Mechanisms underlying the weight loss effects of RYGB and SG: similar, yet different

A Pucci, R L Batterham, A Pucci, R L Batterham

Abstract

The worldwide obesity epidemic continues unabated, adversely impacting upon global health and economies. People with severe obesity suffer the greatest adverse health consequences with reduced life expectancy. Currently, bariatric surgery is the most effective treatment for people with severe obesity, resulting in marked sustained weight loss, improved obesity-associated comorbidities and reduced mortality. Sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB), the most common bariatric procedures undertaken globally, engender weight loss and metabolic improvements by mechanisms other than restriction and malabsorption. It is now clear that a plethora of gastrointestinal (GI) tract-derived signals plays a critical role in energy and glucose regulation. SG and RYGB, which alter GI anatomy and nutrient flow, impact upon these GI signals ultimately leading to weight loss and metabolic improvements. However, whilst highly effective overall, at individual level, post-operative outcomes are highly variable, with a proportion of patients experiencing poor long-term weight loss outcome and gaining little health benefit. RYGB and SG are markedly different anatomically and thus differentially impact upon GI signalling and bodyweight regulation. Here, we review the mechanisms proposed to cause weight loss following RYGB and SG. We highlight similarities and differences between these two procedures with a focus on gut hormones, bile acids and gut microbiota. A greater understanding of these procedure-related mechanisms will allow surgical procedure choice to be tailored to the individual to maximise post-surgery health outcomes and will facilitate the discovery of non-surgical treatments for people with obesity.

Keywords: Bile acids; Gut hormones; Gut microbiota; Roux-en-Y gastric bypass; Sleeve gastrectomy; Type 2 diabetes.

Conflict of interest statement

Conflict of interest

All authors declare that they have no conflict of interest.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Informed consent

Informed consent was obtained from all individual participants included in the studies performed by the authors.

Figures

Fig. 1
Fig. 1
Schematic diagram illustrating the normal upper GI anatomy (a) and the two most commonly performed bariatric surgical procedures in the world with relative percentages. The metabolic procedures: b RYGB and c SG (surgical technique described in details in the main text). RYGB were the 39.6% and SG were the 45.9% of the total procedures performed in 2014 [12]. RYGB Roux-en-Y gastric bypass, SG Sleeve gastrectomy
Fig. 2
Fig. 2
Schematic diagram illustrating the mechanisms involved in regulating feeding behaviour. Nutrient entry into the GI tract causes stomach and intestine distension, secretion of pancreatic enzymes and BA, altered enteric and vagal nerve signalling and exposure of gut enteroendocrine cells to nutrients with altered circulating gut hormone levels (e.g. decrease in orexigenic hormone ghrelin and increase in anorectic hormones PYY3-36 and GLP-1). Gut-derived signals (nutrients, hormones, and neural) and adipokines (e.g. leptin, IL-6, TNF-alpha and adiponectin) act directly and indirectly upon brainstem and hypothalamic arcuate nuclei (first order neurons: orexigenic NPY/AgRP and anorexigenic POMC/CART). ARC neurons interact with second order neurons in the PVN, and to the LHA. All those mechanisms are involved in the regulation of homeostatic hunger. Social factors, emotion, reward, pleasure, increased food availability and sensory cues can influence brain reward and higher cognitive brain regions leading to altered feeding behaviour (hedonic hunger). Taste and olfactory signals can also influence energy intake acting on both homeostatic and brain reward systems. Insulin leptin, GLP-1, PYY and ghrelin are present in saliva with cognate receptors on taste buds and olfactory neurons. AgRP agouti-related peptide, ARC arcuate nucleus, CART cocaine and amphetamine-regulated transcript, FGF-19 fibroblast growth factor-19, GLP-1 glucagon-like peptide 1, IL-6 interleukin-6, LHA lateral hypothalamic area, NPY neuropeptide Y, PNS peripheral nervous system, PVN paraventricular nucleus, PYY3-36 peptide tyrosine–tyrosine 3-36, POMC pro-opiomelanocortin, SNS sympathetic nervous system
Fig. 3
Fig. 3
Schematic diagram illustrating the different biological changes induced by weight loss obtained through dieting (upper part) compared to bariatric/metabolic surgery (lower part). Powerful compensatory biological changes contribute to the high rate of weight recidivism observed following lifestyle intervention weight management. Many homeostatic mechanisms act to defend higher body weight, and these includes hormonal alterations and a decreased energy expenditure leading to increased hunger and energy consumption. In contrast, bariatric surgery leads to a favourable biology that includes increased satiety hormones, reduced ghrelin, enhanced BA secretion and a “lean” microbiota. Together these mechanisms lead to reduced hunger and a shift towards healthier food options with a resetting of body weight “set point” to a lower level facilitating meaningful and sustained weight loss. GLP-1 glucagon-like peptide 1, PYY3-36 peptide tyrosine–tyrosine 3-36. *Suggestion that leptin sensitivity may improve References for this figure [5, 60, 96]
Fig. 4
Fig. 4
Schematic diagram illustrating RYGB and SG and the mechanisms leading to weight loss and resolution of comorbidities. For every mechanism the effect of the procedure is represented with a “↑” when stimulating or “↓” when suppressing. A “+” means that the proposed mechanism is present only after surgery when compared to the pre-operative period. When the effect is stronger for one of the two procedures there is a double arrow compared with a single one. When the effect is missing for one procedure it means that the mechanism is procedure specific. RYGB Roux-en-Y gastric bypass, SG Sleeve gastrectomy, GLP-1 glucagon-like peptide 1, PYY3-36 peptide tyrosine–tyrosine 3-36, GIP gastric inhibitory polypeptide, FGF-19 fibroblast growth factor-19, CCK cholecystokinin

