1
|
Cook TM, Fuller KNZ, Sandoval DA. Insights into the neurobiology of weight loss after bariatric surgery and GLP-1R agonists. Neuropharmacology 2025; 265:110269. [PMID: 39675463 PMCID: PMC11702201 DOI: 10.1016/j.neuropharm.2024.110269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 11/18/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
Obesity and its related complications are growing in prevalence worldwide, with increasing impact to individuals and healthcare systems alike. Currently, the leading treatment approaches for effective and sustained weight loss are bariatric surgery and gut peptide therapeutics. At a high level, both treatment strategies work by hijacking gut-brain axis signaling to reduce food intake. However, we predict that each modality has distinct neuronal mechanisms that are responsible for their success and complications. This review compares the neurobiology of feeding behavior between these two weight loss strategies via a discussion of both clinical and pre-clinical data. The most compelling evidence points to signaling within the hindbrain, hypothalamus, and reward circuits contributing to weight loss. Considerations for treatment, including differing complications between the two treatment approaches, will also be discussed. Based on the data, we pose the hypothesis that these two interventions are acting via distinct mechanisms to induce weight loss. Both interventions have variable degrees of weight loss across the patient population, thus, understanding these distinct mechanisms could help drive individualized medicine to optimize weight loss. This article is part of the Special Issue on "Food intake and feeding states".
Collapse
Affiliation(s)
- Tyler M Cook
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly N Z Fuller
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Darleen A Sandoval
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
2
|
Yang K, Wu YT, He Y, Dai JX, Luo YL, Xie JH, Ding WJ. GLP-1 and IL-6 regulates obesity in the gut and brain. Life Sci 2025; 362:123339. [PMID: 39730038 DOI: 10.1016/j.lfs.2024.123339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Obesity is a chronic metabolic disease characterized by excessive nutrient intake leading to increased subcutaneous or visceral fat, resulting in pathological and physiological changes. The incidence rate of obesity, an important form of metabolic syndrome, is increasing worldwide. Excess appetite is a key pathogenesis of obesity, and the inflammatory response induced by obesity has received increasing attention. This review focuses on the role of appetite-regulating factor (Glucogan-like peptide 1) and inflammatory factor (Interleukin-6) in the gut and brain in individuals with obesity and draws insights from the current literature.
Collapse
Affiliation(s)
- Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ting Wu
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin-Xiu Dai
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yu-Lu Luo
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jing-Hui Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei-Jun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
3
|
Rayas M, Pezzica S, Honka H, Carli F, Peterson R, DeFronzo R, Gastaldelli A, Salehi M. GLP-1 enhances β-cell response to protein ingestion and bariatric surgery amplifies it. Obesity (Silver Spring) 2025; 33:104-115. [PMID: 39635951 DOI: 10.1002/oby.24182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/23/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE The glycemic-independent actions of glucagon-like peptide-1 (GLP-1) in the prandial state in humans are unknown. We examined the contribution of GLP-1 to β-cell secretory response (primary endpoint) and glucose metabolism during protein ingestion under basal glycemia, as well as whether these responses are affected by rerouted gut after gastric bypass (GB) or sleeve gastrectomy (SG). METHODS Insulin secretion rate (ISR) and glucose fluxes during a 50-g oral protein load were compared among 10 nondiabetic individuals with GB, 9 with SG, and 7 non-operated controls (CN), with and without intravenous infusion of exendin(9-39) (Ex-9), a GLP-1 receptor (GLP-1R) antagonist. RESULTS Blocking GLP-1R increased glucose before and after protein ingestion and decreased β-cell sensitivity to glucose in the first 30 min of protein ingestion in all three groups (p < 0.05). Reduction in the premeal ISR by Ex-9 infusion was only observed in CN, whereas diminished prandial ISR3h by GLP-1R blockade was only observed in GB and SG (p < 0.05 for interaction). GLP-1R blockade enhanced post-protein insulin action in GB and SG, but not in CN, and exaggerated endogenous glucose production only GB (p < 0.05 for interaction). CONCLUSIONS These findings are consistent with both pancreatic and extra-pancreatic roles for GLP-1 during protein ingestion in humans that are exaggerated by bariatric surgery.
