J. Physiol. 587, 1593–1605 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Kim, Y.-R. et al. Hepatic triglyceride accumulation via endoplasmic reticulum stress-induced SREBP-1 activation is regulated by ceramide synthases. Exp. Mol. Med. 51, 1–16 (2019).

    PubMed  PubMed Central  Google Scholar 

  192. Boslem, E. et al. A lipidomic screen of palmitate-treated MIN6 β-cells links sphingolipid metabolites with endoplasmic reticulum (ER) stress and impaired protein trafficking. Biochem. J. 435, 267–276 (2011).

    Article  CAS  PubMed  Google Scholar 

  193. Flamment, M., Hajduch, E., Ferré, P. & Foufelle, F. New insights into ER stress-induced insulin resistance. Trends Endocrinol. Metab. 23, 381–390 (2012).

    Article  CAS  PubMed  Google Scholar 

  194. Sumitomo, M. et al. Protein kinase Cdelta amplifies ceramide formation via mitochondrial signaling in prostate cancer cells. J. Clin. Invest. 109, 827–836 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Lee, H.-Y. et al. Mitochondrial-targeted catalase protects against high-fat diet-induced muscle insulin resistance by decreasing intramuscular lipid accumulation. Diabetes 66, 2072–2081 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Riojas-Hernández, A. et al. Enhanced oxidative stress sensitizes the mitochondrial permeability transition pore to opening in heart from Zucker Fa/fa rats with type 2 diabetes. Life Sci. 141, 32–43 (2015).

    Article  PubMed  CAS  Google Scholar 

  197. Taddeo, E. P. et al. Opening of the mitochondrial permeability transition pore links mitochondrial dysfunction to insulin resistance in skeletal muscle. Mol. Metab. 3, 124–134 (2014).

    Article  CAS  PubMed  Google Scholar 

  198. Cho, J. et al. Mitochondrial ATP transporter depletion protects mice against liver steatosis and insulin resistance. Nat. Commun. 8, 14477 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. O’Neill, B. T. et al. Differential role of insulin/IGF-1 receptor signaling in muscle growth and glucose homeostasis. Cell Rep. 11, 1220–1235 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Gastaldelli, A. et al. Effect of physiological hyperinsulinemia on gluconeogenesis in nondiabetic subjects and in type 2 diabetic patients. Diabetes 50, 1807–1812 (2001).

    Article  CAS  PubMed  Google Scholar 

  201. Reaven, G. M., Hollenbeck, C. B. & Chen, Y. D. Relationship between glucose tolerance, insulin secretion, and insulin action in non-obese individuals with varying degrees of glucose tolerance. Diabetologia 32, 52–55 (1989).

    Article  CAS  PubMed  Google Scholar 

  202. Firth, R., Bell, P. & Rizza, R. Insulin action in non-insulin-dependent diabetes mellitus: the relationship between hepatic and extrahepatic insulin resistance and obesity. Metabolism 36, 1091–1095 (1987).

    Article  CAS  PubMed  Google Scholar 

  203. Manning, B. D. & Toker, A. AKT/PKB signaling: navigating the network. Cell 169, 381–405 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. James, D. E., Strube, M. & Mueckler, M. Molecular cloning and characterization of an insulin-regulatable glucose transporter. Nature 338, 83–87 (1989).

    Article  CAS  PubMed  Google Scholar 

  205. Birnbaum, M. J. Identification of a novel gene encoding an insulin-responsive glucose transporter protein. Cell 57, 305–315 (1989).

    Article  CAS  PubMed  Google Scholar 

  206. Bryant, N. J., Govers, R. & James, D. E. Regulated transport of the glucose transporter GLUT4. Nat. Rev. Mol. Cell Biol. 3, 267–277 (2002).

    Article  CAS  PubMed  Google Scholar 

  207. Yeh, J. I., Gulve, E. A., Rameh, L. & Birnbaum, M. J. The effects of wortmannin on rat skeletal muscle. Dissociation of signaling pathways for insulin- and contraction-activated hexose transport. J. Biol. Chem. 270, 2107–2111 (1995).

