Skip to main content

Advertisement

Log in

Emerging Role of AMPK in Brown and Beige Adipose Tissue (BAT): Implications for Obesity, Insulin Resistance, and Type 2 Diabetes

  • Pathogenesis of Type 2 Diabetes and Insulin Resistance (M-E Patti, Section Editor)
  • Published:
Current Diabetes Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

The global prevalence of type 2 diabetes (T2D) is escalating at alarming rates, demanding the development of additional classes of therapeutics to further reduce the burden of disease. Recent studies have indicated that increasing the metabolic activity of brown and beige adipose tissue may represent a novel means to reduce circulating glucose and lipids in people with T2D. The AMP-activated protein kinase (AMPK) is a cellular energy sensor that has recently been demonstrated to be important in potentially regulating the metabolic activity of brown and beige adipose tissue. The goal of this review is to summarize recent work describing the role of AMPK in brown and beige adipose tissue, focusing on its role in adipogenesis and non-shivering thermogenesis.

Recent Findings

Ablation of AMPK in mouse adipocytes results in cold intolerance, a reduction in non-shivering thermogenesis in brown adipose tissue (BAT), and the development of non-alcoholic fatty liver disease (NAFLD) and insulin resistance; effects associated with a defect in mitochondrial specific autophagy (mitophagy) within BAT. The effects of a β3-adrenergic agonist on the induction of BAT thermogenesis and the browning of white adipose tissue (WAT) are also blunted in mice lacking adipose tissue AMPK. A specific AMPK activator, A-769662, also results in the activation of BAT and the browning of WAT, effects which may involve demethylation of the PR domain containing 16 (Prdm16) promoter region, which is important for BAT development.

Summary

AMPK plays an important role in the development and maintenance of brown and beige adipose tissue. Adipose tissue AMPK is reduced in people with insulin resistance, consistent with findings that mice lacking adipocyte AMPK develop greater NAFLD and insulin resistance. These data suggest that pharmacologically targeting adipose tissue AMPK may represent a promising strategy to enhance energy expenditure and reduce circulating glucose and lipids, which may be effective for the treatment of NAFLD and T2D.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. The NCD Risk Factor Collaboration Team. The weight of the world – trends in adult body mass index in 200 countries since 1975: pooled analysis of 1,698 population-based measurement studies with 19.2 million participants. Lancet. 2017;390:2627–42.

    Article  Google Scholar 

  2. The NCD Risk Factor Collaboration Team. Worldwide trends in diabetes since 1980: a pooled analysis of 751 population-based studies with 4.4 million participants. Lancet. 2016;387:1513–30.

    Article  Google Scholar 

  3. Hardie DG. Keeping the home fires burning: AMP-activated protein kinase. J R Soc Interface. 2018;15:20170774.

    Article  PubMed Central  PubMed  Google Scholar 

  4. Oakhill JS, Scott JW, Kemp BE. AMPK functions as an adenylate charge-regulated protein kinase. Trends Endocrinol Metab. 2012;23:125–32.

    Article  CAS  PubMed  Google Scholar 

  5. Zhang CS, Hawley SA, Zong Y, Li M, Wang Z, Gray A, et al. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature. 2017;548:112–6.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Zhang CS, Jiang B, Li M, Zhu M, Peng Y, Zhang YL, et al. The lysosomal v-ATPase-ragulator complex is a common activator for AMPK and mTORC1, acting as a switch between catabolism and anabolism. Cell Metab. 2014;20:526–40.

    Article  CAS  PubMed  Google Scholar 

  7. Zhang YL, Guo H, Zhang CS, Lin SY, Yin Z, Peng Y, et al. AMP as a low-energy charge signal autonomously initiates assembly of axin-ampk-lkb1 complex for AMPK activation. Cell Metab. 2013;18:546–55.

    Article  CAS  PubMed  Google Scholar 

  8. Dite TA, Ling NXY, Scott JW, Hoque A, Galic S, Parker BL, et al. The autophagy initiator ULK1 sensitizes AMPK to allosteric drugs. Nat Commun. 2017;8:1–13.