Collapse
Affiliation(s)
- Maria Rayas
- Department of Pediatrics, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Samantha Pezzica
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Henri Honka
- Division of Diabetes, Department of Medicine, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Fabrizia Carli
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Richard Peterson
- Department of Surgery, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Ralph DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
- Division of Diabetes, Department of Medicine, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Marzieh Salehi
- Division of Diabetes, Department of Medicine, University of Texas Health at San Antonio, San Antonio, Texas, USA
- South Texas Veterans Health Care System, Audie Murphy Hospital, San Antonio, Texas, USA
| |
Collapse
|
4
|
Pasveer YM, Aydin Ö, Groen AK, Meijnikman AS, Nieuwdorp M, Gerdes VEA, van Riel NAW. Does GLP-1 cause post-bariatric hypoglycemia: 'Computer says no'. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108424. [PMID: 39326360 DOI: 10.1016/j.cmpb.2024.108424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/18/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND OBJECTIVE Patients who underwent Roux-en-Y Gastric Bypass surgery for treatment of obesity or diabetes can suffer from post-bariatric hypoglycemia (PBH). It has been assumed that PBH is caused by increased levels of the hormone GLP-1. In this research, we elucidate the role of GLP-1 in PBH with a physiology-based mathematical model. METHODS The Eindhoven Diabetes Simulator (EDES) model, simulating postprandial glucose homeostasis, was adapted to include the effect of GLP-1 on insulin secretion. Parameter sensitivity analysis was used to identify parameters that could cause PBH. Virtual patient models were created by defining sets of models parameters based on 63 participants from the HypoBaria study cohort, before and one year after bariatric surgery. RESULTS Simulations with the virtual patient models showed that glycemic excursions can be correctly simulated for the study population, despite heterogeneity in the glucose, insulin and GLP-1 data. Sensitivity analysis showed that GLP-1 stimulated insulin secretion alone was not able to cause PBH. Instead, analyses showed the increased transit speed of the ingested food resulted in quick and increased glucose absorption in the gut after surgery, which in turn induced postprandial glycemic dips. Furthermore, according to the model post-bariatric increased rate of glucose absorption in combination with different levels of insulin sensitivity can result in PBH. CONCLUSIONS Our model findings implicate that if initial rapid improvement in insulin sensitivity after gastric bypass surgery is followed by a more gradual decrease in insulin sensitivity, this may result in the emergence of PBH after prolonged time (months to years after surgery).
Collapse
Affiliation(s)
- Ysanne M Pasveer
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Ömrüm Aydin
- Department of Vascular Medicine, Amsterdam UMC - AMC, Amsterdam, The Netherlands
| | - Albert K Groen
- Department of Vascular Medicine, Amsterdam UMC - AMC, Amsterdam, The Netherlands
| | - Abraham S Meijnikman
- Department of Vascular Medicine, Amsterdam UMC - AMC, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam UMC - AMC, Amsterdam, The Netherlands
| | - Victor E A Gerdes
- Department of Vascular Medicine, Amsterdam UMC - AMC, Amsterdam, The Netherlands; Department of Bariatric Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands; Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Natal A W van Riel
