Skip to main content

Advertisement

Log in

The Gut as an Endocrine Organ: Role in the Regulation of Food Intake and Body Weight

  • Lipid and Metabolic Effects of Gastrointestinal Surgery (R. Cohen, Section Editor)
  • Published:
Current Atherosclerosis Reports Aims and scope Submit manuscript

Abstract

Abstract

Obesity and its related complications remain a major threat to public health. Efforts to reduce the prevalence of obesity are of paramount importance in improving population health. Through these efforts, our appreciation of the role of gut-derived hormones in the management of body weight has evolved and manipulation of this system serves as the basis for our most effective obesity interventions.

Purpose of the review

We review current understanding of the enteroendocrine regulation of food intake and body weight, focusing on therapies that have successfully embraced the physiology of this system to enable weight loss.

Recent findings

In addition to the role of gut hormones in the regulation of energy homeostasis, our understanding of the potential influence of enteroendocrine peptides in food reward pathways is evolving. So too is the role of gut derived hormones on energy expenditure.

Summary

Gut-derived hormones have the ability to alter feeding behavior. Certain obesity therapies already manipulate this system; however, our evolving understanding of the effects of enteroendocrine signals on hedonic aspects of feeding and energy expenditure may be crucial in identifying future obesity therapies.

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.

Similar content being viewed by others

References

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

  1. Calle EE, Thun MJ, Petrelli JM, Rodriguez C, Heath CW. Body-mass index and mortality in a prospective cohort of U.S. Adults. N Engl J Med. 1999;341(15).

  2. WHO. World Health Organisation Global Health Observatory Data; Overweight and Obesity 2014. Available from: http://www.who.int/gho/ncd/risk_factors/overweight/en/.

  3. Hahn TM, Breininger JF, Baskin DG, Schwartz MW. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nat Neurosci. 1998;1(4).

  4. Lin S, Boey D, Herzog H. NPY and Y receptors: lessons from transgenic and knockout models. Neuropeptides. 2004;38(4):189–200.

    Article  CAS  PubMed  Google Scholar 

  5. Fong TM, Mao C, MacNeil T, Kalyani R, Smith T, Weinberg D, et al. ART (protein product of agouti-related transcript) as an antagonist of MC-3 and MC-4 receptors. Biochem Biophys Res Commun. 1997;237(3):629–31.

    Article  CAS  PubMed  Google Scholar 

  6. Wu Q, Palmiter RD. GABAergic signaling by AgRP neurons prevents anorexia via a melanocortin-independent mechanism. Eur J Pharmacol. 2011;660(1):21–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Schwartz MW, Woods SC, Porte Jr D, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404:661–71.

    CAS  PubMed  Google Scholar 

  8. Alhadeff AL, Grill HJ. Hindbrain nucleus tractus solitarius glucagon-like peptide-1 receptor signaling reduces appetitive and motivational aspects of feeding. Am J Physiol Regul Integr Comp Physiol. 2014;307(4):R465–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Blouet C, Schwartz GJ. Brainstem nutrient sensing in the nucleus of the solitary tract inhibits feeding. Cell Metab. 2012;16(5):579–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ter Horst GJ, de Boer P, Luiten PG, van Willigen JD. Ascending projections from the solitary tract nucleus to the hypothalamus. A Phaseolus vulgaris lectin tracing study in the rat. Neuroscience. 1989;31(3):785–97.

    Article  PubMed  Google Scholar 

  11. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762):656–60.

    Article  CAS  PubMed  Google Scholar 

  12. Romero A, Kirchner H, Heppner K, Pfluger PT, Tschop MH, Nogueiras R. GOAT: the master switch for the ghrelin system? Eur J Endocrinol. 2010;163(1):1–8.

    Article  CAS  PubMed  Google Scholar 

  13. Date Y, Kojima M, Hosoda H, Sawaguchi A, Mondal MS, Suganuma T, et al. Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology. 2000;141(11):4255–61.

    CAS  PubMed  Google Scholar 

  14. Cummings DE, Purnell JQ, Scott Frayo R, Schmidova K, Wisse BE, Weigle DS. A Preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes. 2001;50.