References

    1. WHO Global health observatory (GHO) data 2016 Available from:
    1. Gloy VL, Briel M, Bhatt DL, et al. Bariatric surgery versus non-surgical treatment for obesity: a systematic review and meta-analysis of randomised controlled trials. BMJ. 2013;347:f5934.
    1. Makaronidis JM, Batterham RL. Potential mechanisms mediating sustained weight loss following Roux-en-Y gastric bypass and sleeve gastrectomy. Endocrinol Metab Clin North Am. 2016;45(3):539–552.
    1. Schauer PR, Bhatt DL, Kirwan JP, et al. Bariatric surgery versus intensive medical therapy for diabetes—5-year outcomes. N Engl J Med. 2017;376(7):641–651.
    1. Sjostrom L. Review of the key results from the Swedish Obese Subjects (SOS) trial—a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273(3):219–234.
    1. Radaelli MG, Martucci F, Perra S, et al. NAFLD/NASH in patients with type 2 diabetes and related treatment options. J Endocrinol Invest. 2017
    1. Saber AA, Elgamal MH, McLeod MK. Bariatric surgery: the past, present, and future. Obes Surg. 2008;18(1):121–128.
    1. Payne JH, Dewind LT, Commons RR. Metabolic observations in patients with jejunocolic shunts. Am J Surg. 1963;106:273–289.
    1. Sherman CD, Jr, May AG, Nye W, et al. Clinical and metabolic studies following bowel by-passing for obesity. Ann N Y Acad Sci. 1965;131(1):614–622.
    1. Buchwald H. The evolution of metabolic/bariatric surgery. Obes Surg. 2014;24(8):1126–1135.
    1. Mason EE, Ito C. Gastric bypass in obesity. Surg Clin N Am. 1967;47(6):1345–1351.
    1. Angrisani L, Santonicola A, Iovino P, et al. Bariatric surgery and endoluminal procedures: IFSO worldwide survey 2014. Obes Surg. 2017;27(9):2279–2289.
    1. Manning S, Pucci A, Carter NC, et al. Early postoperative weight loss predicts maximal weight loss after sleeve gastrectomy and Roux-en-Y gastric bypass. Surg Endosc. 2015;29(6):1484–1491.
    1. Pucci A, Tymoszuk U, Cheung WH, et al. Type 2 diabetes remission 2 years post Roux-en-Y gastric bypass and sleeve gastrectomy: the role of the weight loss and comparison of DiaRem and DiaBetter scores. Diabet Med J Br Diabet Assoc. 2018;35(3):360–367.
    1. Olbers T, Lonroth H, Fagevik-Olsen M, et al. Laparoscopic gastric bypass: development of technique, respiratory function, and long-term outcome. Obes Surg. 2003;13(3):364–370.
    1. Abu-Jaish W, Rosenthal RJ. Sleeve gastrectomy: a new surgical approach for morbid obesity. Expert Rev Gastroenterol Hepatol. 2010;4(1):101–119.
    1. Halperin F, Ding SA, Simonson DC, et al. Roux-en-Y gastric bypass surgery or lifestyle with intensive medical management in patients with type 2 diabetes: feasibility and 1-year results of a randomized clinical trial. JAMA Surg. 2014;149(7):716–726.
    1. Courcoulas AP, Belle SH, Neiberg RH, et al. Three-year outcomes of bariatric surgery vs lifestyle intervention for type 2 diabetes mellitus treatment: a randomized clinical trial. JAMA Surg. 2015;150(10):931–940.
    1. Schauer PR, Bhatt DL, Kirwan JP, et al. Bariatric surgery versus intensive medical therapy for diabetes–3-year outcomes. N Engl J Med. 2014;370(21):2002–2013.
