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:

Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes

Key Points

  • Insulin resistance due to obesity is a primary event in the development of type 2 diabetes. Recent work indicates a central role of adipose tissue dysfunction in linking obesity to insulin resistance.

  • Fatty acids and their derivatives are mediators of insulin resistance in skeletal muscle, probably through deleterious effects on the insulin signalling pathway. Increased exposure of skeletal muscle tissue to elevated fatty acids in rodents and humans impairs insulin-stimulated glucose uptake.

  • The impaired ability of adipose tissue to sequester fatty acids in triglyceride stores results in increasing fatty acid concentrations in the circulation and the exposure of skeletal muscle to these high fatty acid levels. This impairment of adipose function can be caused by a chronic inflammatory state that arises within adipose tissue in obese animals and humans.

  • Inflammatory cytokines, including tumour-necrosis factor-α (TNFα), have profound effects on adipocyte metabolism by impairing triglyceride synthesis and storage, and promoting the hydrolysis and release of triglycerides as free fatty acids. These effects are mediated in part through downregulation of the key adipocyte transcription factor PPARγ (peroxisome proliferator-activated receptor-γ).

  • Recent data implicate lipid-droplet proteins, including newly described CIDE family proteins, in the promotion of triglyceride storage and as significant targets of PPARγ regulation. Through effects on these proteins, downregulation of PPARγ can mediate diminished lipid storage ability of inflamed adipose tissue. Expression of these proteins may be determinants of the differential ability to sequester fat away from the circulation and the propensity of obese humans to develop insulin resistance.

Abstract

Acquired resistance to the action of insulin to stimulate glucose transport in skeletal muscle is associated with obesity and promotes the development of type 2 diabetes. In skeletal muscle, insulin resistance can result from high levels of circulating fatty acids that disrupt insulin signalling pathways. However, the severity of insulin resistance varies greatly among obese people. Here we postulate that this variability might reflect differences in levels of lipid-droplet proteins that promote the sequestration of fatty acids within adipocytes in the form of triglycerides, thereby lowering exposure of skeletal muscle to the inhibitory effects of fatty acids.

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

Figure 1: Chronic inflammation in adipose tissue triggers insulin resistance in skeletal muscle.
Figure 2: Chronic inflammation impairs triglyceride deposition in adipose tissue.
Figure 3: PPARγ downregulation by TNFα impairs triglyceride storage in adipose cells.
Figure 4: TNFα decreases triglyceride deposition and increases lipolysis in adipose cells.
Figure 5: TNFα downregulates lipid-droplet proteins and enhances lipolysis in adipose cells.

Similar content being viewed by others

References

  1. Flier, J. S. Obesity wars: molecular progress confronts an expanding epidemic. Cell 116, 337–350 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Hogan, P., Dall, T. & Nikolov, P. Economic costs of diabetes in the US in 2002. Diabetes Care 26, 917–932 (2003).

    Article  PubMed  Google Scholar 

  3. Alberti, K. G. The costs of non-insulin-dependent diabetes mellitus. Diabet. Med. 14, 7–9 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Clee, S. M. & Attie, A. D. The genetic landscape of type 2 diabetes in mice. Endocr. Rev. 28, 48–83 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Sims, E. A. et al. Endocrine and metabolic effects of experimental obesity in man. Recent Prog. Horm. Res. 29, 457–496 (1973).

    CAS  PubMed  Google Scholar 

  6. Freidenberg, G. R., Reichart, D., Olefsky, J. M. & Henry, R. R. Reversibility of defective adipocyte insulin receptor kinase activity in non-insulin-dependent diabetes mellitus. Effect of weight loss. J. Clin. Invest. 82, 1398–1406 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kahn, S. E. The relative contributions of insulin resistance and β-cell dysfunction to the pathophysiology of Type 2 diabetes. Diabetologia 46, 3–19 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Butler, A. E. et al. β-cell deficit and increased β-cell apoptosis in humans with type 2 diabetes. Diabetes 52, 102–110 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Rhodes, C. J. Type 2 diabetes—a matter of β-cell life and death? Science 307, 380–384 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Unger, R. H. Lipotoxicity in the pathogenesis of obesity-dependent NIDDM. Genetic and clinical implications. Diabetes 44, 863–870 (1995).