  15. Masamitsu Nakazato NM, Yukari Date, Masayasu Kojima, Hisayuki Matsuo, Kenji Kangawa & Shigeru Matsukura. A role for ghrelin in the central regulation of feeding. Nature. 2001;409.

  16. Callahan HS, Cummings DE, Pepe MS, Breen PA, Matthys CC, Weigle DS. Postprandial suppression of plasma ghrelin level is proportional to ingested caloric load but does not predict intermeal interval in humans. J Clin Endocrinol Metab. 2004;89(3):1319–24.

    Article  CAS  PubMed  Google Scholar 

  17. Weigle DS, Cummings DE, Newby PD, Breen PA, Frayo RS, Matthys CC, et al. Roles of leptin and ghrelin in the loss of body weight caused by a low fat, high carbohydrate diet. J Clin Endocrinol Metab. 2003;88(4):1577–86.

    Article  CAS  PubMed  Google Scholar 

  18. Shiya T, Nakazato M, Mizuta M, Date Y, Mondal MS, Tanaka M, et al. Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J Clin Endocrinol Metab. 2002;87(1):240–4.

    Article  Google Scholar 

  19. Banks WA, Tschop M, Robinson SM, Heiman ML. Extent and direction of ghrelin transport across the blood–brain barrier is determined by its unique primary structure. J Pharmacol Exp Ther. 2002;302(2):822–7.

    Article  CAS  PubMed  Google Scholar 

  20. Willesen MG, Kristensen P, Romer J. Co-localization of growth hormone secretagogue receptor and NPY mRNA in the arcuate nucleus of the rat. Neuroendocrinology. 1999;70(5):306–16.

    Article  CAS  PubMed  Google Scholar 

  21. Shintani M, Ogawa Y, Ebihara K, Aizawa-Abe M, Miyanaga F, Takaya K, et al. Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes. 2001;5.

  22. Wren AM, Seal LJ, Brynes AE, Frost GS, Murphy KG, Dhillo WS, et al. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab. 2001;86(12):5992–5.

    Article  CAS  PubMed  Google Scholar 

  23. Cummings DE, Weigle DS, Scott Frayo R, Breen PA, Ma MK, Dellinger P, et al. Plasma Ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346(21).

  24. Zigman JM, Jones JE, Lee CE, Saper CB, Elmquist JK. Expression of ghrelin receptor mRNA in the rat and the mouse brain. J Comp Neurol. 2006;494(3):528–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Abizaid A, Liu ZW, Andrews ZB, Shanabrough M, Borok E, Elsworth JD, et al. Ghrelin modulates the activity and synaptic input organization of midbrain dopamine neurons while promoting appetite. J Clin Invest. 2006;116(12):3229–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Perello M, Sakata I, Birnbaum S, Chuang JC, Osborne-Lawrence S, Rovinsky SA, et al. Ghrelin increases the rewarding value of high-fat diet in an orexin-dependent manner. Biol Psychiatry. 2010;67(9):880–6.

    Article  CAS  PubMed  Google Scholar 

  27. Nilsson O, Bilchik AJ, Goldenring JR, Ballantyne GH, Adrian TE, Modlin IM. Distribution and immunocytochemical colocalization of peptide YY and enteroglucagon in endocrine cells of the rabbit colon. Endocrinology. 1991;129(1):139–48.

    Article  CAS  PubMed  Google Scholar 

  28. Tatemoto K, Nakano I, Makk G, Angwin P, Mann M, Schilling J, et al. Isolation and primary structure of human peptide YY. Biochem Biophys Res Commun. 1988;157(2):713–7.

    Article  CAS  PubMed  Google Scholar 

  29. Essah PA, Levy JR, Sistrun SN, Kelly SM, Nestler JE. Effect of macronutrient composition on postprandial peptide YY levels. J Clin Endocrinol Metab. 2007;92(10):4052–5.

    Article  CAS  PubMed  Google Scholar 

  30. Grandt D, Schimiczek M, Beglinger C, Layer P, Goebell H, Eysselein VE, et al. Two molecular forms of peptide YY (PYY) are abundant in human blood: characterization of a radioimmunoassay recognizing PYY 1–36 and PYY 3–36. Regul Pept. 1994;51(2):151–9.

