Skip to main content
Log in

Targeting Islets: Metabolic Surgery Is More than a Bariatric Surgery

  • Review Article
  • Published:
Obesity Surgery Aims and scope Submit manuscript

Abstract

Metabolic surgery is an effective therapy for diabetic patients with obesity. The main mechanisms underlying the effects of metabolic surgery include food intake restriction and the accompanying reduced daily caloric intake and changes in gut hormones and bile acid. Insulin resistance and impaired β-cell function contribute to the development of type 2 diabetes. An increasing number of studies have focused on the central role of islet function in type 2 diabetes. In this article, we review the related high-quality literature and summarize the following mechanisms and principles underlying metabolic surgery in the context of islet function protection: (1) reduced glucotoxicity and chronic inflammation help facilitate better β-cell function and the preservation of β-cell mass following metabolic surgery; (2) based on the increased levels of GLP-1 and PYY after metabolic surgery, gut hormones appear to play a significant role in improving β-cell function through the GLP-1R signaling pathways; (3) the bile acid signaling pathway could affect β-cell function; and (4) the GLP-1R and bile acid signaling pathways could also cause other endocrine cells to contribute to islet function.

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

Abbreviations

IDF:

International Diabetes Federation

T2D:

Type 2 diabetes

RYGB:

Roux-en-Y gastric bypass

SG:

Sleeve gastrostomy

BA:

Bile acid

GPCRs:

G protein-coupled receptors

PKC:

Protein kinase C

PKA:

Protein kinase A

IRS-2:

Insulin receptor substrate-2

PKB:

Protein kinase B

TGF-β:

Transforming growth factor-β

FFA:

Free fatty acid

GLP-1:

Glucagon-like peptide 1

PYY:

Peptide-YY

GIP:

Gastric inhibitory polypeptide

FXR:

Farnesoid X receptor

TGR5:

G protein-coupled bile acid receptor 5

AUC:

Area under the curve

FGF-19:

Fibroblast growth factor-19

GLP-1R:

GLP-1 receptor

IL-1β:

Interleukin-1β

NF-κB:

Nuclear factor-κB

References

  1. Cho NH, Shaw JE, Karuranga S. IDF diabetes atlas: global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271–81.

    Article  CAS  PubMed  Google Scholar 

  2. Yang W, Lu J, Weng J. Prevalence of diabetes among men and women in China. N Engl J Med. 2010;362(12):1090–101.

    Article  CAS  PubMed  Google Scholar 

  3. NCD-RisC NRFC. Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19·2 million participants. Lancet. 2016;387(10026):1377–96.

    Article  Google Scholar 

  4. Ramos-Levi AM, Rubio MA. Comment on Rubino et al. metabolic surgery in the treatment algorithm for type 2 diabetes: a joint statement by International Diabetes Organizations. Diabetes Care 2016;39:861–877. Diabetes Care. 2017;40(7):e90–1.

    Article  PubMed  Google Scholar 

  5. ADA. Standards of medical care in diabetes--2009. Diabetes Care. 2009;32 Suppl 1:S13–61.

    Google Scholar 

  6. Dixon JB, Zimmet P, Alberti KG. Bariatric surgery: an IDF statement for obese type 2 diabetes. Diabet Med. 2011;28(6):628–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Accili D. Lilly lecture 2003: the struggle for mastery in insulin action: from triumvirate to republic. Diabetes. 2004;53(7):1633–42.

    Article  CAS  PubMed  Google Scholar 

  8. Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol. 2011;11(11):738–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov. 2014;13(6):465–76.

    Article  CAS  PubMed  Google Scholar 

  10. Shu CJ, Benoist C, Mathis D. The immune system’s involvement in obesity-driven type 2 diabetes. Semin Immunol. 2012;24(6):436–42.

    Article  CAS  PubMed  Google Scholar 

  11. Ferrannini E. Insulin resistance versus insulin deficiency in non-insulin-dependent diabetes mellitus: problems and prospects. Endocr Rev. 1998;19(4):477–90.