    Article  CAS  Google Scholar 

  9. Kjøbsted R, Hingst JR, Fentz J, Foretz M, Sanz M-N, Pehmøller C, et al. AMPK in skeletal muscle function and metabolism. FASEB J. 2018;32:1741–77.

    Article  PubMed Central  PubMed  Google Scholar 

  10. O’Neill HM, Maarbjerg SJ, Crane JD, Jeppesen J, Jørgensen SB, Schertzer JD, et al. AMP-activated protein kinase (AMPK) beta1beta2 muscle null mice reveal an essential role for AMPK in maintaining mitochondrial content and glucose uptake during exercise. Proc Natl Acad Sci U S A. 2011;108:16092–7.

    Article  PubMed Central  PubMed  Google Scholar 

  11. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invesit. 2001;108:1167–74.

    Article  CAS  Google Scholar 

  12. Hawley SA, Ford RJ, Smith BK, Gowans GJ, Mancini SJ, Pitt RD, et al. The Na+/glucose cotransporter inhibitor canagliflozin activates AMPK by inhibiting mitochondrial function and increasing cellular AMP levels. Diabetes. 2016;65:2784–94.

    Article  CAS  PubMed  Google Scholar 

  13. Fullerton MD, Galic S, Marcinko K, Sikkema S, Pulinilkunnil T, Chen Z, et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat Med. 2013;19:1649–54.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Madiraju AK, Qiu Y, Perry RJ, Rahimi Y, Zhang X-M, Zhang D, et al. Metformin inhibits gluconeogenesis via a redox-dependent mechanism in vivo. Nat Med. 2018;1

  15. Kim EK, Lee SH, Jhun JY, Byun JK, Jeong JH, Lee SY, et al. Metformin prevents fatty liver and improves balance of white/brown adipose in an obesity mouse model by inducing FGF21. Mediators Inflamm. 2016;ID5813030.

  16. Salastekar N, Desai T, Hauser T, Schaefer EJ, Fowler K, Joseph S, et al. Salsalate improves glycaemia in overweight persons with diabetes risk factors of stable statin-treated cardiovascular disease: a 30-month randomized placebo-controlled trial. Diabetes Obes Metab. 2017;19:1458–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Goldfine AB, Fonseca V, Jablonski KA, Pyle L, Staten MA, Shoelson SE. The effects of salsalate on glycemic control in patients with type 2 diabetes: a randomized trial. Ann Intern Med. 2010;152:346–57.

    Article  PubMed Central  PubMed  Google Scholar 

  18. Smith BK, Ford RJ, Desjardins EM, Green AE, Hughes MC, Houde VP, et al. Salsalate (salicylate) uncouples mitochondria, improves glucose homeostasis, and reduces liver lipids independent of AMPK-β1. Diabetes. 2016;65:3352–61.

    Article  CAS  PubMed  Google Scholar 

  19. Hawley SA, Fullerton MD, Ross FA, Schertzer JD, Chevtzoff C, Walker KJ, et al. The ancient drug salicylate directly activates AMP-activated protein kinase. Science. 2012;336:918–22.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Ford RJ, Fullerton MD, Pinkosky SL, Day EA, Scott JW, Oakhill JS, et al. Metformin and salicylate synergistically activate liver AMPK, inhibit lipogenesis and improve insulin sensitivity. Biochem J. 2015;468:125–32.

    Article  CAS  PubMed  Google Scholar 

  21. Myers RW, Guan HP, Ehrhart J, Petrov A, Prahalada S, Tozzo E, et al. Systemic pan-AMPK activator MK-8722 improves glucose homeostasis but induces cardiac hypertrophy. Science. 2017;357:507–11.

    Article  CAS  PubMed  Google Scholar 

  22. Cokorinos EC, Delmore J, Reyes AR, Albuquerque B, Kjøbsted R, Jørgensen NO, et al. Activation of skeletal muscle AMPK promotes glucose disposal and glucose lowering in non-human Primates and mice. Cell Metab. 2017;25:1147–59.

    Article  CAS  PubMed  Google Scholar 

  23. Steneberg P, Edlund T, Edlund H. PAN-AMPK activator O304 improves glucose homeostasis and microvascular perfusion in mice and type 2 diabetes patients. 2018; 3:e99114

  24. Esquejo RM, Salatto CT, Delmore J, Albuquerque B, Reyes A, Shi Y, et al. Activation of liver AMPK with PF-06409577 corrects NAFLD and lowers cholesterol in rodent and primate preclinical models. EBioMedicine. 2018;31:122–32.