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Department of Vascular Medicine, Amsterdam UMC - AMC, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Seo E, Ryu SW. HbA1c Reductions Following Sleeve Gastrectomy Versus Sleeve Gastrectomy With Proximal Jejunal Bypass: Significant Differences in Diabetic Patients. JOURNAL OF METABOLIC AND BARIATRIC SURGERY 2024; 13:71-77. [PMID: 39802591 PMCID: PMC11717990 DOI: 10.17476/jmbs.2024.13.2.71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Sleeve gastrectomy with proximal jejunal bypass (SG with PJB) is often preferred for patients with higher body mass index (BMI) and inadequate glycemic control. This study aimed to compare the outcomes of SG and SG with PJB, focusing on glycemic control and BMI in prediabetic and diabetic patients. Preoperative analysis revealed differences in age and hemoglobin A1c (HbA1c) levels between groups: SG with PJB patients were older (38.72±9.75 vs. 34.93±10.90 years, P=0.002) and had higher HbA1c levels (7.25±1.76 vs. 5.86±0.78%, P<0.001). Patients were stratified into prediabetic and diabetic groups. In the prediabetic group, no preoperative differences were observed between the surgical groups. However, in the diabetic group, patients in the SG with PJB had lower BMI (37.77±5.83 vs. 41.08±8.5 kg/m2, P=0.034) and higher HbA1c levels (7.88±1.72 vs. 6.51±1.37%, P<0.001) compared to the SG, despite stratification. Postoperatively, SG with PJB led to significantly lower BMI at 3 months compared to SG, but this difference was not sustained at 6 and 12 months in the prediabetic group. In diabetic patients, SG with PJB resulted in significantly greater reductions in HbA1c levels compared to SG, even when adjusted for BMI as a covariate. At the 12-month follow-up, although SG with PJB still showed higher HbA1c levels than SG (5.79±0.78 vs. 5.59±0.44%, P=0.031), the difference was smaller compared to the preoperative period, where SG with PJB had significantly higher levels (7.88±1.72 vs. 6.51±1.37%, P<0.001). These findings suggest that SG with PJB may offer superior glycemic control in morbidly obese diabetic patients.
Collapse
Affiliation(s)
- Eunhye Seo
- College of Nursing, Keimyung University, Daegu, Korea
| | - Seung Wan Ryu
- Department of Surgery, Keimyung University Dongsan Medical Centre, Daegu, Korea
| |
Collapse
|
6
|
Hedbäck N, Dichman ML, Hindsø M, Dirksen C, Jørgensen NB, Bojsen-Møller KN, Kristiansen VB, Rehfeld JF, Hartmann B, Holst JJ, Svane MS, Madsbad S. Effect of ghrelin on glucose tolerance, gut hormones, appetite, and food intake after sleeve gastrectomy. Am J Physiol Endocrinol Metab 2024; 327:E396-E410. [PMID: 39082900 PMCID: PMC11427089 DOI: 10.1152/ajpendo.00177.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Ghrelin is an appetite-stimulating hormone secreted from the gastric mucosa in the fasting state, and secretion decreases in response to food intake. After sleeve gastrectomy (SG), plasma concentrations of ghrelin decrease markedly. Whether this affects appetite and glucose tolerance postoperatively is unknown. We investigated the effects of ghrelin infusion on appetite and glucose tolerance in individuals with obesity before and 3 mo after SG. Twelve participants scheduled for SG were included. Before and 3 mo after surgery, a mixed-meal test followed by an ad libitum meal test was performed with concomitant infusions of acyl-ghrelin (1 pmol/kg/min) or placebo. Infusions began 60 min before meal intake to reach a steady state before the mixed-meal and were continued throughout the study day. Two additional experimental days with 0.25 pmol/kg/min and 10 pmol/kg/min of acyl-ghrelin infusions were conducted 3 mo after surgery. Both before and after SG, postprandial glucose concentrations increased dose dependently during ghrelin infusions compared with placebo. Ghrelin infusions inhibited basal and postprandial insulin secretion rates, resulting in lowered measures of β-cell function, but no effect on insulin sensitivity was seen. Ad libitum meal intake was unaffected by the administration of ghrelin. In conclusion, ghrelin infusion increases postprandial plasma glucose concentrations and impairs β-cell function before and after SG but has no effect on ad libitum meal intake. We speculate that the lower concentration of ghrelin after SG may impact glucose metabolism following this procedure.NEW & NOTEWORTHY Ghrelin's effect on glucose tolerance and food intake following sleeve gastrectomy (SG) was evaluated. Acyl-ghrelin was infused during a mixed-meal and ad libitum meals before and 3 mo after surgery. Postprandial glucose concentrations increased during ghrelin infusions, both before and after surgery, while insulin production was inhibited. However, ad libitum meal intake did not differ during ghrelin administration compared with placebo. The decreased ghrelin concentration following SG may contribute to the glycemic control after surgery.