    Article  CAS  PubMed  Google Scholar 

  12. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11(2):98–107.

    Article  CAS  PubMed  Google Scholar 

  13. Cerf ME. Beta cell dysfunction and insulin resistance. Front Endocrinol (Lausanne). 2013;4:37.

    Article  Google Scholar 

  14. Heit JJ, Karnik SK, Kim SK. Intrinsic regulators of pancreatic beta-cell proliferation. Annu Rev Cell Dev Biol. 2006;22:311–38.

    Article  CAS  PubMed  Google Scholar 

  15. Vangoitsenhoven R, Mathieu C, Van der Schueren B. GLP1 and cancer: friend or foe? Endocr Relat Cancer. 2012;19(5):F77–88.

    Article  CAS  PubMed  Google Scholar 

  16. Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes. J Clin Invest. 2006;116(7):1802–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kalupahana NS, Moustaid-Moussa N, Claycombe KJ. Immunity as a link between obesity and insulin resistance. Mol Asp Med. 2012;33(1):26–34.

    Article  CAS  Google Scholar 

  18. Lee BC, Lee J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim Biophys Acta. 2014;1842(3):446–62.

    Article  CAS  PubMed  Google Scholar 

  19. Butler PC, Meier JJ, Butler AE. The replication of beta cells in normal physiology, in disease and for therapy. Nat Clin Pract Endocrinol Metab. 2007;3(11):758–68.

    Article  CAS  PubMed  Google Scholar 

  20. Talchai C, Lin HV, Kitamura T. Genetic and biochemical pathways of beta-cell failure in type 2 diabetes. Diabetes Obes Metab. 2009;11(Suppl 4):38–45.

    Article  CAS  PubMed  Google Scholar 

  21. Merino B, Alonso-Magdalena P, Lluesma M. Pancreatic alpha-cells from female mice undergo morphofunctional changes during compensatory adaptations of the endocrine pancreas to diet-induced obesity. Sci Rep. 2015;5:11622.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Liu H, Javaheri A, Godar RJ. Intermittent fasting preserves beta-cell mass in obesity-induced diabetes via the autophagy-lysosome pathway. Autophagy. 2017;13(11):1952–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tang C, Ahmed K, Gille A. Loss of FFA2 and FFA3 increases insulin secretion and improves glucose tolerance in type 2 diabetes. Nat Med. 2015;21(2):173–7.

    Article  CAS  PubMed  Google Scholar 

  24. Larsen CM, Faulenbach M, Vaag A. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517–26.

    Article  CAS  PubMed  Google Scholar 

  25. Bunck MC, Diamant M, Corner A. One-year treatment with exenatide improves beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a randomized, controlled trial. Diabetes Care. 2009;32(5):762–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xu W, Bi Y, Sun Z. Comparison of the effects on glycaemic control and beta-cell function in newly diagnosed type 2 diabetes patients of treatment with exenatide, insulin or pioglitazone: a multicentre randomized parallel-group trial (the CONFIDENCE study). J Intern Med. 2015;277(1):137–50.

    Article  CAS  PubMed  Google Scholar 

  27. Yang Z, Zhou Z, Li X. Rosiglitazone preserves islet beta-cell function of adult-onset latent autoimmune diabetes in 3 years follow-up study. Diabetes Res Clin Pract. 2009;83(1):54–60.

    Article  CAS  PubMed  Google Scholar 

  28. Zhao Y, Yang L, Xiang Y. Dipeptidyl peptidase 4 inhibitor sitagliptin maintains beta-cell function in patients with recent-onset latent autoimmune diabetes in adults: one year prospective study. J Clin Endocrinol Metab. 2014;99(5):E876–80.