    Article  CAS  PubMed  Google Scholar 

  31. Grandt D, Dahms P, Schimiczek M, Eysselein VE, Reeve Jr JR, Mentlein R. [Proteolytic processing by dipeptidyl aminopeptidase IV generates receptor selectivity for peptide YY (PYY)]. Med Klin (Munich). 1993;88(3):143–5.

    CAS  Google Scholar 

  32. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, et al. Inhibition of food intake in obese subjects by peptide YY3–36. N Engl J Med. 2003;349(10):941–8.

    Article  CAS  PubMed  Google Scholar 

  33. Nonaka N, Shioda S, Niehoff ML, Banks WA. Characterization of blood–brain barrier permeability to PYY3-36 in the mouse. J Pharmacol Exp Ther. 2003;306(3):948–53.

    Article  CAS  PubMed  Google Scholar 

  34. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, et al. Gut hormone PYY(3–36) physiologically inhibits food intake. Nature. 2002;418(6898):650–4.

    Article  CAS  PubMed  Google Scholar 

  35. Shi YC, Lin Z, Lau J, Zhang H, Yagi M, Kanzler I, et al. PYY3-36 and pancreatic polypeptide reduce food intake in an additive manner via distinct hypothalamic dependent pathways in mice. Obesity (Silver Spring). 2013;21(12):E669–78.

    Article  CAS  Google Scholar 

  36. Stanic A, Brumovsky P, Fetissov S, Shuster S, Herzog H, Hokfelt T. Characterization of neuropeptide Y2 receptor protein expression in the mouse brain. I. Distribution in cell bodies and nerve terminals. J Comp Neurol. 2006;499:357–90.

    Article  CAS  PubMed  Google Scholar 

  37. Halatchev IG, Ellacott KL, Fan W, Cone RD. Peptide YY3-36 inhibits food intake in mice through a melanocortin-4 receptor-independent mechanism. Endocrinology. 2004;145(6):2585–90.

    Article  CAS  PubMed  Google Scholar 

  38. Batterham RL. ffytche DH, Rosenthal JM, Zelaya FO, Barker GJ, Withers DJ, et al. PYY modulation of cortical and hypothalamic brain areas predicts feeding behaviour in humans. Nature. 2007;450(7166):106–9.

    Article  CAS  PubMed  Google Scholar 

  39. Orskov C, Bersani M, Johnsen AH, Hajrup P, Holst JJ. Complete sequences of glucagon-like peptide-1 from human and pig small intestine. J Biol Chem. 1989;264(22).

  40. Damholt AB, Buchan AMJ, Holst JJ, Kofod H. Proglucagon processing profile in canine L cells expressing endogenous prohormone convertase 1/3 and prohormone convertase 2. Endocrinology. 1999;140(10):4800–8.

    Article  CAS  PubMed  Google Scholar 

  41. Orskov C, Rabenhoj L, Wettergren A, Kofod H, Hoist JJ. Tissue and plasma concentrations of amidated and glycine-extended glucagon-like peptide I in humans. Diabetes. 1994;43:535–9.

    Article  CAS  PubMed  Google Scholar 

  42. Mentlein R, B. G, W.E. S. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-likepeptide-l(7–36)amide, peptidehistidinemethionine and is responsible for their degradation in human serum. Eur J Biochem. 1993;214:826–835).

  43. Kreymann B, Ghatei MA, Williams G, Bloom SR. Glucagon-like peptide-1 7–36: a physiological incretin in man. Lancet. 1987;330(8571):1300–4.

    Article  Google Scholar 

  44. Orskov C, Poulsen SS, Moller M, Hoist JJ. Glucagon-like peptide I receptors in the subfornical organ and the area postrema are accessible to circulating glucagon-like peptide I. Diabetes. 1996;45:832–5.

    Article  CAS  PubMed  Google Scholar 

  45. Flint A, Raben A, Astrup A, Holst JJ. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Investig. 1998;101(3):515–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Näslund E, Gutniak M, Skogar S, Rössner S, Hellström PM. Glucagon-like peptide 1 increases the period of postprandial satiety and slows gastric emptying in obese men. Am J Clin Nutr. 1998;68:525–30.