    Article  CAS  PubMed  Google Scholar 

  11. Unger, R. H. Lipotoxic diseases. Annu. Rev. Med. 53, 319–336 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Savage, D. B., Petersen, K. F. & Shulman, G. I. Disordered lipid metabolism and the pathogenesis of insulin resistance. Physiol. Rev. 87, 507–520 (2007). A recent review focusing on studies of lipid effects on insulin resistance in human subjects.

    Article  CAS  PubMed  Google Scholar 

  13. Boden, G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes 46, 3–10 (1997). Demonstrates that infusion of free fatty acids acutely induces insulin resistance in human subjects.

    Article  CAS  PubMed  Google Scholar 

  14. Kelley, D. E., Mokan, M., Simoneau, J. A. & Mandarino, L. J. Interaction between glucose and free fatty acid metabolism in human skeletal muscle. J. Clin. Invest. 92, 91–98 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Santomauro, A. T. et al. Overnight lowering of free fatty acids with Acipimox improves insulin resistance and glucose tolerance in obese diabetic and nondiabetic subjects. Diabetes 48, 1836–1841 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Oakes, N. D. et al. Diet-induced muscle insulin resistance in rats is ameliorated by acute dietary lipid withdrawal or a single bout of exercise: parallel relationship between insulin stimulation of glucose uptake and suppression of long-chain fatty acyl-CoA. Diabetes 46, 2022–2028 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Goodpaster, B. H., Thaete, F. L., Simoneau, J. A. & Kelley, D. E. Subcutaneous abdominal fat and thigh muscle composition predict insulin sensitivity independently of visceral fat. Diabetes 46, 1579–1585 (1997).

    Article  CAS  PubMed  Google Scholar 

  18. Perseghin, G. et al. Intramyocellular triglyceride content is a determinant of in vivo insulin resistance in humans: a 1H-13C nuclear magnetic resonance spectroscopy assessment in offspring of type 2 diabetic parents. Diabetes 48, 1600–1606 (1999).

    Article  CAS  PubMed  Google Scholar 

  19. Krssak, M. et al. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia 42, 113–116 (1999).

    Article  CAS  PubMed  Google Scholar 

  20. Szczepaniak, L. S. et al. Measurement of intracellular triglyceride stores by H spectroscopy: validation in vivo. Am. J. Physiol. 276, E977–E989 (1999).

    CAS  PubMed  Google Scholar 

  21. Sovik, O., Vestergaard, H., Trygstad, O. & Pedersen, O. Studies of insulin resistance in congenital generalized lipodystrophy. Acta Paediatr. Suppl. 413, 29–37 (1996).

    Article  CAS  PubMed  Google Scholar 

  22. Moitra, J. et al. Life without white fat: a transgenic mouse. Genes Dev. 12, 3168–3181 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Laustsen, P. G. et al. Lipoatrophic diabetes in Irs1−/−/Irs3−/− double knockout mice. Genes Dev. 16, 3213–3222 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Shimomura, I. et al. Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adipose tissue: model for congenital generalized lipodystrophy. Genes Dev. 12, 3182–3194 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Arioglu, E., Rother, K. I., Reitman, M. L., Premkumar, A. & Taylor, S. I. Lipoatrophy syndromes: when 'too little fat' is a clinical problem. Pediatr. Diabetes 1, 155–168 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Oral, E. A. et al. Leptin-replacement therapy for lipodystrophy. N. Engl. J. Med. 346, 570–578 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Ahima, R. S. & Flier, J. S. Leptin. Annu. Rev. Physiol. 62, 413–437 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Berg, A. H., Combs, T. P. & Scherer, P. E. ACRP30/adiponectin: an adipokine regulating glucose and lipid metabolism. Trends Endocrinol. Metab. 13, 84–89 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Unger, R. H. & Orci, L. Diseases of liporegulation: new perspective on obesity and related disorders. FASEB J. 15, 312–321 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Rosen, E. D. & Spiegelman, B. M. Adipocytes as regulators of energy balance and glucose homeostasis. Nature 444, 847–853 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kalderon, B., Mayorek, N., Berry, E., Zevit, N. & Bar-Tana, J. Fatty acid cycling in the fasting rat. Am. J. Physiol. Endocrinol. Metab. 279, E221–E227 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Christianson, J. L., Nicoloro, S., Straubhaar, J. & Czech, M. P. Stearoyl CoA desaturase 2 is required for PPARγ expression and adipogenesis in cultured 3T3-L1 cells. J. Biol. Chem. 283, 2906–2916 (2007).