    Article  CAS  PubMed  Google Scholar 

  29. Yin YN, Yu QF, Fu N. Effects of four Bifidobacteria on obesity in high-fat diet induced rats. World J Gastroenterol. 2010;16(27):3394–401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Karamanakos SN, Vagenas K, Kalfarentzos F. Weight loss, appetite suppression, and changes in fasting and postprandial ghrelin and peptide-YY levels after Roux-en-Y gastric bypass and sleeve gastrectomy: a prospective, double blind study. Ann Surg. 2008;247(3):401–7.

    Article  PubMed  Google Scholar 

  31. Peterli R, Wolnerhanssen B, Peters T. Improvement in glucose metabolism after bariatric surgery: comparison of laparoscopic Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy: a prospective randomized trial. Ann Surg. 2009;250(2):234–41.

    Article  PubMed  Google Scholar 

  32. Peterli R, Borbely Y, Kern B. Early results of the Swiss Multicentre Bypass or Sleeve Study (SM-BOSS): a prospective randomized trial comparing laparoscopic sleeve gastrectomy and roux-en-Y gastric bypass. Ann Surg. 2013;258(5):690–694, 695.

    Article  PubMed  Google Scholar 

  33. Peterli R, Wolnerhanssen BK, Vetter D. Laparoscopic sleeve gastrectomy versus Roux-Y-gastric bypass for morbid obesity-3-year outcomes of the prospective randomized Swiss Multicenter Bypass or Sleeve Study (SM-BOSS). Ann Surg. 2017;265(3):466–73.

    Article  PubMed  Google Scholar 

  34. Ochner CN, Gibson C, Shanik M. Changes in neurohormonal gut peptides following bariatric surgery. Int J Obes. 2011;35(2):153–66.

    Article  CAS  Google Scholar 

  35. Lee WJ, Chen CY, Chong K. Changes in postprandial gut hormones after metabolic surgery: a comparison of gastric bypass and sleeve gastrectomy. Surg Obes Relat Dis. 2011;7(6):683–90.

    Article  PubMed  Google Scholar 

  36. Thaler JP, Cummings DE. Minireview: hormonal and metabolic mechanisms of diabetes remission after gastrointestinal surgery. Endocrinology. 2009;150(6):2518–25.

    Article  CAS  PubMed  Google Scholar 

  37. Cummings DE, Weigle DS, Frayo RS. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346(21):1623–30.

    Article  PubMed  Google Scholar 

  38. Holdstock C, Engstrom BE, Ohrvall M. Ghrelin and adipose tissue regulatory peptides: effect of gastric bypass surgery in obese humans. J Clin Endocrinol Metab. 2003;88(7):3177–83.

    Article  CAS  PubMed  Google Scholar 

  39. Stoeckli R, Chanda R, Langer I. Changes of body weight and plasma ghrelin levels after gastric banding and gastric bypass. Obes Res. 2004;12(2):346–50.

    Article  CAS  PubMed  Google Scholar 

  40. Rubino F, Gagner M, Gentileschi P. The early effect of the Roux-en-Y gastric bypass on hormones involved in body weight regulation and glucose metabolism. Ann Surg. 2004;240(2):236–42.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Clements RH, Gonzalez QH, Long CI. Hormonal changes after Roux-en-Y gastric bypass for morbid obesity and the control of type-II diabetes mellitus. Am Surg. 2004;70(1):1–4. 4-5

    PubMed  Google Scholar 

  42. DePaula AL, Macedo AL, Schraibman V. Hormonal evaluation following laparoscopic treatment of type 2 diabetes mellitus patients with BMI 20-34. Surg Endosc. 2009;23(8):1724–32.

    Article  PubMed  Google Scholar 

  43. Farey JE, Preda TC, Fisher OM. Effect of laparoscopic sleeve gastrectomy on fasting gastrointestinal, pancreatic, and adipose-derived hormones and on non-esterified fatty acids. Obes Surg. 2017;27(2):399–407.