    PubMed  Google Scholar 

  47. Shughrue PJ, Lane MV, Merchenthaler I. Glucagon-like peptide-1 receptor (GLP1-R) mRNA in the rat hypothalamus. Endocrinology. 1996;137(11):5159–62.

    CAS  PubMed  Google Scholar 

  48. McMahon LR, Wellman PJ. PVN infusion of GLP-1-(7—36) amide suppresses feeding but does not induce aversion or alter locomotion in rats. 1997.

  49. Turton MD, O'Shea D, Gunn I, Beak SA, Edwards CMB, Meeran K, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379(6560):69–72.

    Article  CAS  PubMed  Google Scholar 

  50. Dossat AM, Lilly N, Kay K, Williams DL. Glucagon-like peptide 1 receptors in nucleus accumbens affect food intake. J Neurosci. 2011;31(41):14453–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dickson SL, Shirazi RH, Hansson C, Bergquist F, Nissbrandt H, Skibicka KP. The glucagon-like peptide 1 (GLP-1) analogue, exendin-4, decreases the rewarding value of food: a new role for mesolimbic GLP-1 receptors. J Neurosci. 2012;32(14):4812–20.

    Article  CAS  PubMed  Google Scholar 

  52. van Bloemendaal L, IJzerman RG, ten Kulve JS, Barkhof F, Konrad RJ, Drent ML, et al. GLP-1 receptor activation modulates appetite- and reward-related brain areas in humans. Diabetes. 2014;63:4186–96.

    Article  PubMed  Google Scholar 

  53. ten Kulve JS, Veltman DJ, van Bloemendaal L, Barkhof F, Deacon CF, Holst JJ, et al. Endogenous GLP-1 mediates postprandial reductions in activation in central reward and satiety areas in patients with type 2 diabetes. Diabetologia. 2015;58(12):2688–98.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Munck A, Kervran A, Marie JC, Bataille D, Rosselin G. Glucagon-37 (oxyntomodulin) and glucagon-29 (pancreatic glucagon) in human bowel: analysis by HPLC and radioreceptorassay. Peptides. 1984;5(3):553–61.

    Article  CAS  PubMed  Google Scholar 

  55. Holst JJ. Enteroglucagon. Annu Rev Physiol. 1997;59:257–71.

    Article  CAS  PubMed  Google Scholar 

  56. Zhu L, Tamvakopoulos C, Xie D, Dragovic J, Shen X, Fenyk-Melody JE, et al. The role of dipeptidyl peptidase IV in the cleavage of glucagon family peptides: in vivo metabolism of pituitary adenylate cyclase activating polypeptide-(1–38). J Biol Chem. 2003;278(25):22418–23.

    Article  CAS  PubMed  Google Scholar 

  57. Schjoldager B, Mortensen PE, Myhre J, Christiansen J, Holst JJ. Oxyntomodulin from distal gut. Role in regulation of gastric and pancreatic functions. Dig Dis Sci. 1989;34(9):1411–9.

    Article  CAS  PubMed  Google Scholar 

  58. Gros L, Thorens B, Bataille D, Kervran A. Glucagon-like peptide-1-(7–36) amide, oxyntomodulin, and glucagon interact with a common receptor in a somatostatin-secreting cell line. Endocrinology. 1993;133(2):631–8.

    CAS  PubMed  Google Scholar 

  59. Dakin CL, Gunn I, Small CJ, Edwards CMB, Hay DL, Smith DM, et al. Oxyntomodulin inhibits food intake in the rat. Endocrinology. 2001;142(10):4244–50.

    Article  CAS  PubMed  Google Scholar 

  60. Baggio LL, Huang Q, Brown TJ, Drucker DJ. Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure. Gastroenterology. 2004;127(2):546–58.

    Article  CAS  PubMed  Google Scholar 

  61. Parkinson JR, Chaudhri OB, Kuo YT, Field BC, Herlihy AH, Dhillo WS, et al. Differential patterns of neuronal activation in the brainstem and hypothalamus following peripheral injection of GLP-1, oxyntomodulin and lithium chloride in mice detected by manganese-enhanced magnetic resonance imaging (MEMRI). Neuroimage. 2009;44(3):1022–31.