    Article  PubMed  CAS  Google Scholar 

  33. Frayn, K. N. et al. Regulation of fatty acid movement in human adipose tissue in the postabsorptive-to-postprandial transition. Am. J. Physiol. 266, E308–E317 (1994).

    CAS  PubMed  Google Scholar 

  34. Qatanani, M. & Lazar, M. A. Mechanisms of obesity-associated insulin resistance: many choices on the menu. Genes Dev. 21, 1443–1455 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Rajala, M. W. & Scherer, P. E. Minireview: the adipocyte-at the crossroads of energy homeostasis, inflammation, and atherosclerosis. Endocrinology 144, 3765–3773 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Kershaw, E. E. & Flier, J. S. Adipose tissue as an endocrine organ. J. Clin. Endocrinol. Metab. 89, 2548–2556 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Sartipy, P. & Loskutoff, D. J. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc. Natl Acad. Sci. USA 100, 7265–7270 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Xu, H. et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Invest. 112, 1821–1830 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Curat, C. A. et al. From blood monocytes to adipose tissue-resident macrophages: induction of diapedesis by human mature adipocytes. Diabetes 53, 1285–1292 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Inouye, K. E. et al. Absence of CC chemokine ligand 2 does not limit obesity-associated infiltration of macrophages into adipose tissue. Diabetes 56, 2242–2250 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Lagathu, C. et al. Long-term treatment with interleukin-1β induces insulin resistance in murine and human adipocytes. Diabetologia 49, 2162–2173 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Kanda, H. et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J. Clin. Invest. 116, 1494–1505 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Uysal, K. T., Wiesbrock, S. M., Marino, M. W. & Hotamisligil, G. S. Protection from obesity-induced insulin resistance in mice lacking TNF-α function. Nature 389, 610–614 (1997). Strongly suggests a role for an inflammatory cytokine in mediating insulin resistance.

    Article  CAS  PubMed  Google Scholar 

  45. Solinas, G. et al. JNK1 in hematopoietically derived cells contributes to diet-induced inflammation and insulin resistance without affecting obesity. Cell Metab. 6, 386–397 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Harman-Boehm, I. et al. Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity. J. Clin. Endocrinol. Metab. 92, 2240–2247 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Bruun, J. M., Helge, J. W., Richelsen, B. & Stallknecht, B. Diet and exercise reduce low-grade inflammation and macrophage infiltration in adipose tissue but not in skeletal muscle in severely obese subjects. Am. J. Physiol. Endocrinol. Metab. 290, E961–E967 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Cancello, R. et al. Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes 54, 2277–2286 (2005).

    Article  CAS  PubMed  Google Scholar 

  49. Bouzakri, K. & Zierath, J. R. MAP4K4 gene silencing in human skeletal muscle prevents tumor necrosis factor-α-induced insulin resistance. J. Biol. Chem. 282, 7783–7789 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Tang, X. et al. An RNA interference-based screen identifies MAP4K4/NIK as a negative regulator of PPARγ, adipogenesis, and insulin-responsive hexose transport. Proc. Natl Acad. Sci. USA 103, 2087–2092 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Tesz, G. J. et al. Tumor necrosis factor α (TNFα) stimulates Map4k4 expression through TNFα receptor 1 signaling to c-Jun and activating transcription factor 2. J. Biol. Chem. 282, 19302–19312 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Shulman, G. I. Cellular mechanisms of insulin resistance. J. Clin. Invest. 106, 171–176 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Biddinger, S. B. & Kahn, C. R. From mice to men: insights into the insulin resistance syndromes. Annu. Rev. Physiol. 68, 123–158 (2006).

    Article  CAS  PubMed  Google Scholar 

  54. Ofei, F., Hurel, S., Newkirk, J., Sopwith, M. & Taylor, R. Effects of an engineered human anti-TNF-α antibody (CDP571) on insulin sensitivity and glycemic control in patients with NIDDM. Diabetes 45, 881–885 (1996).

    Article  PubMed  Google Scholar 

  55. Paquot, N., Castillo, M. J., Lefebvre, P. J. & Scheen, A. J. No increased insulin sensitivity after a single intravenous administration of a recombinant human tumor necrosis factor receptor: Fc fusion protein in obese insulin-resistant patients. J. Clin. Endocrinol. Metab. 85, 1316–1319 (2000).