    Article  CAS  PubMed  Google Scholar 

  208. Sano, H. et al. Rab10, a target of the AS160 Rab GAP, is required for insulin-stimulated translocation of GLUT4 to the adipocyte plasma membrane. Cell Metab. 5, 293–303 (2007).

    Article  CAS  PubMed  Google Scholar 

  209. Stöckli, J., Fazakerley, D. J. & James, D. E. GLUT4 exocytosis. J. Cell Sci. 124, 4147–4159 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  210. Kirwan, J. P., Solomon, T. P. J., Wojta, D. M., Staten, M. A. & Holloszy, J. O. Effects of 7 days of exercise training on insulin sensitivity and responsiveness in type 2 diabetes mellitus. Am. J. Physiol. Endocrinol. Metab. 297, E151–E156 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. James, D. E., Kraegen, E. W. & Chisholm, D. J. Effects of exercise training on in vivo insulin action in individual tissues of the rat. J. Clin. Invest. 76, 657–666 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Koh, J.-H. et al. PPARβ is essential for maintaining normal levels of PGC-1α and mitochondria and for the increase in muscle mitochondria induced by exercise. Cell Metab. 25, 1176–1185.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Alibegovic, A. C. et al. Impact of 9 days of bed rest on hepatic and peripheral insulin action, insulin secretion, and whole-body lipolysis in healthy young male offspring of patients with type 2 diabetes. Diabetes 58, 2749–2756 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Mikines, K. J., Richter, E. A., Dela, F. & Galbo, H. Seven days of bed rest decrease insulin action on glucose uptake in leg and whole body. J. Appl. Physiol. 70, 1245–1254 (1991).

    Article  CAS  PubMed  Google Scholar 

  215. Krogh-Madsen, R. et al. A 2-wk reduction of ambulatory activity attenuates peripheral insulin sensitivity. J. Appl. Physiol. 108, 1034–1040 (2010).

    Article  CAS  PubMed  Google Scholar 

  216. Grams, J. & Garvey, W. T. Weight loss and the prevention and treatment of type 2 diabetes using lifestyle therapy, pharmacotherapy, and bariatric surgery: mechanisms action. Curr. Obes. Rep. 4, 287–302 (2015).

    Article  CAS  PubMed  Google Scholar 

  217. Lean, M. E. et al. Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial. Lancet 391, 541–551 (2018).

    Article  PubMed  Google Scholar 

  218. Magkos, F. et al. Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity. Cell Metab. 23, 591–601 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Bray, G. A. & Bouchard, C. The biology of human overfeeding: a systematic review. Obes. Rev. 21, e13040 (2020).

    Article  CAS  PubMed  Google Scholar 

  220. Smith, G. I. et al. One day of mixed meal overfeeding reduces hepatic insulin sensitivity and increases VLDL particle but not VLDL-triglyceride secretion in overweight and obese men. J. Clin. Endocrinol. Metab. 98, 3454–3462 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. McLaughlin, T. et al. Adipose cell size and regional fat deposition as predictors of metabolic response to overfeeding in insulin-resistant and insulin-sensitive humans. Diabetes 65, 1245–1254 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Ludwig, D. S., Willett, W. C., Volek, J. S. & Neuhouser, M. L. Dietary fat: from foe to friend? Science 362, 764–770 (2018).

    Article  CAS  PubMed  Google Scholar 

  223. Weickert, M. O. et al. Effects of supplemented isoenergetic diets differing in cereal fiber and protein content on insulin sensitivity in overweight humans. Am. J. Clin. Nutr. 94, 459–471 (2011).

    Article  CAS  PubMed  Google Scholar 

  224. Linn, T. et al. Effect of long-term dietary protein intake on glucose metabolism in humans. Diabetologia 43, 1257–1265 (2000).

    Article  CAS  PubMed  Google Scholar 

  225. Sluijs, I. et al. Dietary intake of total, animal, and vegetable protein and risk of type 2 diabetes in the European Prospective Investigation into Cancer and Nutrition (EPIC)-NL study. Diabetes Care 33, 43–48 (2010).