Collapse
Affiliation(s)
- Nora Hedbäck
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Biomedical Sciences, SUND, University of Copenhagen, Copenhagen, Denmark
| | - Marie-Louise Dichman
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Biomedical Sciences, SUND, University of Copenhagen, Copenhagen, Denmark
| | - Morten Hindsø
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Carsten Dirksen
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Biomedical Sciences, SUND, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nils Brun Jørgensen
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Kirstine Nyvold Bojsen-Møller
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Viggo B Kristiansen
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, SUND, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, SUND, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Maria Saur Svane
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| |
Collapse
|
7
|
Hansen LS, Gasbjerg LS, Brønden A, Dalsgaard NB, Bahne E, Stensen S, Hellmann PH, Rehfeld JF, Hartmann B, Wewer Albrechtsen NJ, Holst JJ, Vilsbøll T, Knop FK. The role of glucagon-like peptide 1 in the postprandial effects of metformin in type 2 diabetes: a randomized crossover trial. Eur J Endocrinol 2024; 191:192-203. [PMID: 39049802 DOI: 10.1093/ejendo/lvae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024]
Abstract
AIMS Although metformin is widely used for treatment of type 2 diabetes (T2D), its glucose-lowering mechanism remains unclear. Using the glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) antagonist exendin(9-39)NH2, we tested the hypothesis that postprandial GLP-1-mediated effects contribute to the glucose-lowering potential of metformin in T2D. METHODS In a randomized, placebo-controlled, double-blind, crossover study, 15 individuals with T2D (median HbA1c 50 mmol/mol [6.7%], body mass index 30.1 kg/m2, age 71 years) underwent, in randomized order, 14 days of metformin and placebo treatment, respectively. Each treatment period was preceded by 14 days without any glucose-lowering medicine and concluded by two 4 h mixed meal tests performed in randomized order and separated by >24 h with either continuous intravenous exendin(9-39)NH2 or saline infusion. RESULTS Compared to placebo, metformin treatment lowered fasting plasma glucose (mean of differences [MD] 1.4 mmol/L × min [95% CI 0.8-2.0]) as well as postprandial plasma glucose excursions during both saline infusion (MD 186 mmol/L × min [95% CI 64-307]) and exendin(9-39)NH2 infusion (MD 268 mmol/L × min [95% CI 108-427]). The metformin-induced improvement in postprandial glucose tolerance was unaffected by GLP-1R antagonization (MD 82 mmol/L × min [95% CI -6564-170]). Metformin treatment increased fasting plasma GLP-1 (MD 1.7 pmol/L × min [95% CI 0.39-2.9]) but did not affect postprandial GLP-1 responses (MD 820 pmol/L × min [95% CI -1750-111]). CONCLUSIONS Using GLP-1R antagonization, we could not detect GLP-1-mediated postprandial glucose-lowering effect of metformin in individuals with T2D. We show that 2 weeks of metformin treatment increases fasting plasma GLP-1, which may contribute to metformin's beneficial effect on fasting plasma glucose in T2D. Trial registration: Clinicaltrials.gov NCT03246451.