    CAS  PubMed  Google Scholar 

  56. Kraegen, E. W., Cooney, G. J., Ye, J. M., Thompson, A. L. & Furler, S. M. The role of lipids in the pathogenesis of muscle insulin resistance and β cell failure in type II diabetes and obesity. Exp. Clin. Endocrinol. Diabetes. 109 (Suppl. 2), S189–S201 (2001).Article

    Article  CAS  PubMed  Google Scholar 

  57. Aguirre, V., Uchida, T., Yenush, L., Davis, R. & White, M. F. The c-Jun NH2-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser307. J. Biol. Chem. 275, 9047–9054 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. Hirosumi, J. et al. A central role for JNK in obesity and insulin resistance. Nature 420, 333–336 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Gao, Z. et al. Serine phosphorylation of insulin receptor substrate 1 by inhibitor κB kinase complex. J. Biol. Chem. 277, 48115–48121 (2002).

    Article  CAS  PubMed  Google Scholar 

  60. Griffin, M. E. et al. Free fatty acid-induced insulin resistance is associated with activation of protein kinase C θ and alterations in the insulin signaling cascade. Diabetes 48, 1270–1274 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Yu, C. et al. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle. J. Biol. Chem. 277, 50230–50236 (2002).

    Article  CAS  PubMed  Google Scholar 

  62. Kim, J. K. et al. PKC-θ knockout mice are protected from fat-induced insulin resistance. J. Clin. Invest. 114, 823–827 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Dresner, A. et al. Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity. J. Clin. Invest. 103, 253–259 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yuan, M. et al. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkβ. Science 293, 1673–1677 (2001).

    Article  CAS  PubMed  Google Scholar 

  65. Hundal, R. S. et al. Mechanism by which high-dose aspirin improves glucose metabolism in type 2 diabetes. J. Clin. Invest. 109, 1321–1326 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Um, S. H. et al. Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature 431, 200–205 (2004).

    Article  CAS  PubMed  Google Scholar 

  67. Senn, J. J. Toll-like receptor-2 is essential for the development of palmitate-induced insulin resistance in myotubes. J. Biol. Chem. 281, 26865–26875 (2006).

    Article  CAS  PubMed  Google Scholar 

  68. Song, M. J., Kim, K. H., Yoon, J. M. & Kim, J. B. Activation of Toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem. Biophys. Res. Commun. 346, 739–745 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Suganami, T. et al. Role of the Toll-like receptor 4/NF-κB pathway in saturated fatty acid-induced inflammatory changes in the interaction between adipocytes and macrophages. Arterioscler. Thromb. Vasc. Biol. 27, 84–91 (2007).

    Article  CAS  PubMed  Google Scholar 

  70. Shi, H. et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin. Invest. 116, 3015–3025 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Tsukumo, D. M. et al. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes 56, 1986–1998 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Hannun, Y. A. The sphingomyelin cycle and the second messenger function of ceramide. J. Biol. Chem. 269, 3125–3128 (1994).

    Article  CAS  PubMed  Google Scholar 

  73. Shimabukuro, M. et al. Lipoapoptosis in β-cells of obese prediabetic fa/fa rats. Role of serine palmitoyltransferase overexpression. J. Biol. Chem. 273, 32487–32490 (1998).

    Article  CAS  PubMed  Google Scholar 

  74. Summers, S. A., Garza, L. A., Zhou, H. & Birnbaum, M. J. Regulation of insulin-stimulated glucose transporter GLUT4 translocation and Akt kinase activity by ceramide. Mol. Cell. Biol. 18, 5457–5464 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Powell, D. J., Turban, S., Gray, A., Hajduch, E. & Hundal, H. S. Intracellular ceramide synthesis and protein kinase Cζ activation play an essential role in palmitate-induced insulin resistance in rat L6 skeletal muscle cells. Biochem. J. 382, 619–629 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Summers, S. A. & Nelson, D. H. A role for sphingolipids in producing the common features of type 2 diabetes, metabolic syndrome X, and Cushing's syndrome. Diabetes 54, 591–602 (2005).

    Article  CAS  PubMed  Google Scholar 

  77. Holland, W. L. et al. Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance. Cell Metab. 5, 167–179 (2007).

    Article  CAS  PubMed  Google Scholar 

  78. Turinsky, J., O'Sullivan, D. M. & Bayly, B. P. 1, 2-Diacylglycerol and ceramide levels in insulin-resistant tissues of the rat in vivo. J. Biol. Chem. 265, 16880–16885 (1990).