    Article  PubMed  Google Scholar 

  44. Jahansouz C, Xu H, Hertzel AV. Bile acids increase independently from hypocaloric restriction after bariatric surgery. Ann Surg. 2016;264(6):1022–8.

    Article  PubMed  Google Scholar 

  45. Jorgensen NB, Dirksen C, Bojsen-Moller KN. Improvements in glucose metabolism early after gastric bypass surgery are not explained by increases in total bile acids and fibroblast growth factor 19 concentrations. J Clin Endocrinol Metab. 2015;100(3):E396–406.

    Article  CAS  PubMed  Google Scholar 

  46. Khan FH, Shaw L, Zhang W. Fibroblast growth factor 21 correlates with weight loss after vertical sleeve gastrectomy in adolescents. Obesity (Silver Spring). 2016;24(11):2377–83.

    Article  CAS  Google Scholar 

  47. Steinert RE, Peterli R, Keller S. Bile acids and gut peptide secretion after bariatric surgery: a 1-year prospective randomized pilot trial. Obesity (Silver Spring). 2013;21(12):E660–8.

    Article  CAS  Google Scholar 

  48. Ryan KK, Tremaroli V, Clemmensen C. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509(7499):183–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ding L, Sousa KM, Jin L. Vertical sleeve gastrectomy activates GPBAR-1/TGR5 to sustain weight loss, improve fatty liver, and remit insulin resistance in mice. Hepatology. 2016;64(3):760–73.

    Article  CAS  PubMed  Google Scholar 

  50. McGavigan AK, Garibay D, Henseler ZM. TGR5 contributes to glucoregulatory improvements after vertical sleeve gastrectomy in mice. Gut. 2017;66(2):226–34.

    Article  CAS  PubMed  Google Scholar 

  51. Ley RE, Backhed F, Turnbaugh P. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102(31):11070–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ley RE, Turnbaugh PJ, Klein S. Microbial ecology: Human gut microbes associated with obesity. Nature. 2006;444(7122):1022–3.

    Article  CAS  PubMed  Google Scholar 

  53. Liou AP, Paziuk M, Luevano JJ. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5(178):141r–78r.

    Article  CAS  Google Scholar 

  54. Li JV, Ashrafian H, Bueter M. Metabolic surgery profoundly influences gut microbial-host metabolic cross-talk. Gut. 2011;60(9):1214–23.

    Article  CAS  PubMed  Google Scholar 

  55. Zhang H, DiBaise JK, Zuccolo A. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci U S A. 2009;106(7):2365–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Furet JP, Kong LC, Tap J. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59(12):3049–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Talchai C, Xuan S, Lin HV. Pancreatic beta cell dedifferentiation as a mechanism of diabetic beta cell failure. Cell. 2012;150(6):1223–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Taylor BL, Liu FF, Sander M. Nkx6.1 is essential for maintaining the functional state of pancreatic beta cells. Cell Rep. 2013;4(6):1262–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Puri S, Akiyama H, Hebrok M. VHL-mediated disruption of Sox9 activity compromises beta-cell identity and results in diabetes mellitus. Genes Dev. 2013;27(23):2563–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Guo S, Dai C, Guo M. Inactivation of specific beta cell transcription factors in type 2 diabetes. J Clin Invest. 2013;123(8):3305–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang Z, York NW, Nichols CG. Pancreatic beta cell dedifferentiation in diabetes and redifferentiation following insulin therapy. Cell Metab. 2014;19(5):872–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Cinti F, Bouchi R, Kim-Muller JY. Evidence of beta-cell dedifferentiation in human type 2 diabetes. J Clin Endocrinol Metab. 2016;101(3):1044–54.

    Article  CAS  PubMed  Google Scholar 

  63. Savage PJ, Bennion LJ, Flock EV. Diet-induced improvement of abnormalities in insulin and glucagon secretion and in insulin receptor binding in diabetes mellitus. J Clin Endocrinol Metab. 1979;48(6):999–1007.