    Article  PubMed Central  PubMed  Google Scholar 

  25. Sanchez-Gurmaches J, Hung CM, Guertin DA. Emerging complexities in adipocyte origins and identity. Trends Cell Biol. 2016;26:313–26.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Wang W, Seale P. Control of brown and beige fat development. Nat Rev Mol Cell Biol. 2016;17:691–702.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Kazak L, Chouchani ET, Jedrychowski MP, Erickson BK, Shinoda K, Cohen P, et al. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell. 2015;163:643–55.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Ikeda K, Kang Q, Yoneshiro T, Camporez JP, Maki H, Homma M, et al. UCP1-independent signaling involving SERCA2bmediated calcium cycling regulates beige fat thermogenesis and systemic glucose homeostasis. Nat Med. 2017;23:1454–65.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Mulligan JD, Gonzalez AA, Stewart AM, Carey HV, Saupe KW. Upregulation of AMPK during cold exposure occurs via distinct mechanisms in brown and white adipose tissue of the mouse. J Physiol. 2007;580:677–84.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. MacPherson REK, Dragos SM, Ramos S, Sutton C, Frendo-Cumbo S, Castellani L, et al. Reduced ATGL-mediated lipolysis attenuates β-adrenergic-induced AMPK signaling, but not the induction of PKA-targeted genes, in adipocytes and adipose tissue. Am J Phys Cell Phys. 2016;311:C269–76.

    Article  Google Scholar 

  31. Hutchinson DS, Chernogubova E, Dallner OS, Cannon B, Bengtsson T. Beta-adrenoceptors, but not alpha-adrenoceptors, stimulate AMP-activated protein kinase in brown adipocytes independently of uncoupling protein-1. Diabetologia. 2005;48:2386–95.

    Article  CAS  PubMed  Google Scholar 

  32. • Mottillo EP, Desjardins EM, Crane JD, Smith BK, Green AE, Ducommun S, et al. Lack of adipocyte AMPK exacerbates insulin resistance and hepatic steatosis through brown and beige adipose tissue function. Cell Metab. 2016;24:118–29. Characterization of an inducible, adipocyte-specific AMPK β1β2 knockout mouse model showing that AMPK is required for cold and β-adrenergic-stimulated thermogenesis, required for the browning of WAT, and, when absent, results in aggravated insulin resistance and hepatic lipid accumulation in response to high-fat diet. This characterization is explained through the role of AMPK in maintaining mitochondrial homeostasis through the regulation of mitophagy

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Blondin DP, Labbé SM, Noll C, Kunach M, Phoenix S, Guérin B, et al. Selective impairment of glucose but not fatty acid or oxidative metabolism in brown adipose tissue of subjects with type 2 diabetes. Diabetes. 2015;64:2388–97.

    Article  CAS  PubMed  Google Scholar 

  34. Ong FJ, Ahmed BA, Oreskovich SM, Blondin DP, Haq T, Konyer NB, et al. Recent advances in the detection of brown adipose tissue in adult humans: a review 2018;132:1039–54.

  35. Xu XJ, Gauthier M, Hess DT, Apovian CM, Cacicedo JM, Gokce N, et al. Insulin sensitive and resistant obesity in humans : AMPK activity , oxidative stress , and depot-specifi c changes in gene expression in adipose tissue. J Lipid Res. 2012;53:792–801.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. Fritzen AMH, Lundsgaard AM, Jordy AB, Poulsen SK, Stender S, Pilegaard H, et al. New nordic diet-induced weight loss is accompanied by changes in metabolism and AMPK signaling in adipose tissue. J Clin Endocrinol Metab. 2015;100:3509–19.

    Article  CAS  PubMed  Google Scholar 

  37. Julia Xu X, Apovian C, Hess D, Carmine B, Saha A, Ruderman N. Improved insulin sensitivity 3 months after RYGB surgery is associated with increased subcutaneous adipose tissue AMPK activity and decreased oxidative stress. Diabetes. 2015;64:3155–9.