    Article  CAS  PubMed  Google Scholar 

  79. Adams, J. M. et al. Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes 53, 25–31 (2004).

    Article  CAS  PubMed  Google Scholar 

  80. Tamori, Y., Masugi, J., Nishino, N. & Kasuga, M. Role of peroxisome proliferator-activated receptor-g in maintenance of the characteristics of mature 3T3-L1 adipocytes. Diabetes 51, 2045–2055 (2002).

    Article  CAS  PubMed  Google Scholar 

  81. Imai, T. et al. Peroxisome proliferator-activated receptor γ is required in mature white and brown adipocytes for their survival in the mouse. Proc. Natl Acad. Sci. USA 101, 4543–4547 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Zhang, B. et al. Negative regulation of peroxisome proliferator-activated receptor-γ gene expression contributes to the antiadipogenic effects of tumor necrosis factor-α. Mol. Endocrinol. 10, 1457–1466 (1996).

    CAS  PubMed  Google Scholar 

  83. Stephens, J. M., Lee, J. & Pilch, P. F. Tumor necrosis factor-α-induced insulin resistance in 3T3-L1 adipocytes is accompanied by a loss of insulin receptor substrate-1 and GLUT4 expression without a loss of insulin receptor-mediated signal transduction. J. Biol. Chem. 272, 971–976 (1997).

    Article  CAS  PubMed  Google Scholar 

  84. Ruan, H., Hacohen, N., Golub, T. R., Van Parijs, L. & Lodish, H. F. Tumor necrosis factor-α suppresses adipocyte-specific genes and activates expression of preadipocyte genes in 3T3-L1 adipocytes: nuclear factor-κB activation by TNF-α is obligatory. Diabetes 51, 1319–1336 (2002).

    Article  CAS  PubMed  Google Scholar 

  85. Puri, V., Virbasius, J. V., Guilherme, A. & Czech, M. P. RNAi screens reveal novel metabolic regulators: RIP140, MAP4k4 and the lipid droplet associated fat specific protein (FSP) 27. Acta Physiol. (Oxf.) 192, 103–115 (2008).

    Article  CAS  Google Scholar 

  86. Floyd, Z. E. & Stephens, J. M. Interferon-γ-mediated activation and ubiquitin-proteasome-dependent degradation of PPARγ in adipocytes. J. Biol. Chem. 277, 4062–4068 (2002).

    Article  CAS  PubMed  Google Scholar 

  87. Medina, E. A. et al. Tumour necrosis factor-α decreases Akt protein levels in 3T3-L1 adipocytes via the caspase-dependent ubiquitination of Akt. Endocrinology 146, 2726–2735 (2005).

    Article  CAS  PubMed  Google Scholar 

  88. Diradourian, C., Girard, J. & Pegorier, J. P. Phosphorylation of PPARs: from molecular characterization to physiological relevance. Biochimie 87, 33–38 (2005).

    Article  CAS  PubMed  Google Scholar 

  89. Hauser, S. et al. Degradation of the peroxisome proliferator-activated receptor γ is linked to ligand-dependent activation. J. Biol. Chem. 275, 18527–18533 (2000).

    Article  CAS  PubMed  Google Scholar 

  90. Shao, D. et al. Interdomain communication regulating ligand binding by PPAR-γ. Nature 396, 377–380 (1998).

    Article  CAS  PubMed  Google Scholar 

  91. Hu, E., Kim, J. B., Sarraf, P. & Spiegelman, B. M. Inhibition of adipogenesis through MAP kinase-mediated phosphorylation of PPARγ. Science 274, 2100–2103 (1996).

    Article  CAS  PubMed  Google Scholar 

  92. Camp, H. S. & Tafuri, S. R. Regulation of peroxisome proliferator-activated receptor γ activity by mitogen-activated protein kinase. J. Biol. Chem. 272, 10811–10816 (1997).

    Article  CAS  PubMed  Google Scholar 

  93. Ristow, M., Muller-Wieland, D., Pfeiffer, A., Krone, W. & Kahn, C. R. Obesity associated with a mutation in a genetic regulator of adipocyte differentiation. N. Engl. J. Med. 339, 953–959 (1998).