    1. Peterli R, Wolnerhanssen BK, Peters T, et al. Effect of laparoscopic sleeve gastrectomy vs laparoscopic Roux-en-Y gastric bypass on weight loss in patients with morbid obesity: the SM-BOSS randomized clinical trial. JAMA. 2018;319(3):255–265.
    1. Salminen P, Helmio M, Ovaska J, et al. Effect of laparoscopic sleeve gastrectomy vs laparoscopic Roux-en-Y Gastric bypass on weight loss at 5 years among patients with morbid obesity: the SLEEVEPASS randomized clinical trial. JAMA. 2018;319(3):241–254.
    1. Ignat M, Vix M, Imad I, et al. Randomized trial of Roux-en-Y gastric bypass versus sleeve gastrectomy in achieving excess weight loss. Br J Surg. 2017;104(3):248–256.
    1. Osland E, Yunus RM, Khan S, et al. Weight loss outcomes in laparoscopic vertical sleeve gastrectomy (LVSG) versus laparoscopic Roux-en-Y gastric bypass (LRYGB) procedures: a meta-analysis and systematic review of randomized controlled trials. Surg Laparosc Endosc Percutaneous Tech. 2017;27(1):8–18.
    1. Kashyap SR, Bhatt DL, Wolski K, et al. Metabolic effects of bariatric surgery in patients with moderate obesity and type 2 diabetes: analysis of a randomized control trial comparing surgery with intensive medical treatment. Diabetes Care. 2013;36(8):2175–2182.
    1. Miras AD, le Roux CW. Mechanisms underlying weight loss after bariatric surgery. Nat Rev Gastroenterol Hepatol. 2013;10(10):575–584.
    1. Makaronidis JM, Batterham RL. Obesity, body weight regulation and the brain: insights from fMRI. Br J Radiol. 2018
    1. Scott WR, Batterham RL. Roux-en-Y gastric bypass and laparoscopic sleeve gastrectomy: understanding weight loss and improvements in type 2 diabetes after bariatric surgery. Am J Physiol Regul Integr Comp Physiol. 2011;301(1):R15–R27.
    1. Batterham RL, Cowley MA, Small CJ, et al. Gut hormone PYY(3-36) physiologically inhibits food intake. Nature. 2002;418(6898):650–654.
    1. Manning S, Pucci A, Batterham RL. GLP-1: a mediator of the beneficial metabolic effects of bariatric surgery? Physiology (Bethesda, Md) 2015;30(1):50–62.
    1. Batterham RL, Ffytche DH, Rosenthal JM, et al. PYY modulation of cortical and hypothalamic brain areas predicts feeding behaviour in humans. Nature. 2007;450(7166):106–109.
    1. Guida C, Stephen S, Guitton R, et al. The role of PYY in pancreatic islet physiology and surgical control of diabetes. Trends Endocrinol Metab TEM. 2017;28(8):626–636.
    1. Pucci A, Finer N. New medications for treatment of obesity: metabolic and cardiovascular effects. Can J Cardiol. 2015;31(2):142–152.
    1. Muller TD, Nogueiras R, Andermann ML, et al. Ghrelin. Mol Metab. 2015;4(6):437–460.
    1. Berthoud HR. Metabolic and hedonic drives in the neural control of appetite: who is the boss? Curr Opin Neurobiol. 2011;21(6):888–896.
    1. Seeley RJ, Chambers AP, Sandoval DA. The role of gut adaptation in the potent effects of multiple bariatric surgeries on obesity and diabetes. Cell Metab. 2015;21(3):369–378.
    1. Thomas C, Gioiello A, Noriega L, et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009;10(3):167–177.
    1. Dufer M, Horth K, Wagner R, et al. Bile acids acutely stimulate insulin secretion of mouse beta-cells via farnesoid X receptor activation and K(ATP) channel inhibition. Diabetes. 2012;61(6):1479–1489.