    Article  CAS  PubMed  Google Scholar 

  38. Albers PH, Bojsen-Møller KN, Dirksen C, Serup AK, Kristensen DE, Frystyk J, et al. Enhanced insulin signaling in human skeletal muscle and adipose tissue following gastric bypass surgery. Am J Phys Regul Integr Comp Phys. 2015;309:R510–24.

    CAS  Google Scholar 

  39. Qi J, Gong J, Zhao T, Zhao J, Lam P, Ye J, et al. Downregulation of AMP-activated protein kinase by Cidea-mediated ubiquitination and degradation in brown adipose tissue. EMBO J. 2008;27:1537–48.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Perdikari A, Kulenkampff E, Rudigier C, Neubauer H, Luippold G, Redemann N, et al. A high-throughput , image-based screen to identify kinases involved in brown adipocyte development. Sci Signal. 2017;10:eaaf5357.

    Article  CAS  PubMed  Google Scholar 

  41. Zhao J, Yang Q, Zhang L, Liang X, Sun X, Wang B, et al. AMPKα1 deficiency suppresses brown adipogenesis in favor of fibrogenesis during brown adipose tissue development. Biochem Biophys Res Commun. 2017;491:508–14.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. •• Yang Q, Liang X, Sun X, Zhang L, Fu X, Rogers CJ, et al. AMPK/α-Ketoglutarate axis dynamically mediates DNA demethylation in the Prdm16 promoter and brown adipogenesis. Cell Metab. 2016;24:542–54. The authors show a novel mechanism by which AMPK is essential in brown adipose tissue development through elevating α-ketoglutarate production and subsequently increasing the demethylation of the Prdm16 promoter

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  43. Dagon Y, Avraham Y, Berry EM. AMPK activation regulates apoptosis, adipogenesis, and lipolysis by eIF2α in adipocytes. Biochem Biophys Res Commun. 2006;340:43–7.

    Article  CAS  PubMed  Google Scholar 

  44. Vila-Bedmar R, Lorenzo M, Fernández-Veledo S. Adenosine 5′-monophosphate-activated protein kinase-mammalian target of rapamycin cross talk regulates brown adipocyte differentiation. Endocrinology. 2010;151:980–92.

    Article  CAS  PubMed  Google Scholar 

  45. Garcia D, Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Mol Cell. 2017;66:789–800.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Polak P, Cybulski N, Feige JN, Auwerx J, Rüegg MA, Hall MN. Adipose-specific knockout of raptor results in lean mice with enhanced mitochondrial respiration. Cell Metab. 2008;8:399–410.

    Article  CAS  PubMed  Google Scholar 

  47. Olsen JM, Sato M, Dallner OS, Sandström AL, Pisani DF, Chambard JC, et al. Glucose uptake in brown fat cells is dependent on mTOR complex 2-promoted GLUT1 translocation. J Cell Biol. 2014;207:365–74.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  48. Labbé SM, Mouchiroud M, Caron A, Secco B, Freinkman E, Lamoureux G, et al. MTORC1 is required for brown adipose tissue recruitment and metabolic adaptation to cold. Sci Rep. 2016;6:1–17.

    Article  CAS  Google Scholar 

  49. Olsen JM, Csikasz RI, Dehvari N, Lu L, Sandström A, Öberg AI, et al. β3-Adrenergically induced glucose uptake in brown adipose tissue is independent of UCP1 presence or activity: mediation through the mTOR pathway. Mol Metab. 2017;6:611–9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  50. Su X, Wellen KE, Rabinowitz JD. Metabolic control of methylation and acetylation. Curr Opin Chem Biol. 2016;30:52–60.