    Article  CAS  PubMed  Google Scholar 

  94. Rangwala, S. M. et al. Genetic modulation of PPARγ phosphorylation regulates insulin sensitivity. Dev. Cell 5, 657–663 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Feige, J. N. & Auwerx, J. Transcriptional coregulators in the control of energy homeostasis. Trends Cell Biol. 17, 292–301 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Leonardsson, G. et al. Nuclear receptor corepressor RIP140 regulates fat accumulation. Proc. Natl Acad. Sci. USA 101, 8437–8442 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Powelka, A. M. et al. Suppression of oxidative metabolism and mitochondrial biogenesis by the transcriptional corepressor RIP140 in mouse adipocytes. J. Clin. Invest. 116, 125–136 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Wu, Z. et al. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92, 829–839 (1998).

    Article  PubMed  Google Scholar 

  99. Finck, B. N. & Kelly, D. P. PGC-1 coactivators: inducible regulators of energy metabolism in health and disease. J. Clin. Invest. 116, 615–622 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Large, V., Peroni, O., Letexier, D., Ray, H. & Beylot, M. Metabolism of lipids in human white adipocyte. Diabetes Metab. 30, 294–309 (2004).

    Article  CAS  PubMed  Google Scholar 

  101. Arner, P. Human fat cell lipolysis: biochemistry, regulation and clinical role. Best Pract. Res. Clin. Endocrinol Metab. 19, 471–482 (2005).

    Article  CAS  PubMed  Google Scholar 

  102. Degerman, E., Resjo, S., Landstrom, T. R. & Manganiello, V. Methods to study phosphorylation and activation of the hormone-sensitive adipocyte phosphodiesterase type 3B in rat adipocytes. Methods Mol. Biol. 155, 167–180 (2001).

    CAS  PubMed  Google Scholar 

  103. Granneman, J. G. & Moore, H. P. Location, location: protein trafficking and lipolysis in adipocytes. Trends Endocrinol. Metab. 19, 3–9 (2008).

    Article  CAS  PubMed  Google Scholar 

  104. Zimmermann, R. et al. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science 306, 1383–1386 (2004).

    Article  CAS  PubMed  Google Scholar 

  105. Ryden, M. et al. Comparative studies of the role of hormone-sensitive lipase and adipose triglyceride lipase in human fat cell lipolysis. Am. J. Physiol. Endocrinol. Metab. 292, E1847–E1855 (2007).

    Article  CAS  PubMed  Google Scholar 

  106. Brasaemle, D. L. Thematic review series: adipocyte biology. The perilipin family of structural lipid droplet proteins: stabilization of lipid droplets and control of lipolysis. J. Lipid Res. 48, 2547–2559 (2007).

    Article  CAS  PubMed  Google Scholar 

  107. Ducharme, N. A. & Bickel, P. E. Minireview: lipid droplets in lipogenesis and lipolysis. Endocrinology 149, 942–949 (2008). Summarizes recent developments in the field of lipid-droplet structure and function.

    Article  CAS  PubMed  Google Scholar 

  108. Wellen, K. E. & Hotamisligil, G. S. Obesity-induced inflammatory changes in adipose tissue. J. Clin. Invest. 112, 1785–1788 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Langin, D. & Arner, P. Importance of TNFα and neutral lipases in human adipose tissue lipolysis. Trends Endocrinol. Metab. 17, 314–320 (2006).

    Article  CAS  PubMed  Google Scholar 

  110. Hotamisligil, G. S., Arner, P., Caro, J. F., Atkinson, R. L. & Spiegelman, B. M. Increased adipose tissue expression of tumor necrosis factor-α in human obesity and insulin resistance. J. Clin. Invest. 95, 2409–2415 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Engfeldt, P. & Arner, P. Lipolysis in human adipocytes, effects of cell size, age and of regional differences. Horm. Metab. Res. Suppl. 19, 26–29 (1988).

    CAS  PubMed  Google Scholar 

  112. 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 

  113. Tontonoz, P., Hu, E., Devine, J., Beale, E. G. & Spiegelman, B. M. PPAR γ 2 regulates adipose expression of the phosphoenolpyruvate carboxykinase gene. Mol. Cell. Biol. 15, 351–357 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Dalen, K. T. et al. Adipose tissue expression of the lipid droplet-associating proteins S3-12 and perilipin is controlled by peroxisome proliferator-activated receptor-γ. Diabetes 53, 1243–1252 (2004).

    Article  CAS  PubMed  Google Scholar 

  115. Wolins, N. E. et al. OXPAT/PAT-1 is a PPAR-induced lipid droplet protein that promotes fatty acid utilization. Diabetes 55, 3418–3428 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Kadereit, B. et al. Evolutionarily conserved gene family important for fat storage. Proc. Natl Acad. Sci. USA 105, 94–99 (2008).