    Article  CAS  PubMed  Google Scholar 

  64. Greenwood RH, Mahler RF, Hales CN. Improvement in insulin secretion in diabetes after diazoxide. Lancet. 1976;1(7957):444–7.

    Article  CAS  PubMed  Google Scholar 

  65. Wajchenberg BL. Beta-cell failure in diabetes and preservation by clinical treatment. Endocr Rev. 2007;28(2):187–218.

    Article  CAS  PubMed  Google Scholar 

  66. Zhou X, Qian B, Ji N. Pancreatic hyperplasia after gastric bypass surgery in a GK rat model of non-obese type 2 diabetes. J Endocrinol. 2016;228(1):13–23.

    Article  CAS  PubMed  Google Scholar 

  67. Qian B, Zhou X, Li B. Reduction of pancreatic beta-cell dedifferentiation after gastric bypass surgery in diabetic rats. J Mol Cell Biol. 2014;6(6):531–4.

    Article  PubMed  Google Scholar 

  68. Roslin MS, Dudiy Y, Weiskopf J. Comparison between RYGB, DS, and VSG effect on glucose homeostasis. Obes Surg. 2012;22(8):1281–6.

    Article  PubMed  Google Scholar 

  69. Mari A, Manco M, Guidone C. Restoration of normal glucose tolerance in severely obese patients after bilio-pancreatic diversion: role of insulin sensitivity and beta cell function. Diabetologia. 2006;49(9):2136–43.

    Article  CAS  PubMed  Google Scholar 

  70. Nannipieri M, Mari A, Anselmino M. The role of beta-cell function and insulin sensitivity in the remission of type 2 diabetes after gastric bypass surgery. J Clin Endocrinol Metab. 2011;96(9):E1372–9.

    Article  CAS  PubMed  Google Scholar 

  71. Chiellini C, Iaconelli A, Familiari P. Study of the effects of transoral gastroplasty on insulin sensitivity and secretion in obese subjects. Nutr Metab Cardiovasc Dis. 2010;20(3):202–7.

    Article  CAS  PubMed  Google Scholar 

  72. Bosello O, Armellini F, Pelloso M. Glucose tolerance in jejunoileal bypass for morbid obesity: a fifteen month follow-up. Diabete Metab. 1978;4(3):159–62.

    CAS  PubMed  Google Scholar 

  73. Sirinek KR, O’Dorisio TM, Hill D. Hyperinsulinism, glucose-dependent insulinotropic polypeptide, and the enteroinsular axis in morbidly obese patients before and after gastric bypass. Surgery. 1986;100(4):781–7.

    CAS  PubMed  Google Scholar 

  74. Karayiannakis AJ, Syrigos KN, Zbar A. The effect of vertical banded gastroplasty on glucose-induced beta-endorphin response. J Surg Res. 1998;80(2):123–8.

    Article  CAS  PubMed  Google Scholar 

  75. Kopp HP, Kopp CW, Festa A. Impact of weight loss on inflammatory proteins and their association with the insulin resistance syndrome in morbidly obese patients. Arterioscler Thromb Vasc Biol. 2003;23(6):1042–7.

    Article  CAS  PubMed  Google Scholar 

  76. Korner J, Inabnet W, Febres G. Prospective study of gut hormone and metabolic changes after adjustable gastric banding and Roux-en-Y gastric bypass. Int J Obes. 2009;33(7):786–95.

    Article  CAS  Google Scholar 

  77. de Carvalho CP, Marin DM, de Souza AL. GLP-1 and adiponectin: effect of weight loss after dietary restriction and gastric bypass in morbidly obese patients with normal and abnormal glucose metabolism. Obes Surg. 2009;19(3):313–20.

    Article  PubMed  Google Scholar 

  78. De Paula AL, Stival AR, Halpern A. Improvement in insulin sensitivity and beta-cell function following ileal interposition with sleeve gastrectomy in type 2 diabetic patients: potential mechanisms. J Gastrointest Surg. 2011;15(8):1344–53.