    Article  CAS  PubMed  Google Scholar 

  51. Than A, He HL, Chua SH, Xu D, Sun L, Leow MKS, et al. Apelin enhances brown adipogenesis and browning of white adipocytes. J Biol Chem. 2015;290:14679–91.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Trajkovski M, Lodish H. MicroRNA networks regulate development of brown adipocytes. Trends Endocrinol Metab. 2013;24:442–50.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Zhang H, Guan M, Townsend KL, Huang TL, An D, Yan X, et al. MicroRNA- 455 regulates brown adipogenesis via a novel HIF 1 an-AMPK-PGC 1 a signaling network. EMBO Rep. 2015;16:1378–93.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  54. Yan M, Audet-Walsh É, Manteghi S, Dufour CR, Walker B, Baba M, et al. Chronic AMPK activation via loss of FLCN induces functional beige adipose tissue through PGC-1 α / ERR α. Genes Dev. 2016;30:1034–46.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  55. Imran KM, Rahman N, Yoon D, Jeon M, Lee BT, Kim YS. Cryptotanshinone promotes commitment to the brown adipocyte lineage and mitochondrial biogenesis in C3H10T1/2 mesenchymal stem cells via AMPK and p38-MAPK signaling. Biochim Biophys Acta Mol Cell Biol Lipids. 1862;2017:1110–20.

    Google Scholar 

  56. Imran KM, Yoon D, Kim YS. A pivotal role of AMPK signaling in medicarpin-mediated formation of brown and beige. Biofactors. 2018;44:168–79.

    Article  CAS  PubMed  Google Scholar 

  57. Wang S, Liang X, Yang Q, Fu X, Zhu M, Rodgers BD, et al. Resveratrol enhances brown adipocyte formation and function by activating AMP-activated protein kinase (AMPK) α1 in mice fed high-fat diet. Mol Nutr Food Res. 2017;61:1–11.

    CAS  Google Scholar 

  58. • Wu L, Zhang L, Li B, Jiang H, Duan Y, Xie Z, et al. AMP-activated protein kinase (AMPK) regulates energy metabolism through modulating thermogenesis in adipose tissue. Front Physiol. 2018;9:1–11. This report demonstrates that chronic administration of A-769662, an AMPK activator, reduces body weight gain, increases energy expenditure, improves cold tolerance, and promotes the browning of inguinal WAT in high-fat diet-fed mice

    Google Scholar 

  59. Herzig S, Shaw RJ. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19:121–35.

    Article  CAS  PubMed  Google Scholar 

  60. Sharp LZ, Shinoda K, Ohno H, Scheel DW, Tomoda E, Ruiz L, et al. Human BAT possesses molecular signatures that resemble beige/Brite cells. PLoS One. 2012;7:e49452.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  61. Lidell ME, Betz MJ, Leinhard OD, Heglind M, Elander L, Slawik M, et al. Evidence for two types of brown adipose tissue in humans. Nat Med. 2013;19:631–4.

    Article  CAS  PubMed  Google Scholar 

  62. Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang A, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 2012;150:366–76.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Ikeda K, Maretich P, Kajimura S. The common and distinct features of Brown and Beige adipocytes. Trends Endocrinol Metab. 2018;29:191–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zhu Q, Ghoshal S, Rodrigues A, Gao S, Asterian A, Kamenecka TM, et al. Adipocyte-specific deletion of Ip6k1 reduces diet-induced obesity by enhancing AMPK-mediated thermogenesis. J Clin Invest. 2016;126:4273–88.

    Article  PubMed Central  PubMed  Google Scholar 

  65. Chung YW, Ahmad F, Tang Y, Hockman SC, Kee HJ, Berger K, et al. White to beige conversion in PDE3B KO adipose tissue through activation of AMPK signaling and mitochondrial function. Sci Rep. 2017;7:1–13.

    Article  CAS  Google Scholar 

  66. Geerling JJ, Boon MR, van der Zon GC, van den Berg SAA, van den Hoek AM, Lombes M, et al. Metformin lowers plasma triglycerides by promoting VLDL-triglyceride clearance by Brown adipose tissue in mice. Diabetes. 2014;63:880–91.

    Article  CAS  PubMed  Google Scholar 

  67. Zhang Z, Zhang H, Li B, Meng X, Wang J, Zhang Y, et al. Berberine activates thermogenesis in white and brown adipose tissue. Nat Commun. 2014;5:5493.

    Article  CAS  PubMed  Google Scholar 

  68. Zhang X, Zhang Q, Wang X, Zhang L, Qu W, Bao B, et al. Dietary luteolin activates browning and thermogenesis in mice through an AMPK/PGC1 α pathway-mediated mechanism. Int J Obes. 2016;40:1841–9.

