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:

Targeting inflammation in the treatment of type 2 diabetes: time to start

Key Points

  • Inflammation has an important role in the pathogenesis of type 2 diabetes and associated complications.

  • Clinical studies have demonstrated that interleukin-1 (IL-1) antagonists, salsalate and tumour necrosis factor (TNF) antagonists improve glucose metabolism.

  • Anti-inflammatory drugs may have disease-modifying and long-lasting effects.

  • Future genetic and biomarker studies may profile responders to a specific anti-inflammatory treatment.

  • Combining or sequentially using multiple anti-inflammatory drugs may provide a tailored solution for treating patients with type 2 diabetes.

Abstract

The role of inflammation in the pathogenesis of type 2 diabetes and associated complications is now well established. Several conditions that are driven by inflammatory processes are also associated with diabetes, including rheumatoid arthritis, gout, psoriasis and Crohn's disease, and various anti-inflammatory drugs have been approved or are in late stages of development for the treatment of these conditions. This Review discusses the rationale for the use of some of these anti-inflammatory treatments in patients with diabetes and what we could expect from their use. Future immunomodulatory treatments may not target a specific disease, but could instead act on a dysfunctional pathway that causes several conditions associated with the metabolic syndrome.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The development of type 2 diabetes.
Figure 2: Islet inflammation in type 2 diabetes.
Figure 3: Storage of excessive nutrients in adipose tissues leads to an inflammatory response and insulin resistance.

Similar content being viewed by others

References

  1. Cerasi, E. Insulin deficiency and insulin resistance in the pathogenesis of NIDDM: is a divorce possible? Diabetologia 38, 992–997 (1995).

    Article  CAS  PubMed  Google Scholar 

  2. Kahn, S. E., Hull, R. L. & Utzschneider, K. M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444, 840–846 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Bonner-Weir, S. Islet growth and development in the adult. J. Mol. Endocrinol. 24, 297–302 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Kahn, B. B. Type 2 diabetes: when insulin secretion fails to compensate for insulin resistance. Cell 92, 593–596 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Robertson, R. P., Harmon, J., Tran, P. O. & Poitout, V. β-cell glucose toxicity, lipotoxicity, and chronic oxidative stress in type 2 diabetes. Diabetes 53 (Suppl. 1), 119–124 (2004).

    Article  Google Scholar 

  6. Weir, G. C. & Bonner-Weir, S. Five stages of evolving β-cell dysfunction during progression to diabetes. Diabetes 53 (Suppl. 3), 16–21 (2004).

    Article  Google Scholar 

  7. Prentki, M. & Nolan, C. J. Islet β cell failure in type 2 diabetes. J. Clin. Invest. 116, 1802–1812 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hull, R. L., Westermark, G. T., Westermark, P. & Kahn, S. E. Islet amyloid: a critical entity in the pathogenesis of type 2 diabetes. J. Clin. Endocrinol. Metab. 89, 3629–3643 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Harding, H. P. & Ron, D. Endoplasmic reticulum stress and the development of diabetes: a review. Diabetes 51 (Suppl. 3), 455–461 (2002).

    Article  Google Scholar 

  10. Hotamisligil, G. S. & Erbay, E. Nutrient sensing and inflammation in metabolic diseases. Nature Rev. Immunol. 8, 923–934 (2008).

    Article  CAS  Google Scholar 

  11. Donath, M. Y., Storling, J., Maedler, K. & Mandrup-Poulsen, T. Inflammatory mediators and islet β-cell failure: a link between type 1 and type 2 diabetes. J. Mol. Med. 81, 455–470 (2003).

    Article  CAS  PubMed  Google Scholar 

  12. Ehses, J. A., Ellingsgaard, H., Boni-Schnetzler, M. & Donath, M. Y. Pancreatic islet inflammation in type 2 diabetes: from α and β cell compensation to dysfunction. Arch. Physiol. Biochem. 115, 240–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Westwell-Roper, C. Y., Ehses, J. A. & Verchere, C. B. Resident macrophages mediate islet amyloid polypeptide-induced islet IL-1β production and β cell dysfunction. Diabetes 63, 1698–1711 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Westwell-Roper, C. et al. IL-1 blockade attenuates islet amyloid polypeptide-induced proinflammatory cytokine release and pancreatic islet graft dysfunction. J. Immunol. 187, 2755–2765 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Masters, S. L. et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1β in type 2 diabetes. Nature Immunol. 11, 897–904 (2010).