    Article  CAS  PubMed  Google Scholar 

  226. Rosenbaum, M. et al. Glucose and lipid homeostasis and inflammation in humans following an isocaloric ketogenic diet. Obesity 27, 971–981 (2019).

    Article  CAS  PubMed  Google Scholar 

  227. Roberts, M. N. et al. A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab. 26, 539–546.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Bisschop, P. H. et al. Dietary fat content alters insulin-mediated glucose metabolism in healthy men. Am. J. Clin. Nutr. 73, 554–559 (2001).

    Article  CAS  PubMed  Google Scholar 

  229. Lundsgaard, A.-M. et al. Mechanisms preserving insulin action during high dietary fat intake. Cell Metab. 29, 50–63.e4 (2019).

    Article  CAS  PubMed  Google Scholar 

  230. Sargrad, K. R., Homko, C., Mozzoli, M. & Boden, G. Effect of high protein vs high carbohydrate intake on insulin sensitivity, body weight, hemoglobin A1c, and blood pressure in patients with type 2 diabetes mellitus. J. Am. Diet. Assoc. 105, 573–580 (2005).

    Article  CAS  PubMed  Google Scholar 

  231. Seidelmann, S. B. et al. Dietary carbohydrate intake and mortality: a prospective cohort study and meta-analysis. Lancet Public. Health 3, e419–e428 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  232. Randle, P. J. Fuel selection in animals. Biochem. Soc. Trans. 14, 799–806 (1986).

    Article  CAS  PubMed  Google Scholar 

  233. Moltke, I. et al. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Nature 512, 190–193 (2014). Human genetic evidence supporting a role for the AKT target TBC1D4 in the regulation of muscle glucose transport.

    Article  CAS  PubMed  Google Scholar 

  234. Manousaki, D. et al. Toward precision medicine: TBC1D4 disruption is common among the inuit and leads to underdiagnosis of type 2 diabetes. Diabetes Care 39, 1889–1895 (2016).

    Article  CAS  PubMed  Google Scholar 

  235. Palla, L., Higgins, J. P. T., Wareham, N. J. & Sharp, S. J. Challenges in the use of literature-based meta-analysis to examine gene-environment interactions. Am. J. Epidemiol. 171, 1225–1232 (2010).

    Article  PubMed  Google Scholar 

  236. Martínez-González, M. A. et al. Benefits of the Mediterranean diet: insights from the PREDIMED study. Prog. Cardiovasc. Dis. 58, 50–60 (2015).

    Article  PubMed  Google Scholar 

  237. Kilpeläinen, T. O. et al. Physical activity attenuates the influence of FTO variants on obesity risk: a meta-analysis of 218,166 adults and 19,268 children. PLoS Med. 8, e1001116 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  238. Sonestedt, E. et al. Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity. Am. J. Clin. Nutr. 90, 1418–1425 (2009).

    Article  CAS  PubMed  Google Scholar 

  239. Heianza, Y. et al. Macronutrient intake-associated FGF21 genotype modifies effects of weight-loss diets on 2-year changes of central adiposity and body composition: the POUNDS lost trial. Diabetes Care 39, 1909–1914 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Hall, K. D. Challenges of human nutrition research. Science 367, 1298–1300 (2020).

    Article  CAS  PubMed  Google Scholar 

  241. Franz, M. J. et al. Academy of nutrition and dietetics nutrition practice guideline for type 1 and type 2 diabetes in adults: systematic review of evidence for medical nutrition therapy effectiveness and recommendations for integration into the nutrition care process. J. Acad. Nutr. Diet. 117, 1659–1679 (2017).

    Article  PubMed  Google Scholar 

  242. Powell, K. E. et al. The scientific foundation for the Physical Activity Guidelines for Americans, 2nd Edition. J. Phys. Act. Health 16, 1–11 (2018).

    Article  Google Scholar 

  243. American Diabetes Association. 5. Facilitating behavior change and well-being to improve health outcomes: standards of medical care in diabetes-2020. Diabetes Care 43, S48–S65 (2020).

    Article  Google Scholar 

  244. Pernicova, I. & Korbonits, M. Metformin — mode of action and clinical implications for diabetes and cancer. Nat. Rev. Endocrinol. 10, 143–156 (2014).