    Article  PubMed  Google Scholar 

  62. Kosinski JR, Hubert J, Carrington PE, Chicchi GG, Mu J, Miller C, et al. The glucagon receptor is involved in mediating the body weight-lowering effects of oxyntomodulin. Obesity (Silver Spring). 2012;20(8):1566–71.

    Article  CAS  Google Scholar 

  63. Wynne K, Park AJ, Small CJ, Meeran K, Ghatei MA, Frost GS, et al. Oxyntomodulin increases energy expenditure in addition to decreasing energy intake in overweight and obese humans: a randomised controlled trial. Int J Obes (Lond). 2006;30(12):1729–36.

    Article  CAS  Google Scholar 

  64. Nakagawa A, Satake H, Nakabayashi H, Nishizawa M, Furuya K, Nakano S, et al. Receptor gene expression of glucagon-like peptide-1, but not glucose-dependent insulinotropic polypeptide, in rat nodose ganglion cells. Auton Neurosci. 2004;110(1):36–43.

    Article  CAS  PubMed  Google Scholar 

  65. Koda S, Date Y, Murakami N, Shimbara T, Hanada T, Toshinai K, et al. The role of the vagal nerve in peripheral PYY3-36-induced feeding reduction in rats. Endocrinology. 2005;146(5):2369–75.

    Article  CAS  PubMed  Google Scholar 

  66. Abbott CR, Monteiro M, Small CJ, Sajedi A, Smith KL, Parkinson JR, et al. The inhibitory effects of peripheral administration of peptide YY(3–36) and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res. 2005;1044(1):127–31.

    Article  CAS  PubMed  Google Scholar 

  67. Ruttimann EB, Arnold M, Hillebrand JJ, Geary N, Langhans W. Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology. 2009;150(3):1174–81.

    Article  CAS  PubMed  Google Scholar 

  68. le Roux CW, Neary NM, Halsey TJ, Small CJ, Martinez-Isla AM, Ghatei MA, et al. Ghrelin does not stimulate food intake in patients with surgical procedures involving vagotomy. J Clin Endocrinol Metab. 2005;90(8):4521–4.

    Article  PubMed  Google Scholar 

  69. Arnold M, Mura A, Langhans W, Geary N. Gut vagal afferents are not necessary for the eating-stimulatory effect of intraperitoneally injected ghrelin in the rat. J Neurosci. 2006;26(43):11052–60.

    Article  CAS  PubMed  Google Scholar 

  70. Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H, et al. The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats. Gastroenterology. 2002;123(4):1120–8.

    Article  CAS  PubMed  Google Scholar 

  71. Zhang JH, Nolan JD, Kennie SL, Johnston IM, Dew T, Dixon PH, et al. Potent stimulation of fibroblast growth factor 19 expression in the human ileum by bile acids. Am J Physiol Gastrointest Liver Physiol. 2013;304(10):G940–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Ma Y, Huang Y, Yan L, Gao M, Liu D. Synthetic FXR agonist GW4064 prevents diet-induced hepatic steatosis and insulin resistance. Pharm Res. 2013;30(5):1447–57.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Fang S, Suh JM, Reilly SM, Yu E, Osborn O, Lackey D, et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat Med. 2015;21(2):159–65. This paper reports on increased energy expenditure mediated through intestinal FXR activation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, et al. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med. 2013;369(2):145–54.

    Article  CAS  PubMed  Google Scholar 

  75. Sumithran P, Prendergast LA, Delbridge E, Purcell K, Shulkes A, Kriketos A, et al. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365(17):1597–604.

    Article  CAS  PubMed  Google Scholar 

  76. Torsten Olbers HL. Monika Fegervik-Olsen and Lars Lundell. Laparoscopic gastric bypass: development of technique, respiratory function, and long-term outcome. Obes Surg. 2003;13:364–70.

    Article  PubMed  Google Scholar 

  77. Carswell KA, Vincent RP, Belgaumkar AP, Sherwood RA, Amiel SA, Patel AG, et al. The effect of bariatric surgery on intestinal absorption and transit time. Obes Surg. 2014;24(5):796–805.

    Article  PubMed  Google Scholar 

  78. Marmuse PMDCJ-P. Laparoscopic sleeve gastrectomy as an initial bariatric operation for high-risk patients: initial results in 10 patients. Obes Surg. 2005;15:1030–3.