    Article  CAS  Google Scholar 

  16. Jourdan, T. et al. Activation of the Nlrp3 inflammasome in infiltrating macrophages by endocannabinoids mediates β cell loss in type 2 diabetes. Nature Med. 19, 1132–1140 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Oslowski, C. M. et al. Thioredoxin-interacting protein mediates ER stress-induced β cell death through initiation of the inflammasome. Cell Metab. 16, 265–273 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lerner, A. G. et al. IRE1α induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell Metab. 16, 250–264 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Maedler, K. et al. Glucose-induced β cell production of IL-1β contributes to glucotoxicity in human pancreatic islets. J. Clin. Invest. 110, 851–860 (2002). This is the first description of the role of IL-1β in type 2 diabetes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhou, R., Tardivel, A., Thorens, B., Choi, I. & Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nature Immunol. 11, 136–140 (2010).

    Article  CAS  Google Scholar 

  21. Boni-Schnetzler, M. et al. Free fatty acids induce a proinflammatory response in islets via the abundantly expressed interleukin-1 receptor I. Endocrinology 150, 5218–5229 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Pal, D. et al. Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nature Med. 18, 1279–1285 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Maedler, K. et al. FLIP switches Fas-mediated glucose signaling in human pancreatic β cells from apoptosis to cell replication. Proc. Natl Acad. Sci. USA 99, 8236–8241 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ehses, J. A. et al. Increased number of islet-associated macrophages in type 2 diabetes. Diabetes 56, 2356–2370 (2007). This is the original description of macrophage infiltration in islets of patients with type 2 diabetes.

    Article  CAS  PubMed  Google Scholar 

  25. Richardson, S. J., Willcox, A., Bone, A. J., Foulis, A. K. & Morgan, N. G. Islet-associated macrophages in type 2 diabetes. Diabetologia 52, 1686–1688 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Butcher, M. J. et al. Association of proinflammatory cytokines and islet resident leucocytes with islet dysfunction in type 2 diabetes. Diabetologia 57, 491–501 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Boni-Schnetzler, M. et al. Increased interleukin (IL)-1β messenger ribonucleic acid expression in β-cells of individuals with type 2 diabetes and regulation of IL-1β in human islets by glucose and autostimulation. J. Clin. Endocrinol. Metab. 93, 4065–4074 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Di Marzo, V. et al. Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature 410, 822–825 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Osei-Hyiaman, D. et al. Endocannabinoid activation at hepatic CB1 receptors stimulates fatty acid synthesis and contributes to diet-induced obesity. J. Clin. Invest. 115, 1298–1305 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bendtzen, K. et al. Cytotoxicity of human pI 7 interleukin-1 for pancreatic islets of Langerhans. Science 232, 1545–1547 (1986).

    Article  CAS  PubMed  Google Scholar 

  31. Mandrup-Poulsen, T. et al. Affinity-purified human interleukin I is cytotoxic to isolated islets of Langerhans. Diabetologia 29, 63–67 (1986).

    Article  CAS  PubMed  Google Scholar 

  32. Hotamisligil, G. S. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell 140, 900–917 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Martinez, J., Verbist, K., Wang, R. & Green, D. R. The relationship between metabolism and the autophagy machinery during the innate immune response. Cell Metab. 17, 895–900 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Skurk, T., Alberti-Huber, C., Herder, C. & Hauner, H. Relationship between adipocyte size and adipokine expression and secretion. J. Clin. Endocrinol. Metab. 92, 1023–1033 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Stienstra, R. et al. The inflammasome-mediated caspase-1 activation controls adipocyte differentiation and insulin sensitivity. Cell. Metab. 12, 593–605 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Koenen, T. B. et al. Hyperglycemia activates caspase-1 and TXNIP-mediated IL-1β transcription in human adipose tissue. Diabetes 60, 517–524 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wen, H. et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nature Immunol. 12, 408–415 (2011).