    Article  CAS  PubMed  Google Scholar 

  245. American Diabetes Association. 9. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes — 2020. Diabetes Care 43, S98–S110 (2020).

    Article  Google Scholar 

  246. Packer, M. Potentiation of insulin signaling contributes to heart failure in type 2 diabetes: a hypothesis supported by both mechanistic studies and clinical trials. JACC Basic Transl. Sci. 3, 415–419 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  247. Cariou, B., Charbonnel, B. & Staels, B. Thiazolidinediones and PPARγ agonists: time for a reassessment. Trends Endocrinol. Metab. 23, 205–215 (2012).

    Article  CAS  PubMed  Google Scholar 

  248. Steinberg, G. R. & Carling, D. AMP-activated protein kinase: the current landscape for drug development. Nat. Rev. Drug Discov. 18, 527–551 (2019).

    Article  CAS  PubMed  Google Scholar 

  249. Myers, R. W. et al. Systemic pan-AMPK activator MK-8722 improves glucose homeostasis but induces cardiac hypertrophy. Science 357, 507–511 (2017).

    Article  CAS  PubMed  Google Scholar 

  250. Esquejo, R. M. et al. Activation of liver AMPK with PF-06409577 corrects NAFLD and lowers cholesterol in rodent and primate preclinical models. EBioMedicine 31, 122–132 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  251. Cokorinos, E. C. et al. Activation of skeletal muscle AMPK promotes glucose disposal and glucose lowering in non-human primates and mice. Cell Metab. 25, 1147–1159.e10 (2017).

    Article  CAS  PubMed  Google Scholar 

  252. Lee, C. et al. The mitochondrial-derived peptide MOTS-c promotes metabolic homeostasis and reduces obesity and insulin resistance. Cell Metab. 21, 443–454 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Steneberg, P. et al. PAN-AMPK activator O304 improves glucose homeostasis and microvascular perfusion in mice and type 2 diabetes patients. JCI Insight 3, e99114 (2018).

    Article  PubMed Central  Google Scholar 

  254. Dassano, A., Loretelli, C. & Fiorina, P. Idebenone and T2D: A new insulin-sensitizing drug for personalized therapy. Pharmacol. Res. 139, 469–470 (2019).

    Article  CAS  PubMed  Google Scholar 

  255. Harrison, S. A. et al. Insulin sensitizer MSDC-0602K in non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled phase IIb study. J. Hepatol. 72, 613–626 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

D.E.J. is an Australian Research Council Laureate Fellow. The authors are extremely grateful to S. Klein for his thoughtful discussion of the manuscript and to P. Titchenell, G. Cooney, A. Diaz and M. Nelson for carefully reading and commenting on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to David E. James or Morris J. Birnbaum.

Additional information

Competing interests

D.E.J. and J.S. declare no competing interests. M.J.B. is a full-time employee and stockholder of Pfizer Inc. and sits on the Board of Directors of Cerevel Therapeutics.

Peer review information

Nature Reviews Molecular Cell Biology thanks M. Czech, B. Goodpaster, T. Kadowaki, D. Neufer and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Supplementary information

Glossary

Hepatic glucose output

Refers to the release of glucose from the liver into the bloodstream that is upregulated during fasting and suppressed with insulin. It is mediated via the breakdown of liver glycogen (glycogenolysis) or recycling of breakdown products such as lactate from peripheral tissues (gluconeogenesis).

Impaired fasting glucose

According to the American Diabetes Association, this is defined as fasting plasma glucose levels between 100 and 125 mg dl−1 (between 5.6 and 6.9 mmol l−1).

Impaired glucose tolerance

According to the American Diabetes Association, this is defined as 2-h plasma glucose levels between 140 and 199 mg dl−1 (between 7.8 and 11.0 mmol l−1) measured during a 75-g oral glucose tolerance test.

Relative hyperinsulinaemia

The situation where individuals with type 2 diabetes despite having insufficient insulin secretion to normalize their hyperglycaemia still display significantly higher circulating insulin levels than those observed in healthy people.