    1. Lundasen T, Galman C, Angelin B, et al. Circulating intestinal fibroblast growth factor 19 has a pronounced diurnal variation and modulates hepatic bile acid synthesis in man. J Intern Med. 2006;260(6):530–536.
    1. Fang S, Suh JM, Reilly SM, et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat Med. 2015;21(2):159–165.
    1. Watanabe M, Houten SM, Mataki C, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 2006;439(7075):484–489.
    1. Suzuki T, Aoyama J, Hashimoto M, et al. Correlation between postprandial bile acids and body fat mass in healthy normal-weight subjects. Clin Biochem. 2014;47(12):1128–1131.
    1. Turnbaugh PJ, Ley RE, Hamady M, et al. The human microbiome project. Nature. 2007;449(7164):804–810.
    1. Martinez KB, Pierre JF, Chang EB. The gut microbiota: the gateway to improved metabolism. Gastroenterol Clin N Am. 2016;45(4):601–614.
    1. Cummings DE. Taste and the regulation of food intake: it’s not just about flavor. Am J Clin Nutr. 2015;102(4):717–718.
    1. Piaggi P, Vinales KL, Basolo A, et al. Energy expenditure in the etiology of human obesity: spendthrift and thrifty metabolic phenotypes and energy-sensing mechanisms. J Endocrinol Invest. 2018;41(1):83–89.
    1. Glicksman C, Pournaras DJ, Wright M, et al. Postprandial plasma bile acid responses in normal weight and obese subjects. Ann Clin Biochem. 2010;47(Pt 5):482–484.
    1. Aron-Wisnewsky J, Clement K. The effects of gastrointestinal surgery on gut microbiota: potential contribution to improved insulin sensitivity. Curr Atheroscler Rep. 2014;16(11):454.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457(7228):480–484.
    1. Turnbaugh PJ, Ridaura VK, Faith JJ, et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1(6):6ra14.
    1. Backhed F, Ding H, Wang T, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 2004;101(44):15718–15723.
    1. Tambo A, Roshan MH, Pace NP. The microbial hypothesis: contributions of adenovirus infection and metabolic endotoxaemia to the pathogenesis of obesity. Int J Chronic Dis. 2016;2016:7030795.
    1. Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031.
    1. Barkeling B, King NA, Naslund E, et al. Characterization of obese individuals who claim to detect no relationship between their eating pattern and sensations of hunger or fullness. Int J Obes (2005) 2007;31(3):435–439.
    1. Sumithran P, Prendergast LA, Delbridge E, et al. Long-term persistence of hormonal adaptations to weight loss. N Engl J Med. 2011;365(17):1597–1604.
    1. Leibel RL, Hirsch J. Diminished energy requirements in reduced-obese patients. Metab Clin Exp. 1984;33(2):164–170.
    1. Browning KN, Fortna SR, Hajnal A. Roux-en-Y gastric bypass reverses the effects of diet-induced obesity to inhibit the responsiveness of central vagal motoneurones. J Physiol. 2013;591(9):2357–2372.
    1. Mann T, Tomiyama AJ, Westling E, et al. Medicare’s search for effective obesity treatments: diets are not the answer. Am Psychol. 2007;62(3):220–233.
    1. Ionut V, Bergman RN. Mechanisms responsible for excess weight loss after bariatric surgery. J Diabetes Sci Technol. 2011;5(5):1263–1282.
    1. Odstrcil EA, Martinez JG, Santa Ana CA, et al. The contribution of malabsorption to the reduction in net energy absorption after long-limb Roux-en-Y gastric bypass. Am J Clin Nutr. 2010;92(4):704–713.