    Article  PubMed  Google Scholar 

  79. Melissas J, Leventi A, Klinaki I, Perisinakis K, Koukouraki S, de Bree E, et al. Alterations of global gastrointestinal motility after sleeve gastrectomy: a prospective study. Ann Surg. 2013;258(6):976–82.

    Article  PubMed  Google Scholar 

  80. Sjöström L, Lindroos A, Peltonen M, Torgerson J, Bouchard C, Carlsson B, et al. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351:2683–93.

    Article  PubMed  Google Scholar 

  81. le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A, Laurenius A, et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246(5):780–5.

    Article  PubMed  Google Scholar 

  82. le Roux CW, Aylwin SJB, Batterham RL, Borg CM, Coyle F, Prasad V, et al. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg. 2006;243(1):108–14.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Laferrere B, Swerdlow N, Bawa B, Arias S, Bose M, Olivan B, et al. Rise of oxyntomodulin in response to oral glucose after gastric bypass surgery in patients with type 2 diabetes. J Clin Endocrinol Metab. 2010;95(8):4072–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Yousseif A, Emmanuel J, Karra E, Millet Q, Elkalaawy M, Jenkinson AD, et al. Differential effects of laparoscopic sleeve gastrectomy and laparoscopic gastric bypass on appetite, circulating acyl-ghrelin, peptide YY3-36 and active GLP-1 levels in non-diabetic humans. Obes Surg. 2014;24(2):241–52.

    Article  PubMed  Google Scholar 

  85. Korner J, Bessler M, Cirilo LJ, Conwell IM, Daud A, Restuccia NL, et al. Effects of Roux-en-Y gastric bypass surgery on fasting and postprandial concentrations of plasma ghrelin, peptide YY, and insulin. J Clin Endocrinol Metab. 2005;90(1):359–65.

    Article  CAS  PubMed  Google Scholar 

  86. Dimitriadis E, Daskalakis M, Kampa M, Peppe A, Papadakis JA, Melissas J. Alterations in gut hormones after laparoscopic sleeve gastrectomy: a prospective clinical and laboratory investigational study. Ann Surg. 2013;257(4):647–54.

    Article  PubMed  Google Scholar 

  87. Karamanakos SN, Vagenas K, Kalfarentzos F, Alexandrides TK. 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 

  88. Scholtz S, Miras AD, Chhina N, Prechtl CG, Sleeth ML, Daud NM, et al. Obese patients after gastric bypass surgery have lower brain-hedonic responses to food than after gastric banding. Gut. 2014;63(6):891–902. This paper demonstrates decreased neural food reward pathways following RYGB surgery.

  89. Goldstone AP, Miras AD, Scholtz S, Jackson S, Neff KJ, Penicaud L, et al. Link between increased satiety gut hormones and reduced food reward after gastric bypass surgery for obesity. J Clin Endocrinol Metab. 2016;101(2):599–609. This paper demonstrates decreased activity in neural food reward pathways following RYGB surgery.

  90. Werling M, Fandriks L, Olbers T, Bueter M, Sjostrom L, Lonroth H, et al. Roux-en-Y gastric bypass surgery increases respiratory quotient and energy expenditure during food intake. PLoS One. 2015;10(6):e0129784.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Pournaras DJ, Glicksman C, Vincent RP, Kuganolipava S, Alaghband-Zadeh J, Mahon D, et al. The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology. 2012;153(8):3613–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Maruyama T, Miyamoto Y, Nakamura T, Tamai Y, Okada H, Sugiyama E, et al. Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun. 2002;298:714–9.

    Article  CAS  PubMed  Google Scholar 

  93. Wu T, Bound MJ, Standfield SD, Gedulin B, Jones KL, Horowitz M, et al. Effects of rectal administration of taurocholic acid on glucagon-like peptide-1 and peptide YY secretion in healthy humans. Diabetes Obes Metab. 2013;15(5):474–7.

    Article  CAS  PubMed  Google Scholar 

  94. Watanabe M, Houten SM, Mataki C, Christoffolete MA, Kim BW, Sato H, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 2006;439(7075):484–9.