Collapse
Affiliation(s)
- Laura S Hansen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Andreas Brønden
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Pharmacology, Copenhagen University Hospital-Bispebjerg and Frederiksberg, DK-2400 Copenhagen NV, Denmark
| | - Niels B Dalsgaard
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Emilie Bahne
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Signe Stensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Pernille H Hellmann
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | | | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Tina Vilsbøll
- Clinical Research, Steno Diabetes Center Copenhagen, University of Copenhagen, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, University of Copenhagen, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Holst JJ, Madsbad S, Bojsen-Møller KN, Dirksen C, Svane M. New Lessons from the gut: Studies of the role of gut peptides in weight loss and diabetes resolution after gastric bypass and sleeve gastrectomy. Peptides 2024; 176:171199. [PMID: 38552903 DOI: 10.1016/j.peptides.2024.171199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
It has been known since 2005 that the secretion of several gut hormones changes radically after gastric bypass operations and, although more moderately, after sleeve gastrectomy but not after gastric banding. It has therefore been speculated that increased secretion of particularly GLP-1 and Peptide YY (PYY), which both inhibit appetite and food intake, may be involved in the weight loss effects of surgery and for improvements in glucose tolerance. Experiments involving inhibition of hormone secretion with somatostatin, blockade of their actions with antagonists, or blockade of hormone formation/activation support this notion. However, differences between results of bypass and sleeve operations indicate that distinct mechanisms may also be involved. Although the reductions in ghrelin secretion after sleeve gastrectomy would seem to provide an obvious explanation, experiments with restoration of ghrelin levels pointed towards effects on insulin secretion and glucose tolerance rather than on food intake. It seems clear that changes in GLP-1 secretion are important for insulin secretion after bypass and appear to be responsible for postbariatric hypoglycemia in glucose-tolerant individuals; however, with time the improvements in insulin sensitivity, which in turn are secondary to the weight loss, may be more important. Changes in bile acid metabolism do not seem to be of particular importance in humans.
Collapse
Affiliation(s)
- Jens Juul Holst
- The NovoNordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Denmark.
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| | | | - Carsten Dirksen
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| | - Maria Svane
- Department of Endocrinology, Hvidovre Hospital, University of Copenhagen, Denmark
| |
Collapse
|
9
|
Honka H, Bhattacharjee J, Zadeh M, Kohli R, Gastaldelli A, Salehi M. Vagal activation alters prandial bile acid composition and glycemia in patients with hypoglycemia after Roux-en-Y gastric bypass surgery. Neurogastroenterol Motil 2024; 36:e14763. [PMID: 38342974 PMCID: PMC11043014 DOI: 10.1111/nmo.14763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/07/2023] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Altered prandial glycemic response after Roux-en-Y gastric bypass (RYGB) is exaggerated in patients with post-RYGB hypoglycemia. Increased contribution of glucagon-like peptide 1 (GLP-1) to prandial insulin secretion plays a key role in developing hypoglycemia after RYGB, but the role of nonhormonal gut factors remains unknown. Here, the effect of vagal activation on prandial bile acid (BA) composition in relation to glucose, insulin and gut hormone responses was examined in a small size group of nondiabetic subjects after RYGB with intact gallbladder compared to nonoperated controls. METHODS Concentrations of blood glucose, hormones, and BAs were measured in two RYGB subjects with documented hypoglycemia (HGB), three asymptomatic RYGB-treated subjects (AGB), and four nonoperated controls with intact gallbladders during a meal-tolerance test with (MTT-Sham) and without (MTT) preceding modified sham feeding (chew and spit). KEY RESULTS Meal ingestion raised serum total BAs in RYGB-treated subjects without any effect in nonoperated controls. Modified sham feeding similarly increased meal-induced responses of conjugated BAs (CBAs) in all subjects (p < 0.05 compared to MTT alone), whereas unconjugated BAs (UBAs), mainly deoxycholic and chenodeoxycholic acid, were raised only in the HGB group (p < 0.001 for interaction). Prandial UBAs had an inverse correlation with glucose nadir (r = -0.75, p < 0.05) and were directly associated with ISR and GLP-1 during MTT-Sham. CONCLUSIONS & INFERENCES In this small cohort, vagal activation by modified sham feeding increases prandial CBAs in both operated and nonoperated subjects but enhances UBAs only in patients with documented post-RYGB hypoglycemia. Our findings highlight a potential role for nonhormonal gut factors, such as BA and gut microbiome, in glucose abnormalities after RYGB.