    1. Manning S, Pucci A, Batterham RL. Roux-en-Y gastric bypass: effects on feeding behavior and underlying mechanisms. J Clin Investig. 2015;125(3):939–948.
    1. Dar MS, Chapman WH, 3rd, Pender JR, et al. GLP-1 response to a mixed meal: what happens 10 years after Roux-en-Y gastric bypass (RYGB)? Obes Surg. 2012;22(7):1077–1083.
    1. Yousseif A, Emmanuel J, Karra E, 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):241–252.
    1. Cummings DE, Weigle DS, Frayo RS, et al. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346(21):1623–1630.
    1. Chandarana K, Drew ME, Emmanuel J, et al. Subject standardization, acclimatization, and sample processing affect gut hormone levels and appetite in humans. Gastroenterology. 2009;136(7):2115–2126.
    1. Dirksen C, Jorgensen NB, Bojsen-Moller KN, et al. Gut hormones, early dumping and resting energy expenditure in patients with good and poor weight loss response after Roux-en-Y gastric bypass. Int J Obes (2005) 2013;37(11):1452–1459.
    1. le Roux CW, Welbourn R, Werling M, et al. Gut hormones as mediators of appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg. 2007;246(5):780–785.
    1. Svane MS, Jorgensen NB, Bojsen-Moller KN, et al. Peptide YY and glucagon-like peptide-1 contribute to decreased food intake after Roux-en-Y gastric bypass surgery. Int J Obes (2005) 2016;40(11):1699–1706.
    1. Pucci A, Cheung WH, Jones J, et al. A case of severe anorexia, excessive weight loss and high peptide YY levels after sleeve gastrectomy. Endocrinol Diabetes Metab Case Rep. 2015;2015:150020.
    1. Rao RS, Kini S. GIP and bariatric surgery. Obes Surg. 2011;21(2):244–252.
    1. Salinari S, Bertuzzi A, Asnaghi S, et al. First-phase insulin secretion restoration and differential response to glucose load depending on the route of administration in type 2 diabetic subjects after bariatric surgery. Diabetes Care. 2009;32(3):375–380.
    1. Rubino F, Gagner M, Gentileschi P, et al. 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):236–242.
    1. Suzuki K, Jayasena CN, Bloom SR. Obesity and appetite control. Exp Diabetes Res. 2012;2012:824305.
    1. Peterli R, Steinert RE, Woelnerhanssen B, et al. Metabolic and hormonal changes after laparoscopic Roux-en-Y gastric bypass and sleeve gastrectomy: a randomized, prospective trial. Obes Surg. 2012;22(5):740–748.
    1. Jacobsen SH, Olesen SC, Dirksen C, et al. Changes in gastrointestinal hormone responses, insulin sensitivity, and beta-cell function within 2 weeks after gastric bypass in non-diabetic subjects. Obes Surg. 2012;22(7):1084–1096.
    1. Sillakivi T, Suumann J, Kirsimagi U, et al. Plasma levels of gastric biomarkers in patients after bariatric surgery: biomarkers after bariatric surgery. Hepatogastroenterology. 2013;60(128):2129–2132.
    1. Egerod KL, Engelstoft MS, Grunddal KV, et al. A major lineage of enteroendocrine cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatostatin. Endocrinology. 2012;153(12):5782–5795.
    1. Steinert RE, Peterli R, Keller S, et al. Bile acids and gut peptide secretion after bariatric surgery: a 1-year prospective randomized pilot trial. Obesity (Silver Spring, Md) 2013;21(12):E660–E668.