    Article  CAS  Google Scholar 

  38. Cinti, S. et al. Adipocyte death defines macrophage localization and function in adipose tissue of obese mice and humans. J. Lipid Res. 46, 2347–2355 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. 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 

  40. 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). References 39 and 40 identify macrophages in adipose tissue and show that their numbers increase with obesity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Prieur, X. et al. Differential lipid partitioning between adipocytes and tissue macrophages modulates macrophage lipotoxicity and M2/M1 polarization in obese mice. Diabetes 60, 797–809 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ye, J. Emerging role of adipose tissue hypoxia in obesity and insulin resistance. Int. J. Obes. 33, 54–66 (2009).

    Article  CAS  Google Scholar 

  43. Cox, L. M. & Blaser, M. J. Pathways in microbe-induced obesity. Cell Metab. 17, 883–894 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ley, R. E. et al. Obesity alters gut microbial ecology. Proc. Natl Acad. Sci. USA 102, 11070–11075 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Turnbaugh, P. J. et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031 (2006).

    Article  PubMed  Google Scholar 

  46. Ley, R. E., Turnbaugh, P. J., Klein, S. & Gordon, J. I. Microbial ecology: human gut microbes associated with obesity. Nature 444, 1022–1023 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Cani, P. D. et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 1761–1772 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Cani, P. D. et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 57, 1470–1481 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Nguyen, M. T. et al. A subpopulation of macrophages infiltrates hypertrophic adipose tissue and is activated by free fatty acids via Toll-like receptors 2 and 4 and JNK-dependent pathways. J. Biol. Chem. 282, 35279–35292 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Vandanmagsar, B. et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nature Med. 17, 179–188 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Dinarello, C. A. Immunological and inflammatory functions of the interleukin-1 family. Annu. Rev. Immunol. 27, 519–550 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Hotamisligil, G. S., Shargill, N. S. & Spiegelman, B. M. Adipose expression of tumor necrosis factor-α: direct role in obesity-linked insulin resistance. Science 259, 87–91 (1993). This is the first description of the role of TNF in type 2 diabetes.

    Article  CAS  PubMed  Google Scholar 

  53. 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 

  54. 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 

  55. Dominguez, H. et al. Metabolic and vascular effects of tumor necrosis factor-α blockade with etanercept in obese patients with type 2 diabetes. J. Vasc. Res. 42, 517–525 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Bernstein, L. E., Berry, J., Kim, S., Canavan, B. & Grinspoon, S. K. Effects of etanercept in patients with the metabolic syndrome. Arch. Intern. Med. 166, 902–908 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Stanley, T. L. et al. TNF-α antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. J. Clin. Endocrinol. Metab. 96, E146–E150 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Yazdani-Biuki, B. et al. Improvement of insulin sensitivity in insulin resistant subjects during prolonged treatment with the anti-TNF-α antibody infliximab. Eur. J. Clin. Invest. 34, 641–642 (2004).

    Article  CAS  PubMed  Google Scholar 

  59. Kiortsis, D. N., Mavridis, A. K., Vasakos, S., Nikas, S. N. & Drosos, A. A. Effects of infliximab treatment on insulin resistance in patients with rheumatoid arthritis and ankylosing spondylitis. Ann. Rheum. Dis. 64, 765–766 (2005).

    Article  CAS  PubMed  Google Scholar 

  60. Yazdani-Biuki, B. et al. Relapse of diabetes after interruption of chronic administration of anti-tumor necrosis factor-α antibody infliximab: a case observation. Diabetes Care 29, 1712–1713 (2006).

    Article  PubMed  Google Scholar 

  61. Gonzalez-Gay, M. A. et al. Anti-tumor necrosis factor-α blockade improves insulin resistance in patients with rheumatoid arthritis. Clin. Exp. Rheumatol. 24, 83–86 (2006).

    CAS  PubMed  Google Scholar 

  62. Huvers, F. C., Popa, C., Netea, M. G., van den Hoogen, F. H. & Tack, C. J. Improved insulin sensitivity by anti-TNFα antibody treatment in patients with rheumatic diseases. Ann. Rheum. Dis. 66, 558–559 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Marra, M. et al. Effect of etanercept on insulin sensitivity in nine patients with psoriasis. Int. J. Immunopathol. Pharmacol. 20, 731–736 (2007).