Gluconeogenesis

A metabolic pathway that is particularly important in liver metabolism in which glucose is regenerated essentially via reverse glycolysis from a range of substrates, such as lactate, amino acids, glycerol or acetate.

Metabolic diseases

Diseases that exhibit dysregulated metabolism of carbohydrates and lipids. Metabolic diseases include type 2 diabetes, cardiovascular disease and liver disease.

14-3-3 proteins

A ubiquitously expressed family of proteins comprising seven members in mammals that bind to Ser/Thr phosphorylation motifs and are thought to be one of the major conduits by which kinases such as AKT regulate the function of substrates.

Retromer

A heteropentameric complex of proteins originally discovered in yeast to regulate protein trafficking between endosomes and the trans-Golgi network

Fat depots

Distinct fat depots are named after their location. Subcutaneous fat is the layer of fat found just beneath the skin, whereas visceral or abdominal fat is located in the peritoneal cavity. Subcutaneous fat serves as an energy store, a protective layer of tissue and a potential source of heat for thermoregulation. Visceral fat plays a more important role in releasing fat into the circulation for use by other tissues.

Lipodystrophy

A condition whereby individuals are unable to produce adipose tissue leading to excessive deposition of fat ectopically. The two common forms are congenital, caused by mutations in genes that regulate adipogenesis, or acquired. The most common form of acquired lipodystrophy is caused by older generations of antiretroviral drugs used to treat HIV. Both forms are associated with insulin resistance and other metabolic disorders.

Insulinomas

Small pancreatic tumours that produce and secrete excess insulin into the circulation. This is a rare endocrine disorder that is treated surgically.

Western diet

A diet that generally contains a high content (40–60%) of fat and is rich in simple sugars (sucrose).

C57BL/6J mice

Inbred mouse strain that is the preferred mouse model used in metabolic research to study the effects of a Western diet and for genetic manipulations.

Metabolic flux

The rate at which metabolites are metabolized by intracellular pathways. The metabolic flux is modulated by changing the levels or activity of metabolic enzymes, or by altering the supply or demand of the metabolite.

TBC1D4

A RAB GTPase-activating protein that regulates the GTPase RAB10 and several other RABs. It is phosphorylated by AKT and is integral to insulin regulation of glucose transport in muscle and fat.

Novel PKCs

A subclass of protein kinase C (PKC), a family of Ser/Thr kinases that are activated by diacylglycerols (DAGs) without the need for Ca2+.

JNK

JUN N-terminal kinase is a Ser/Thr kinase that belongs to the family of mitogen-activated protein kinases (MAPKs), which respond to stress stimuli, including cytokines.

mTOR

A Ser/Thr kinase that plays a major role in nutrient sensing in all eukaryotes.

S6 kinase

A Ser/Thr kinase named after its best-known substrate, the ribosomal protein S6. S6 kinase is activated by mTOR, and phosphorylation of its substrate S6 induces protein synthesis.

PDGF receptor

(PDGFR). A receptor that, like the insulin receptor, belongs to the receptor tyrosine kinase family of cell surface receptors. PDGF binding to PDGFR triggers a signal transduction pathway analogous to insulin, involving PI3K and AKT. Importantly, PDGF signalling bypasses IRS and thus provides a tool to test the role of IRS in insulin resistance.

Carbonylation

A protein oxidation reaction that is commonly found on the amino acids lysine and arginine, among other amino acids.

Lipid peroxidation

A process whereby oxidants attack (unsaturated) lipids and via multiple reactions this results in the formation of toxic and/or mutagenic lipid hydroperoxides. The lipid peroxidation process is a chain reaction that propagates until it is terminated by antioxidants.

Adipokine

A molecule, such as a protein, a lipid, a metabolite or a microRNA, that is secreted from adipose tissue to potentially regulate functions in other tissues. It is notable that most adipokines are not produced by adipocytes but derive from stromal cells in the adipose tissue.

Beige adipocytes

A subtype of thermogenic adipocytes that arises in white adipose tissue, most commonly in subcutaneous fat, in response to various stimuli, such as cold exposure, catecholamines, exercise, thiazolidinediones and injury. They are capable of uncoupled mitochondrial respiration via the activity of uncoupling proteins such as UCP1 to produce heat at the expense of ATP, and their emergence has been associated with improved metabolic health.