    Article  CAS  PubMed  Google Scholar 

  95. Sachdev S, Wang Q, Billington C, Connett J, Ahmed L, Inabnet W, et al. 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. 2015;26(5):957–65.

    Article  Google Scholar 

  96. Nauck M, Frid A, Hermansen K, Shah NS, Tankova T, Mitha IH, et al. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care. 2009;32(1):84–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373(1):11–22.

    Article  PubMed  Google Scholar 

  98. Secher A, Jelsing J, Baquero AF, Hecksher-Sorensen J, Cowley MA, Dalboge LS, et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest. 2014;124(10):4473–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Farr OM, Sofopoulos M, Tsoukas MA, Dincer F, Thakkar B, Sahin-Efe A, et al. GLP-1 receptors exist in the parietal cortex, hypothalamus and medulla of human brains and the GLP-1 analogue liraglutide alters brain activity related to highly desirable food cues in individuals with diabetes: a crossover, randomised, placebo-controlled trial. Diabetologia. 2016;59(5):945–65.

    Article  Google Scholar 

  100. Fishman E, Melanson D, Lamport R, Levine A. A novel endoscopic delivery system for placement of a duodenal-jejunal implant for the treatment of obesity and type 2 diabetes. Conf Proc IEEE Eng Med Biol Soc. 2008;2008:2501–3.

    PubMed  Google Scholar 

  101. Schouten R, Rijs CS, Bouvy ND, Hameeteman W, Koek GH, Janssen IM, et al. A multicenter, randomized efficacy study of the EndoBarrier Gastrointestinal Liner for presurgical weight loss prior to bariatric surgery. Ann Surg. 2010;251(2):236–43.

    Article  PubMed  Google Scholar 

  102. Gersin KS, Rothstein RI, Rosenthal RJ, Stefanidis D, Deal SE, Kuwada TS, et al. Open-label, sham-controlled trial of an endoscopic duodenojejunal bypass liner for preoperative weight loss in bariatric surgery candidates. Gastrointest Endosc. 2010;71(6):976–82.

    Article  PubMed  Google Scholar 

  103. Rohde U, Hedback N, Gluud LL, Vilsboll T, Knop FK. Effect of the EndoBarrier Gastrointestinal Liner on obesity and type 2 diabetes: a systematic review and meta-analysis. Diabetes Obes Metab. 2016;18(3):300–5.

    Article  CAS  PubMed  Google Scholar 

  104. de Jonge C, Rensen SS, Verdam FJ, Vincent RP, Bloom SR, Buurman WA, et al. Endoscopic duodenal-jejunal bypass liner rapidly improves type 2 diabetes. Obes Surg. 2013;23(9):1354–60.

    Article  PubMed  Google Scholar 

  105. de Jonge C, Rensen SS, Verdam FJ, Vincent RP, Bloom SR, Buurman WA, et al. Impact of duodenal-jejunal exclusion on satiety hormones. Obes Surg. 2016;26(3):672–8.

    Article  PubMed  Google Scholar 

  106. Habegger KM, Al-Massadi O, Heppner KM, Myronovych A, Holland J, Berger J, et al. Duodenal nutrient exclusion improves metabolic syndrome and stimulates villus hyperplasia. Gut. 2014;63(8):1238–46.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Neil G. Docherty.

Ethics declarations

Conflict of Interest

Audrey Melvin and Neil G. Docherty declare that they have no conflict of interest.

Carel W. le Roux declares personal fees from NovoNordisk, Herbalife and Henry Stewart Talks for education in Obesity; honoraria and travel fees for being an invited speaker at conferences; and money paid to his institution from Science Foundation Ireland, from patents, royalties, for payment for manuscript preparation, and from stocks.

Human and Animal Rights and Informed Consent

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

Additional information

This article is part of the Topical Collection on Lipid and Metabolic Effects of Gastrointestinal Surgery

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Melvin, A., le Roux, C.W. & Docherty, N.G. The Gut as an Endocrine Organ: Role in the Regulation of Food Intake and Body Weight. Curr Atheroscler Rep 18, 49 (2016). https://doi.org/10.1007/s11883-016-0599-9

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11883-016-0599-9

Keywords

Navigation