    Article  CAS  PubMed  Google Scholar 

  64. Timper, K., Hruz, P., Beglinger, C. & Donath, M. Y. Infliximab in the treatment of Crohn disease and type 1 diabetes. Diabetes Care 36, e90–91 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Solomon, D. H. et al. Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA 305, 2525–2531 (2011).

    Article  CAS  PubMed  Google Scholar 

  66. Antohe, J. L. et al. Diabetes mellitus risk in rheumatoid arthritis: reduced incidence with anti-tumor necrosis factor α therapy. Arthritis Care Res. 64, 215–221 (2012).

    Article  CAS  Google Scholar 

  67. Larsen, C. M. et al. Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes. Diabetes Care 32, 1663–1668 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Larsen, C. M. et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N. Engl. J. Med. 356, 1517–1526 (2007). This is a proof-of-concept clinical study demonstrating the potential of immunomodulation in patients with type 2 diabetes.

    Article  CAS  PubMed  Google Scholar 

  69. van Asseldonk, E. J. et al. Treatment with anakinra improves disposition index but not insulin sensitivity in nondiabetic subjects with the metabolic syndrome: a randomized, double-blind, placebo-controlled study. J. Clin. Endocrinol. Metab. 96, 2119–2126 (2011).

    Article  CAS  PubMed  Google Scholar 

  70. van Asseldonk, E. J. P. et al. One week of treatment with the IL-1 receptor antagonist anakinra improves insulin sensitivity in patients with type 1 diabetes mellitus: results from a clinical trial. In: 48th Annual Meeting of the European Association for the Study of Diabetes [online], Abstract 560 (2012).

    Google Scholar 

  71. van Popper, P. C. M., Van Asseldonk, E. V. A., Netea, M. G. & Tack, C. J. The interleukin-1 receptor antagonist anakinra improves β cell function in subjects with impaired glucose tolerance. In: 49th Annual Meeting of the European Association for the Study of Diabetes [online], Abstract 739 (2013).

    Google Scholar 

  72. Cavelti-Weder, C. et al. Effects of gevokizumab on glycemia and inflammatory markers in type 2 diabetes. Diabetes Care 35, 1654–1662 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hensen, J., Howard, C. P., Walter, V. & Thuren, T. Impact of interleukin-1β antibody (canakinumab) on glycaemic indicators in patients with type 2 diabetes mellitus: results of secondary endpoints from a randomized, placebo-controlled trial. Diabetes Metab. 39, 524–531 (2013).

    Article  CAS  PubMed  Google Scholar 

  74. Rissanen, A. et al., Howard, C. P., Botha, J., Thuren, T. & for the Global Investigators. Effect of anti-IL-1β antibody (canakinumab) on insulin secretion rates in impaired glucose tolerance or type 2 diabetes: results of a randomized, placebo-controlled trial. Diabetes Obes. Metab. 14, 1088–1096 (2012).

    Article  CAS  PubMed  Google Scholar 

  75. Sloan-Lancaster, J. et al. Double-blind, randomized study evaluating the glycemic and anti-inflammatory effects of subcutaneous LY2189102, a neutralizing IL-1β antibody, in patients with type 2 diabetes. Diabetes Care 36, 2239–2246 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Yuan, M. et al. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkβ. Science 293, 1673–1677 (2001). This is the first description of the role of salsalate via inhibition of NF-κB in type 2 diabetes.

    Article  CAS  PubMed  Google Scholar 

  77. Fleischman, A., Shoelson, S. E., Bernier, R. & Goldfine, A. B. Salsalate improves glycemia and inflammatory parameters in obese young adults. Diabetes Care 31, 289–294 (2008).

    Article  CAS  PubMed  Google Scholar 

  78. Goldfine, A. B. et al. Use of salsalate to target inflammation in the treatment of insulin resistance and type 2 diabetes. Clin. Transl. Sci. 1, 36–43 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Koska, J. et al. The effect of salsalate on insulin action and glucose tolerance in obese non-diabetic patients: results of a randomised double-blind placebo-controlled study. Diabetologia 52, 385–393 (2009).