Leptin

The first adipokine to be discovered, by Friedman and colleagues in 1994. The obese (ob) gene long known to regulate appetite in rodents was found to encode leptin and was subsequently shown to play a key role in metabolic homeostasis and the starvation response in mammals.

Regulatory T cells

A subpopulation of T cells that are involved in immune function.

C-reactive protein

(CRP). A protein secreted primarily by the liver in response to inflammatory cytokines. In the clinic, circulating levels of CRP are a measure of inflammation or infection.

Genome-wide association studies

A statistical method that identifies genetic variants that are associated with a particular trait, disease or phenotype.

Prediabetes

The clinical term to describe individuals who exhibit ‘impaired glucose tolerance’ but do not fulfil the requirements for type 2 diabetes classification.

Ceramides

A class of lipids composed of sphingosine and a fatty acid. Distinct ceramide species contain different fatty acids with various carbon chain lengths, and the ceramides are distinguished and named after the carbon length of the fatty acid.

AMPK

Adenosine 5′-monophosphate (AMP)-activated kinase. AMPK is activated in most cells in response to cellular and/or metabolic stress to buffer cellular energy charge.

Diacylglycerols

(DAGs). Two fatty acid moieties linked to glycerol. DAGs were first identified through their role in binding and activating various members of the protein kinase C (PKC) family.

Oxidative phosphorylation

(OXPHOS). The most notorious function of mitochondria, which is to oxidize various nutrients such as fat and carbohydrate via a series of complex chemical reactions to create the basic energy unit ATP.

Mitophagy

The engulfment of mitochondria by an autophagosome, undergoing autophagy. Dysfunctional mitochondria are targeted for mitophagy for removal and degradation. Mitochondrial fission is necessary for mitophagy.

Mitochondrial fission

A process of mitochondrial fragmentation catalysed by regulated fission machinery. Mitochondrial fission plays an essential role in the removal of damaged mitochondria from cells but has also been associated with reduced bioenergetics.

Coenzyme Q

(CoQ). Also known as ubiquinone, this coenzyme plays an essential role in transporting electrons between various components of the electron transport chain and so is essential to mitochondrial energy production. CoQ also serves as an antioxidant.

Acylcarnitines

A form of fatty acid that are conjugated to carnitine to be transported into the mitochondria for oxidative metabolism.

Endoplasmic reticulum stress

Endoplasmic reticulum (ER) stress is a form of stress in which the ability of the ER to fold proteins is exceeded, leading to the initiation of the ER stress response, an ancient signal transduction pathway that initiates a series of events to help clear the ER of misfolded proteins.

Sodium–glucose co-transporter 2

Also known as SGLT2 or SLC5A2, a transmembrane glucose transporter protein that is predominantly responsible for glucose reabsorption in the kidney.

Glucagon-like peptide 1

(GLP1). A small peptide hormone derived from the proglucagon protein that is secreted from enteroendocrine L cells in the intestine. Among its functions is an incretin activity, which promotes insulin secretion from the pancreas.

Mitochondrial permeability transition pore

(mPTP). A multisubunit protein complex that forms a non-specific channel in the inner mitochondrial membrane. The mPTP opens in response to calcium and/or oxidative stress and allows transit of molecules of up to 1,500 Da from inside mitochondria to the cytosol.

Palmitate

The most prevalent saturated fatty acid found in animals. It is obtained either from dietary sources or by biosynthesis from de novo lipogenesis. It contains an aliphatic tail with a chain length of 16 carbons.

Insulin-like growth factor 1

A hormone that is very similar to insulin that plays a central role in early development in mammals and probably other animals. It is produced and secreted by the liver in response to growth hormone.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

James, D.E., Stöckli, J. & Birnbaum, M.J. The aetiology and molecular landscape of insulin resistance. Nat Rev Mol Cell Biol 22, 751–771 (2021). https://doi.org/10.1038/s41580-021-00390-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-021-00390-6

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