    Article  PubMed  Google Scholar 

  79. Lee WJ, Ser KH, Chong K. Laparoscopic sleeve gastrectomy for diabetes treatment in nonmorbidly obese patients: efficacy and change of insulin secretion. Surgery. 2010;147(5):664–9.

    Article  PubMed  Google Scholar 

  80. Umeda LM, Silva EA, Carneiro G. Early improvement in glycemic control after bariatric surgery and its relationships with insulin, GLP-1, and glucagon secretion in type 2 diabetic patients. Obes Surg. 2011;21(7):896–901.

    Article  PubMed  Google Scholar 

  81. Campos GM, Rabl C, Peeva S. Improvement in peripheral glucose uptake after gastric bypass surgery is observed only after substantial weight loss has occurred and correlates with the magnitude of weight lost. J Gastrointest Surg. 2010;14(1):15–23.

    Article  PubMed  Google Scholar 

  82. Laferrere B, Heshka S, Wang K. Incretin levels and effect are markedly enhanced 1 month after Roux-en-Y gastric bypass surgery in obese patients with type 2 diabetes. Diabetes Care. 2007;30(7):1709–16.

    Article  CAS  PubMed  Google Scholar 

  83. Cummings DE, Cohen RV. Response to Comment on: Cohen et al. Effects of gastric bypass surgery in patients with type 2 diabetes and only mild obesity. Diabetes Care 2012;35:1420–1428. Diabetes Care. 2013;36(4):e59.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Cummings DE. Metabolic surgery for type 2 diabetes. Nat Med. 2012;18(5):656–8.

    Article  CAS  PubMed  Google Scholar 

  85. Ramracheya RD, McCulloch LJ, Clark A. PYY-dependent restoration of impaired insulin and glucagon secretion in type 2 diabetes following Roux-En-Y gastric bypass surgery. Cell Rep. 2016;15(5):944–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Alarcon C, Boland BB, Uchizono Y. Pancreatic beta-cell adaptive plasticity in obesity increases insulin production but adversely affects secretory function. Diabetes. 2016;65(2):438–50.

    Article  CAS  PubMed  Google Scholar 

  87. Martinussen C, Bojsen-Moller KN, Dirksen C. Immediate enhancement of first-phase insulin secretion and unchanged glucose effectiveness in patients with type 2 diabetes after Roux-en-Y gastric bypass. Am J Physiol Endocrinol Metab. 2015;308(6):E535–44.

    Article  CAS  PubMed  Google Scholar 

  88. Ehses JA, Perren A, Eppler E. Increased number of islet-associated macrophages in type 2 diabetes. Diabetes. 2007;56(9):2356–70.

    Article  CAS  PubMed  Google Scholar 

  89. Richardson SJ, Willcox A, Bone AJ. Islet-associated macrophages in type 2 diabetes. Diabetologia. 2009;52(8):1686–8.

    Article  CAS  PubMed  Google Scholar 

  90. Boni-Schnetzler M, Thorne J, Parnaud G. Increased interleukin (IL)-1beta messenger ribonucleic acid expression in beta -cells of individuals with type 2 diabetes and regulation of IL-1beta in human islets by glucose and autostimulation. J Clin Endocrinol Metab. 2008;93(10):4065–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Eguchi K, Manabe I, Oishi-Tanaka Y. Saturated fatty acid and TLR signaling link beta cell dysfunction and islet inflammation. Cell Metab. 2012;15(4):518–33.