    Article  CAS  PubMed  Google Scholar 

  80. Goldfine, A. B. et al. The effects of salsalate on glycemic control in patients with type 2 diabetes: a randomized trial. Ann. Intern. Med. 152, 346–357 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Goldfine, A. B. et al. Salicylate (salsalate) in patients with type 2 diabetes: a randomized trial. Ann. Intern.Med. 159, 1–12 (2013). This Phase III clinical trial confirms that salsalate improves glycaemia in patients with type 2 diabetes.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Goldfine, A. B. et al. A randomised trial of salsalate for insulin resistance and cardiovascular risk factors in persons with abnormal glucose tolerance. Diabetologia 56, 714–723 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Faghihimani, E. et al. Salsalate improves glycemic control in patients with newly diagnosed type 2 diabetes. Acta Diabetol. 50, 537–543 (2013).

    Article  CAS  PubMed  Google Scholar 

  84. Ramos-Zavala, M. G. et al. Effect of diacerein on insulin secretion and metabolic control in drug-naive patients with type 2 diabetes: a randomized clinical trial. Diabetes Care 34, 1591–1594 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Freigang, S. et al. Fatty acid-induced mitochondrial uncoupling elicits inflammasome-independent IL-1α and sterile vascular inflammation in atherosclerosis. Nature Immunol. 14, 1045–1053 (2013).

    Article  CAS  Google Scholar 

  86. Timper, K. et al. IL-1α antagonism in type 2 diabetes. In: 95th Annual Meeting of the Endocrine Society [online], Abstract SUN-856 (2013).

    Google Scholar 

  87. 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 

  88. 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 

  89. Weisberg, S. P. et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J. Clin. Invest. 116, 115–124 (2006).

    Article  CAS  PubMed  Google Scholar 

  90. Hanefeld, M. et al. Orally-administered chemokine receptor CCR2 antagonist CCX140-B in type 2 diabetes: a pilot double-blind, randomized clinical trial. J. Diabetes Metab. http://dx.doi.org/10.4172/2155-6156.1000225 (2012).

  91. Pedersen, B. K. & Febbraio, M. A. Muscle as an endocrine organ: focus on muscle-derived interleukin-6. Physiol. Rev. 88, 1379–1406 (2008).

    Article  CAS  PubMed  Google Scholar 

  92. Pedersen, B. K. & Febbraio, M. A. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nature Rev. Endocrinol. 8, 457–465 (2012).

    Article  CAS  Google Scholar 

  93. Carey, A. L. et al. Interleukin-6 increases insulin-stimulated glucose disposal in humans and glucose uptake and fatty acid oxidation in vitro via AMP-activated protein kinase. Diabetes 55, 2688–2697 (2006).

    Article  CAS  PubMed  Google Scholar 

  94. Jansson, J. O. & Wallenius, V. Point-counterpoint: Interleukin-6 does/does not have a beneficial role in insulin sensitivity and glucose homeostasis. J. Appl. Physiol. 102, 821; author reply 825 (2007).

    PubMed  Google Scholar 

  95. Mooney, R. A. Counterpoint: interleukin-6 does not have a beneficial role in insulin sensitivity and glucose homeostasis. J. Appl. Physiol. 102, 816–818; discussion 818–819 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Pedersen, B. K. & Febbraio, M. A. Point: interleukin-6 does have a beneficial role in insulin sensitivity and glucose homeostasis. J. Appl. Physiol. 102, 814–816 (2007).

    Article  CAS  PubMed  Google Scholar 

  97. Sabio, G. et al. A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science 322, 1539–1543 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Weigert, C., Lehmann, R. & Schleicher, E. D. Point-counterpoint: interleukin-6 does/does not have a beneficial role in insulin sensitivity and glucose homeostasis. J. Appl. Physiol. 102, 820–821; author reply 825 (2007).

    Article  PubMed  Google Scholar 

  99. Wunderlich, F. T. et al. Interleukin-6 signaling in liver-parenchymal cells suppresses hepatic inflammation and improves systemic insulin action. Cell Metab. 12, 237–249 (2010).

    Article  CAS  PubMed  Google Scholar 

  100. Ellingsgaard, H. et al. Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and α cells. Nature Med. 17, 1481–1489 (2011).