    Article  CAS  PubMed  Google Scholar 

  117. Puri, V. et al. Fat-specific protein 27, a novel lipid droplet protein that enhances triglyceride storage. J. Biol. Chem. 282, 34213–34218 (2007).

    Article  CAS  PubMed  Google Scholar 

  118. Liang, L., Zhao, M., Xu, Z., Yokoyama, K. K. & Li, T. Molecular cloning and characterization of CIDE-3, a novel member of the cell-death-inducing DNA-fragmentation-factor (DFF45)-like effector family. Biochem. J. 370, 195–203 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Inohara, N., Koseki, T., Chen, S., Wu, X. & Nunez, G. CIDE, a novel family of cell death activators with homology to the 45 kDa subunit of the DNA fragmentation factor. EMBO J. 17, 2526–2533 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Puri, V. et al. Cidea: a novel lipid droplet protein associated with insulin sensitivity in humans. Proc. Natl Acad. Sci. USA (in the press).

  121. Brasaemle, D. L., Dolios, G., Shapiro, L. & Wang, R. Proteomic analysis of proteins associated with lipid droplets of basal and lipolytically stimulated 3T3-L1 adipocytes. J. Biol. Chem. 279, 46835–46842 (2004).

    Article  CAS  PubMed  Google Scholar 

  122. Martinez-Botas, J. et al. Absence of perilipin results in leanness and reverses obesity in Leprdb/db mice. Nature Genet. 26, 474–479 (2000).

    Article  CAS  PubMed  Google Scholar 

  123. Nordstrom, E. A. et al. A human-specific role of cell death-inducing DFFA (DNA fragmentation factor-α)-like effector A (CIDEA) in adipocyte lipolysis and obesity. Diabetes 54, 1726–1734 (2005).

    Article  PubMed  Google Scholar 

  124. Wolins, N. E., Brasaemle, D. L. & Bickel, P. E. A proposed model of fat packaging by exchangeable lipid droplet proteins. FEBS Lett. 580, 5484–5491 (2006).

    Article  CAS  PubMed  Google Scholar 

  125. Perugini, R. A. et al. Metabolic characterization of nondiabetic severely obese patients undergoing Roux-en-Y gastric bypass: preoperative classification predicts the effects of gastric bypass on insulin-glucose homeostasis. J. Gastrointest. Surg. 11, 1083–1090 (2007).

    Article  PubMed  Google Scholar 

  126. Kim, H. J. et al. Depot-specific regulation of perilipin by rosiglitazone in a diabetic animal model. Metabolism 56, 676–685 (2007).

    Article  CAS  PubMed  Google Scholar 

  127. Smith, R. E. & Horwitz, B. A. Brown fat and thermogenesis. Physiol. Rev. 49, 330–425 (1969).

    Article  CAS  PubMed  Google Scholar 

  128. Nedergaard, J., Bengtsson, T. & Cannon, B. Unexpected evidence for active brown adipose tissue in adult humans. Am. J. Physiol. Endocrinol. Metab. 293, E444–E452 (2007).

    Article  CAS  PubMed  Google Scholar 

  129. Katagiri, H., Yamada, T. & Oka, Y. Adiposity and cardiovascular disorders: disturbance of the regulatory system consisting of humoral and neuronal signals. Circ. Res. 101, 27–39 (2007).

    Article  CAS  PubMed  Google Scholar 

  130. Bartness, T. J. & Song, C. K. Thematic review series: adipocyte biology. Sympathetic and sensory innervation of white adipose tissue. J. Lipid Res. 48, 1655–1672 (2007).

    Article  CAS  PubMed  Google Scholar 

  131. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nature Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  132. Patti, M. E. et al. Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc. Natl Acad. Sci. USA 100, 8466–8471 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Petersen, K. F. et al. Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science 300, 1140–1142 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Petersen, K. F., Dufour, S., Befroy, D., Garcia, R. & Shulman, G. I. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N. Engl. J. Med. 350, 664–671 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Richardson, D. K. et al. Lipid infusion decreases the expression of nuclear encoded mitochondrial genes and increases the expression of extracellular matrix genes in human skeletal muscle. J. Biol. Chem. 280, 10290–10297 (2005).

    Article  CAS  PubMed  Google Scholar 

  136. Sparks, L. M. et al. A high-fat diet coordinately downregulates genes required for mitochondrial oxidative phosphorylation in skeletal muscle. Diabetes 54, 1926–1933 (2005).