    Article  CAS  Google Scholar 

  69. Zou T, Wang B, Yang Q, de Avila JM, Zhu MJ, You J, et al. Raspberry promotes brown and beige adipocyte development in mice fed high-fat diet through activation of AMP-activated protein kinase (AMPK) α1. J Nutr Biochem. 2018;55:157–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Lee M-S, Shin Y, Jung S, Kim Y. Effects of epigallocatechin-3-gallate on thermogenesis and mitochondrial biogenesis in brown adipose tissues of diet-induced obese mice. Food Nutr Res. 2017;61:1325307.

    Article  PubMed Central  PubMed  Google Scholar 

  71. Kirkwood JS, Legette LCL, Miranda CL, Jiang Y, Stevens JF. A metabolomics-driven elucidation of the anti-obesity mechanisms of xanthohumol. J Biol Chem. 2013;288:19000–13.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Zhang F, Ai W, Hu X, Meng Y, Yuan C, Su H, et al. Phytol stimulates the browning of white adipocytes through the activation of AMP-activated protein kinase (AMPK) α in mice fed high-fat diet. Food Funct. 2018;9:2043–50.

    Article  CAS  PubMed  Google Scholar 

  73. Shan T, Liang X, Bi P, Kuang S. Myostatin knockout drives browning of white adipose tissue through activating the AMPK-PGC1-Fndc5 pathway in muscle. FASEB J. 2013;27:1981–9.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  74. Berti L, Irmler M, Zdichavsky M, Meile T, Böhm A, Stefan N, et al. Fibroblast growth factor 21 is elevated in metabolically unhealthy obesity and affects lipid deposition, adipogenesis, and adipokine secretion of human abdominal subcutaneous adipocytes. Mol Metab. 2015;4:519–27.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  75. Chau MDL, Gao J, Yang Q, Wu Z, Gromada J. Fibroblast growth factor 21 regulates energy metabolism by activating the AMPK–SIRT1–PGC-1α pathway. Proc Natl Acad Sci. 2010;107:12553–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Mottillo EP, Desjardins EM, Fritzen AM, Zou VZ, Crane JD, Yabut JM, et al. FGF21 does not require adipocyte AMP-activated protein kinase (AMPK) or the phosphorylation of acetyl-CoA carboxylase (ACC) to mediate improvements in whole-body glucose homeostasis. Mol Metab. 2017;6:471–81.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  77. Wan Z, Root-mccaig J, Castellani L, Kemp BE, Steinberg GR, Wright DC. Evidence for the role of AMPK in regulating PGC-1 alpha expression and mitochondrial proteins in mouse epididymal adipose tissue. Obesity. 2014;22:730–8.

    Article  CAS  PubMed  Google Scholar 

  78. Wu L, Zhang L, Li B, Jiang H, Duan Y, Xie Z, et al. AMP-activated protein kinase (AMPK) regulates energy metabolism through modulating thermogenesis in adipose tissue. Front Physiol. 2018;9:1–23.

    Google Scholar 

Download references

Acknowledgments

The authors would like to express their gratitude to all the researchers who have contributed to the field of AMPK in brown/beige adipose tissue, especially those whose research they could not cite due to constraints on review length.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gregory R. Steinberg.

Ethics declarations

Conflict of Interest

Eric Desjardins declares no conflicts of interest. Dr. Steinberg reports grants, personal fees, and non-financial support from Esperion Therapeutics; non-financial support from Pfizer, Merck, Sanofi, and Nestle; and personal fees from Novo Nordisk and Eli Lilly.

Informed Consent

This article does not contain any experiments with human or animal subjects.

Additional information

This article is part of the Topical Collection on Pathogenesis of Type 2 Diabetes and Insulin Resistance

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Desjardins, E.M., Steinberg, G.R. Emerging Role of AMPK in Brown and Beige Adipose Tissue (BAT): Implications for Obesity, Insulin Resistance, and Type 2 Diabetes. Curr Diab Rep 18, 80 (2018). https://doi.org/10.1007/s11892-018-1049-6

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11892-018-1049-6

Keywords

Navigation