    Article  CAS  PubMed  Google Scholar 

  101. Lazar, M. A. How obesity causes diabetes: not a tall tale. Science 307, 373–375 (2005).

    Article  CAS  PubMed  Google Scholar 

  102. Rose-John, S. IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6. Int. J. Biol. Sci. 8, 1237–1247 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Imai, Y. et al. Interaction between cytokines and inflammatory cells in islet dysfunction, insulin resistance and vascular disease. Diabetes Obes. Metab. 15 (Suppl. 3), 117–129 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Haigis, M. C. & Sinclair, D. A. Mammalian sirtuins: biological insights and disease relevance. Annu. Rev. Pathol. 5, 253–295 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Misawa, T. et al. Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nature Immunol. 14, 454–460 (2013).

    Article  CAS  Google Scholar 

  106. Donath, M. Y. When metabolism met immunology. Nature Immunol. 14, 421–422 (2013).

    Article  CAS  Google Scholar 

  107. Biason-Lauber, A. et al. Identification of a SIRT1 mutation in a family with type 1 diabetes. Cell Metab. 17, 448–455 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Yoshino, J., Mills, K. F., Yoon, M. J. & Imai, S. Nicotinamide mononucleotide, a key NAD+ intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab. 14, 528–536 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Canto, C. et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 458, 1056–1060 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Canto, C. et al. The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 15, 838–847 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Altmeyer, M. & Hottiger, M. O. Poly(ADP-ribose) polymerase 1 at the crossroad of metabolic stress and inflammation in aging. Aging 1, 458–469 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  112. Isoda, K. et al. Metformin inhibits proinflammatory responses and nuclear factor-κB in human vascular wall cells. Arterioscler. Thromb. Vasc. Biol. 26, 611–617 (2006).

    Article  CAS  PubMed  Google Scholar 

  113. Lee, H. M. et al. Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes 62, 194–204 (2013).

    Article  CAS  PubMed  Google Scholar 

  114. Ricote, M., Li, A. C., Willson, T. M., Kelly, C. J. & Glass, C. K. The peroxisome proliferator-activated receptor-γ is a negative regulator of macrophage activation. Nature 391, 79–82 (1998).

    Article  CAS  PubMed  Google Scholar 

  115. Jiang, C., Ting, A. T. & Seed, B. PPAR-γ agonists inhibit production of monocyte inflammatory cytokines. Nature 391, 82–86 (1998).

    Article  CAS  PubMed  Google Scholar 

  116. Diaz-Delfin, J., Morales, M. & Caelles, C. Hypoglycemic action of thiazolidinediones/peroxisome proliferator-activated receptor γ by inhibition of the c-Jun NH2-terminal kinase pathway. Diabetes 56, 1865–1871 (2007).

    Article  CAS  PubMed  Google Scholar 

  117. Vallerie, S. N. & Hotamisligil, G. S. The role of JNK proteins in metabolism. Sci. Transl. Med. 2, 60rv65 (2010).

    Article  CAS  Google Scholar 

  118. Ferdaoussi, M. et al. Exendin-4 protects β-cells from interleukin-1β-induced apoptosis by interfering with the c-Jun NH2-terminal kinase pathway. Diabetes 57, 1205–1215 (2008).

    Article  CAS  PubMed  Google Scholar 

  119. Pugazhenthi, U., Velmurugan, K., Tran, A., Mahaffey, G. & Pugazhenthi, S. Anti-inflammatory action of exendin-4 in human islets is enhanced by phosphodiesterase inhibitors: potential therapeutic benefits in diabetic patients. Diabetologia 53, 2357–2368 (2010).

    Article  CAS  PubMed  Google Scholar 

  120. Omar, B. A. et al. Enhanced β cell function and anti-inflammatory effect after chronic treatment with the dipeptidyl peptidase-4 inhibitor vildagliptin in an advanced-aged diet-induced obesity mouse model. Diabetologia 56, 1752–1760 (2013).

    Article  CAS  PubMed  Google Scholar 

  121. Chaudhuri, A. et al. Exenatide exerts a potent antiinflammatory effect. J. Clin. Endocrinol. Metab. 97, 198–207 (2012).