    Article  CAS  PubMed  Google Scholar 

  137. Petersen, K. F. et al. Reversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hyperglycemia by moderate weight reduction in patients with type 2 diabetes. Diabetes 54, 603–608 (2005).

    Article  CAS  PubMed  Google Scholar 

  138. Reznick, R. M. & Shulman, G. I. The role of AMP-activated protein kinase in mitochondrial biogenesis. J. Physiol. 574, 33–39 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Morino, K., Petersen, K. F. & Shulman, G. I. Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction. Diabetes 55 (Suppl. 2), S9–S15 (2006).Article

    Article  CAS  PubMed  Google Scholar 

  140. Turner, N. et al. Excess lipid availability increases mitochondrial fatty acid oxidative capacity in muscle: evidence against a role for reduced fatty acid oxidation in lipid-induced insulin resistance in rodents. Diabetes 56, 2085–2092 (2007).

    Article  CAS  PubMed  Google Scholar 

  141. 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 

Download references

Acknowledgements

We thank the members of our laboratory group for excellent discussions on the issues addressed here. We acknowledge funding for our studies covered here by the National Institutes of Health (grants DK030648 and DK030898), including the University of Massachusetts Medical School Diabetes and Endocrinology Research Center (DK32520).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael P. Czech.

Related links

Related links

FURTHER INFORMATION

Michael P. Czech's homepage

Glossary

White adipose tissue

The predominant fat storage tissue in animals, consisting mostly of adipocytes but also other cell types such as mast cells and macrophages. It is distributed in a number of subcutaneous and visceral depots.

Free fatty acid

A carboxylic acid with aliphatic chains of 4–28 carbons, which can be esterified with glycerol to form triglycerides, the main stored form of lipid.

Acipimox

A nicotinic-acid analogue that reduces plasma and intracellular fatty acid levels by suppressing lipolysis.

Adipokine

A cytokine or hormone that is secreted by adipose tissue.

Lipodystrophy

Abnormality of the adipose tissue that is associated with total or partial loss of body fat. This might have a genetic origin in humans or occur as a result of other diseases.

MAP4K4

(Mitogen-activated protein kinase kinase kinase kinase-4). A mammalian Ser/Thr protein kinase related to Saccharomyces cerevisiae Sterile-20 (STE20).

Omental adipose tissue

The fat depot found within the peritoneum, in close association with the stomach and other internal organs.

Diacylglycerol

A molecule that consists of two fatty acid chains esterified with glycerol, produced by the cleavage of membrane phosphatidylinositol 4,5-bisphosphate. Diacylglycerol functions as a signalling molecule by activating protein kinase C.

Salicylates

A group of derivatives of salicylic acid, including aspirin and acetylsalicylic acid, which are widely used as analgesics and anti-inflammatory medications.

Toll-like receptors

Cell-surface receptors that recognize a wide variety of molecules, primarily markers of foreign organisms including bacteria. Main functions include the activation of the innate immune response in infection.

Sphingolipids

Lipids that consist of the aliphatic alcohol sphingosine linked to a fatty acid chain and a variety of head groups. Sphingolipids include ceramides, sphingomyelin and glycosphingolipids.

Glucocorticoids

Steroid hormones, including cortisol, that are produced in the adrenal gland. These hormones have potent effects on energy metabolism in the liver, fat and elsewhere.

C/EBPα

(CCAAT/enhancer-binding protein-α). A transcription factor that has a key role in the differentiation of adipocytes.

Adrenoreceptors

A class of G-protein-coupled receptors activated by catecholamines. Adrenoreceptors in white fat stimulate lipolysis in response to catecholamines.

Catecholamines

A group of amine hormones including adrenaline and noradrenaline. These are produced by the adrenal gland in response to starvation and other stresses.

Perilipin

A main protein component of adipocyte lipid droplets, which surrounds the lipids and participates in the regulation of lipolysis.

Rosiglitazone

An antidiabetic drug, one of several thiazolidinediones (TZDs) used therapeutically. Many TZDs are known to be high-affinity ligand activators of PPARγ.

HOMA-IR

(Homeostatic model assessment of insulin resistance). An estimation of the degree of insulin resistance that is calculated from clinical measurement of fasting blood glucose and insulin levels.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Guilherme, A., Virbasius, J., Puri, V. et al. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat Rev Mol Cell Biol 9, 367–377 (2008). https://doi.org/10.1038/nrm2391

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm2391

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