    Article  CAS  PubMed  Google Scholar 

  92. Jones HB, Nugent D, Jenkins R. Variation in characteristics of islets of Langerhans in insulin-resistant, diabetic and non-diabetic-rat strains. Int J Exp Pathol. 2010;91(3):288–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Masters SL, Dunne A, Subramanian SL. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1beta in type 2 diabetes. Nat Immunol. 2010;11(10):897–904.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Youm YH, Adijiang A, Vandanmagsar B. Elimination of the NLRP3-ASC inflammasome protects against chronic obesity-induced pancreatic damage. Endocrinology. 2011;152(11):4039–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Nicol LE, Grant WF, Comstock SM. Pancreatic inflammation and increased islet macrophages in insulin-resistant juvenile primates. J Endocrinol. 2013;217(2):207–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Maedler K, Sergeev P, Ris F. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest. 2017;127(4):1589.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Cancello R, Rouault C, Guilhem G. Urokinase plasminogen activator receptor in adipose tissue macrophages of morbidly obese subjects. Obes Facts. 2011;4(1):17–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Dillard TH, Purnell JQ, Smith MD. Omentectomy added to Roux-en-Y gastric bypass surgery: a randomized, controlled trial. Surg Obes Relat Dis. 2013;9(2):269–75.

    Article  PubMed  Google Scholar 

  99. Moschen AR, Molnar C, Geiger S. Anti-inflammatory effects of excessive weight loss: potent suppression of adipose interleukin 6 and tumour necrosis factor alpha expression. Gut. 2010;59(9):1259–64.

    Article  CAS  PubMed  Google Scholar 

  100. Pardina E, Ferrer R, Baena-Fustegueras JA. Only C-reactive protein, but not TNF-alpha or IL6, reflects the improvement in inflammation after bariatric surgery. Obes Surg. 2012;22(1):131–9.

    Article  PubMed  Google Scholar 

  101. Haider DG, Schindler K, Prager G. Serum retinol-binding protein 4 is reduced after weight loss in morbidly obese subjects. J Clin Endocrinol Metab. 2007;92(3):1168–71.

    Article  CAS  PubMed  Google Scholar 

  102. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132(6):2131–57.

    Article  CAS  PubMed  Google Scholar 

  103. Ye J, Hao Z, Mumphrey MB. GLP-1 receptor signaling is not required for reduced body weight after RYGB in rodents. Am J Physiol Regul Integr Comp Physiol. 2014;306(5):R352–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Wilson-Perez HE, Chambers AP, Ryan KK. Vertical sleeve gastrectomy is effective in two genetic mouse models of glucagon-like Peptide 1 receptor deficiency. Diabetes. 2013;62(7):2380–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Bottcher G, Ahren B, Lundquist I. Peptide YY: intrapancreatic localization and effects on insulin and glucagon secretion in the mouse. Pancreas. 1989;4(3):282–8.

    Article  CAS  PubMed  Google Scholar 

  106. Upchurch BH, Aponte GW, Leiter AB. Expression of peptide YY in all four islet cell types in the developing mouse pancreas suggests a common peptide YY-producing progenitor. Development. 1994;120(2):245–52.

    CAS  PubMed  Google Scholar 

  107. Chandarana K, Batterham R. Curr Opin Endocrinol Diabetes Obes. 2008;15(1):65–72.

    Article  CAS  PubMed  Google Scholar 

  108. Boey D, Sainsbury A, Herzog H. The role of peptide YY in regulating glucose homeostasis. Peptides. 2007;28(2):390–5.

    Article  CAS  PubMed  Google Scholar 

  109. Sam AH, Gunner DJ, King A. Selective ablation of peptide YY cells in adult mice reveals their role in beta cell survival. Gastroenterology. 2012;143(2):459–68.

    Article  CAS  PubMed  Google Scholar 

  110. Shi YC, Loh K, Bensellam M. Pancreatic PYY is critical in the control of insulin secretion and glucose homeostasis in female mice. Endocrinology. 2015;156(9):3122–36.