    Article  CAS  PubMed  Google Scholar 

  122. Makdissi, A. et al. Sitagliptin exerts an antinflammatory action. J. Clin. Endocrinol. Metab. 97, 3333–3341 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Feldman, M., Jialal, I., Devaraj, S. & Cryer, B. Effects of low-dose aspirin on serum C-reactive protein and thromboxane B2 concentrations: a placebo-controlled study using a highly sensitive C-reactive protein assay. J. Am. Coll. Cardiol. 37, 2036–2041 (2001).

    Article  CAS  PubMed  Google Scholar 

  124. Ridker, P. M., Rifai, N., Pfeffer, M. A., Sacks, F. & Braunwald, E. Long-term effects of pravastatin on plasma concentration of C-reactive protein. The Cholesterol and Recurrent Events (CARE) Investigators. Circulation 100, 230–235 (1999).

    Article  CAS  PubMed  Google Scholar 

  125. Ridker, P. M. et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N. Engl. J. Med. 359, 2195–2207 (2008).

    Article  CAS  PubMed  Google Scholar 

  126. Jialal, I., Miguelino, E., Griffen, S. C. & Devaraj, S. Concomitant reduction of low-density lipoprotein-cholesterol and biomarkers of inflammation with low-dose simvastatin therapy in patients with type 1 diabetes. J. Clin. Endocrinol. Metab. 92, 3136–3140 (2007).

    Article  CAS  PubMed  Google Scholar 

  127. Montero, M. T. et al. Hydroxymethylglutaryl-coenzyme A reductase inhibition stimulates caspase-1 activity and Th1-cytokine release in peripheral blood mononuclear cells. Atherosclerosis 153, 303–313 (2000).

    Article  CAS  PubMed  Google Scholar 

  128. Coward, W. R., Marei, A., Yang, A., Vasa-Nicotera, M. M. & Chow, S. C. Statin-induced proinflammatory response in mitogen-activated peripheral blood mononuclear cells through the activation of caspase-1 and IL-18 secretion in monocytes. J. Immunol. 176, 5284–5292 (2006).

    Article  CAS  PubMed  Google Scholar 

  129. Kuijk, L. M. et al. HMG-CoA reductase inhibition induces IL-1β release through Rac1/PI3K/PKB-dependent caspase-1 activation. Blood 112, 3563–3573 (2008).

    Article  CAS  PubMed  Google Scholar 

  130. Kuijk, L. M. et al. Statin synergizes with LPS to induce IL-1β release by THP-1 cells through activation of caspase-1. Mol. Immunol. 45, 2158–2165 (2008).

    Article  CAS  PubMed  Google Scholar 

  131. Mandey, S. H., Kuijk, L. M., Frenkel, J. & Waterham, H. R. A role for geranylgeranylation in interleukin-1β secretion. Arthritis Rheum. 54, 3690–3695 (2006).

    Article  CAS  PubMed  Google Scholar 

  132. Liao, Y. H. et al. HMG-CoA reductase inhibitors activate caspase-1 in human monocytes depending on ATP release and P2X7 activation. J. Leukoc. Biol. 93, 289–299 (2013).

    Article  CAS  PubMed  Google Scholar 

  133. Sattar, N. et al. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 375, 735–742 (2010).

    Article  CAS  PubMed  Google Scholar 

  134. Jandeleit-Dahm, K. A., Tikellis, C., Reid, C. M., Johnston, C. I. & Cooper, M. E. Why blockade of the renin-angiotensin system reduces the incidence of new-onset diabetes. J. Hypertens. 23, 463–473 (2005).

    Article  CAS  PubMed  Google Scholar 

  135. van der Zijl, N. J., Moors, C. C., Goossens, G. H., Blaak, E. E. & Diamant, M. Does interference with the renin-angiotensin system protect against diabetes? Evidence and mechanisms. Diabetes Obes. Metab. 14, 586–595 (2012).

    Article  CAS  PubMed  Google Scholar 

  136. Fischer, E. et al. Aldosterone excess impairs first phase insulin secretion in primary aldosteronism. J. Clin. Endocrinol. Metab. 98, 2513–2520 (2013).