    1. Pournaras DJ, Glicksman C, Vincent RP, et al. The role of bile after Roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology. 2012;153(8):3613–3619.
    1. Ahmad NN, Pfalzer A. Kaplan LM (2013) Roux-en-Y gastric bypass normalizes the blunted postprandial bile acid excursion associated with obesity. Int J Obes. 2005;37(12):1553–1559.
    1. Liou AP, Paziuk M, Luevano JM, Jr, et al. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5(178):178141.
    1. Ryan KK, Tremaroli V, Clemmensen C, et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509(7499):183–188.
    1. Midtvedt T, Norman A, Nygaard K. Bile acid transforming micro-organisms in rats with an intestinal blind segment. Acta Pathologica et microbiologica Scandinavica. 1969;77(1):162–166.
    1. Liu H, Hu C, Zhang X, et al. Role of gut microbiota, bile acids and their cross-talk in the effects of bariatric surgery on obesity and type 2 diabetes. J Diabetes Investig. 2018;9(1):13–20.
    1. Tremaroli V, Karlsson F, Werling M, et al. Roux-en-Y gastric bypass and vertical banded gastroplasty induce long-term changes on the human gut microbiome contributing to fat mass regulation. Cell Metab. 2015;22(2):228–238.
    1. Furet JP, Kong LC, Tap J, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59(12):3049–3057.
    1. Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4554–4561.
    1. Murphy R, Tsai P, Jullig M, et al. Differential changes in gut microbiota after gastric bypass and sleeve gastrectomy bariatric surgery vary according to diabetes remission. Obes Surg. 2017;27(4):917–925.
    1. Medina DA, Pedreros JP, Turiel D, et al. Distinct patterns in the gut microbiota after surgical or medical therapy in obese patients. PeerJ. 2017;5:e3443.
    1. Guo Y, Liu CQ, Shan CX, et al. Gut microbiota after Roux-en-Y gastric bypass and sleeve gastrectomy in a diabetic rat model: increased diversity and associations of discriminant genera with metabolic changes. Diabetes/Metab Res Rev. 2017
    1. Cavin JB, Couvelard A, Lebtahi R, et al. Differences in alimentary glucose absorption and intestinal disposal of blood glucose after Roux-en-Y gastric bypass vs sleeve gastrectomy. Gastroenterology. 2016;150(2):454–464.
    1. Saeidi N, Meoli L, Nestoridi E, et al. Reprogramming of intestinal glucose metabolism and glycemic control in rats after gastric bypass. Science (New York, NY) 2013;341(6144):406–410.
    1. Baud G, Daoudi M, Hubert T, et al. Bile diversion in Roux-en-Y gastric bypass modulates sodium-dependent glucose intestinal uptake. Cell Metab. 2016;23(3):547–553.
    1. Zakeri R, Batterham RL. Potential mechanisms underlying the effect of bariatric surgery on eating behaviour. Curr Opin Endocrinol Diabetes Obes. 2018;25(1):3–11.
    1. Berthoud HR. Vagal and hormonal gut-brain communication: from satiation to satisfaction. Neurogastroenterol Motil. 2008;20(Suppl 1):64–72.
    1. Berthoud HR, Shin AC, Zheng H. Obesity surgery and gut-brain communication. Physiol Behav. 2011;105(1):106–119.
    1. Leibel RL, Seeley RJ, Darsow T, et al. Biologic responses to weight loss and weight regain: report from an American diabetes association research symposium. Diabetes. 2015;64(7):2299–2309.

Source: PubMed

Подписаться