    Article  CAS  PubMed  Google Scholar 

  111. Garibay D, Lou J, Lee SA. Beta cell GLP-1R signaling alters alpha cell proglucagon processing after vertical sleeve gastrectomy in mice. Cell Rep. 2018;23(4):967–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Mokadem M, Zechner JF, Margolskee RF. Effects of Roux-en-Y gastric bypass on energy and glucose homeostasis are preserved in two mouse models of functional glucagon-like peptide-1 deficiency. Mol Metab. 2014;3(2):191–201.

    Article  CAS  PubMed  Google Scholar 

  113. Sachdev S, Wang Q, Billington C. FGF 19 and bile acids increase following Roux-en-Y gastric bypass but not after medical management in patients with type 2 diabetes. Obes Surg. 2016;26(5):957–65.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Nemati R, Lu J, Dokpuang D. Increased bile acids and FGF19 after sleeve gastrectomy and Roux-en-Y gastric bypass correlate with improvement in type 2 diabetes in a randomized trial. Obes Surg. 2018;28(9):2672–86.

    Article  PubMed  Google Scholar 

  115. Kohli R, Kirby M, Setchell KD. Intestinal adaptation after ileal interposition surgery increases bile acid recycling and protects against obesity-related comorbidities. Am J Physiol Gastrointest Liver Physiol. 2010;299(3):G652–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Cariou B, van Harmelen K, Duran-Sandoval D. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J Biol Chem. 2006;281(16):11039–49.

    Article  CAS  PubMed  Google Scholar 

  117. Prawitt J, Abdelkarim M, Stroeve JH. Farnesoid X receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes. 2011;60(7):1861–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Morton GJ, Matsen ME, Bracy DP. FGF19 action in the brain induces insulin-independent glucose lowering. J Clin Invest. 2013;123(11):4799–808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Ryan KK, Kohli R, Gutierrez-Aguilar R. Fibroblast growth factor-19 action in the brain reduces food intake and body weight and improves glucose tolerance in male rats. Endocrinology. 2013;154(1):9–15.

    Article  CAS  PubMed  Google Scholar 

  120. Baud G, Daoudi M, Hubert T. Bile diversion in Roux-en-Y gastric bypass modulates sodium-dependent glucose intestinal uptake. Cell Metab. 2016;23(3):547–53.

    Article  CAS  PubMed  Google Scholar 

  121. Whalley NM, Pritchard LE, Smith DM. Processing of proglucagon to GLP-1 in pancreatic alpha-cells: is this a paracrine mechanism enabling GLP-1 to act on beta-cells? J Endocrinol. 2011;211(1):99–106.

    Article  CAS  PubMed  Google Scholar 

  122. Kumar DP, Rajagopal S, Mahavadi S. Activation of transmembrane bile acid receptor TGR5 stimulates insulin secretion in pancreatic beta cells. Biochem Biophys Res Commun. 2012;427(3):600–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Kumar DP, Asgharpour A, Mirshahi F. Activation of transmembrane bile acid receptor TGR5 modulates pancreatic islet alpha cells to promote glucose homeostasis. J Biol Chem. 2016;291(13):6626–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported by the National Key R&D Program of China (2016YFC1305000, 2016YFC1305001), the National Science and Technology Infrastructure Program (2015BAI12B13), the National Natural Science Foundation of China (81770775, 91749118, 81370017, 81130015, and 81000316), the Planned Science and Technology Project of Hunan Province (2017RS3015) and Natural Science Foundation of Hunan Province, China (14JJ3034), and the National Basic Research Program of China (2014CB910500).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Zhiguang Zhou.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflicts of interest.

Ethical Approval Statement

This article does not contain any studies with human participants or animals performed by any of the authors.

Informed Consent Statement

Does not apply.

Additional information

Publisher’s Note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, X., Zhang, J. & Zhou, Z. Targeting Islets: Metabolic Surgery Is More than a Bariatric Surgery. OBES SURG 29, 3001–3009 (2019). https://doi.org/10.1007/s11695-019-03979-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11695-019-03979-1

Keywords

Navigation