    Article  CAS  PubMed  Google Scholar 

  137. Fliser, D. et al. Antiinflammatory effects of angiotensin II subtype 1 receptor blockade in hypertensive patients with microinflammation. Circulation 110, 1103–1107 (2004).

    Article  CAS  PubMed  Google Scholar 

  138. Manabe, S., Okura, T., Watanabe, S., Fukuoka, T. & Higaki, J. Effects of angiotensin II receptor blockade with valsartan on pro-inflammatory cytokines in patients with essential hypertension. J. Cardiovasc. Pharmacol. 46, 735–739 (2005).

    Article  CAS  PubMed  Google Scholar 

  139. Pavlatou, M. G. et al. Angiotensin blockade in diabetic patients decreases insulin resistance-associated low-grade inflammation. Eur. J. Clin. Invest. 41, 652–658 (2011).

    Article  CAS  PubMed  Google Scholar 

  140. Garcia, G. E. ANG II receptor antagonists as modulators of macrophages polarization. Am. J. Physiol. Renal Physiol. 298, F868–F869 (2010).

    Article  CAS  PubMed  Google Scholar 

  141. Fujisaka, S. et al. Telmisartan improves insulin resistance and modulates adipose tissue macrophage polarization in high-fat-fed mice. Endocrinology 152, 1789–1799 (2011).

    Article  CAS  PubMed  Google Scholar 

  142. Gallwitz, B. et al. 2-year efficacy and safety of linagliptin compared with glimepiride in patients with type 2 diabetes inadequately controlled on metformin: a randomised, double-blind, non-inferiority trial. Lancet 380, 475–483 (2012).

    Article  CAS  PubMed  Google Scholar 

  143. Scirica, B. M. et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N. Engl. J. Med. 369, 1317–1326 (2013).

    Article  CAS  PubMed  Google Scholar 

  144. Mirza, R. E., Fang, M. M., Ennis, W. J. & Koh, T. J. Blocking interleukin-1β induces a healing-associated wound macrophage phenotype and improves healing in type 2 diabetes. Diabetes 62, 2579–2587 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Dinarello, C. A., Simon, A. & van der Meer, J. W. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nature Rev. Drug Discov. 11, 633–652 (2012).

    Article  CAS  Google Scholar 

  146. Martinon, F., Petrilli, V., Mayor, A., Tardivel, A. & Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440, 237–241 (2006).

    Article  CAS  PubMed  Google Scholar 

  147. Dinarello, C. A. How interleukin-1β induces gouty arthritis. Arthritis Rheum. 62, 3140–3144 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Schlesinger, N. et al. Canakinumab for acute gouty arthritis in patients with limited treatment options: results from two randomised, multicentre, active-controlled, double-blind trials and their initial extensions. Ann. Rheum. Dis. 71, 1839–1848 (2012).

    Article  CAS  PubMed  Google Scholar 

  149. Yeung, H. et al. Psoriasis severity and the prevalence of major medical comorbidity: a population-based study. JAMA Dermatol. 149, 1173–1179 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  150. Libby, P., Ridker, P. M., Hansson, G. K. & Leducq Transatlantic Network on Atherothrombosis. Inflammation in atherosclerosis: from pathophysiology to practice. J. Am. Coll.Cardiol. 54, 2129–2138 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Ridker, P. M., Thuren, T., Zalewski, A. & Libby, P. Interleukin-1β inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am. Heart J. 162, 597–605 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author wishes to thank his scientific collaborators who have contributed so much to these studies, in particular K. Maedler, M. Faulenbach, C. M. Larsen, D. M. Schumann, J. Ehses, H. Ellingsgaard, C. Cavelti-Weder, K. Timper, M. Böni-Schnetzler, P. A. Halban, T. Mandrup Poulsen and C. A. Dinarello.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Marc Y. Donath.

Ethics declarations

Competing interests

M.Y.D. is listed as the inventor on a patent (WO6709) filed in 2003 for the use of an interleukin-1 receptor antagonist for the treatment of, or prophylaxis against, type 2 diabetes.

Related links

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Donath, M. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov 13, 465–476 (2014). https://doi.org/10.1038/nrd4275

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd4275

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research