Collapse
Affiliation(s)
- Henri Honka
- University of Texas Health Science Center, Division of Diabetes, San Antonio, TX
| | - Jashdeep Bhattacharjee
- Children’s Hospital Los Angeles, Division of Gastroenterology, Hepatology and Nutrition, Los Angeles, CA
| | - Mansour Zadeh
- University of Texas Health Science Center, Department of Microbiology, Immunology and Molecular Genetics, San Antonio, TX
| | - Rohit Kohli
- Children’s Hospital Los Angeles, Division of Gastroenterology, Hepatology and Nutrition, Los Angeles, CA
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology-National Research Council, Pisa, Italy
| | - Marzieh Salehi
- University of Texas Health Science Center, Division of Diabetes, San Antonio, TX
- University of Cincinnati College of Medicine, Department of Medicine, Cincinnati, OH
- South Texas Veteran Health Care System, Audie Murphy Hospital, San Antonio, TX
| |
Collapse
|
10
|
Bailey CJ, Flatt PR. Duodenal enteroendocrine cells and GIP as treatment targets for obesity and type 2 diabetes. Peptides 2024; 174:171168. [PMID: 38320643 DOI: 10.1016/j.peptides.2024.171168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/08/2024]
Abstract
The duodenum is an important source of endocrine and paracrine signals controlling digestion and nutrient disposition, notably including the main incretin hormone glucose-dependent insulinotropic polypeptide (GIP). Bariatric procedures that prevent nutrients from contact with the duodenal mucosa are particularly effective interventions to reduce body weight and improve glycaemic control in obesity and type 2 diabetes. These procedures take advantage of increased nutrient delivery to more distal regions of the intestine which enhances secretion of the other incretin hormone glucagon-like peptide-1 (GLP-1). Preclinical experiments have shown that either an increase or a decrease in the secretion or action of GIP can decrease body weight and blood glucose in obesity and non-insulin dependent hyperglycaemia, but clinical studies involving administration of GIP have been inconclusive. However, a synthetic dual agonist peptide (tirzepatide) that exerts agonism at receptors for GIP and GLP-1 has produced marked weight-lowering and glucose-lowering effects in people with obesity and type 2 diabetes. This appears to result from chronic biased agonism in which the novel conformation of the peptide triggers enhanced signalling by the GLP-1 receptor through reduced internalisation while reducing signalling by the GIP receptor directly or via functional antagonism through increased internalisation and degradation.
Collapse
Affiliation(s)
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA Northern Ireland, UK
| |
Collapse
|
11
|
Kong X, Feng L, Yan D, Li B, Yang Y, Ma X. FXR-mediated epigenetic regulation of GLP-1R expression contributes to enhanced incretin effect in diabetes after RYGB. J Cell Mol Med 2024; 28:e16339. [PMID: 33611845 PMCID: PMC10941525 DOI: 10.1111/jcmm.16339] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 01/07/2023] Open
Abstract
In this study, we investigated how Roux-en-Y gastric bypass (RYGB) enhances glucagon-like peptide 1 (GLP-1) response in GK rats and explored the potential link between RYGB-stimulated BAs/FXR signalling and GLP-1R-linked signalling in β-cells, a key pathway that regulates glucose-stimulated insulin secretion (GSIS). Here we show that RYGB restores GLP-1R expression in GK rat islets. This involves increased total BAs as well as chenodeoxycholic acid (CDCA), leading to FXR activation, increasing FXR binding to the promoter of Glp-1r and enhancing occupancy of histone acetyltransferase steroid receptor coactivator-1 (SRC1), thus increasing histone H3 acetylation at the promoter. These coordinated events bring about increased GLP-1R expression, resulting in greater GLP-1 response in β-cells. Moreover, ablation of FXR suppressed the stimulatory effects of GLP-1. Thus, this study unravels the crucial role of the BAs/FXR/SRC1 axis-controlled GLP-1R expression in β-cells, which results in enhanced incretin effect and normalized blood glucose of GK rats after RYGB.
Collapse
Affiliation(s)
- Xiangchen Kong
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Linxian Feng
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Dan Yan
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Bingfeng Li
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Yanhui Yang
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| | - Xiaosong Ma
- Shenzhen University Diabetes InstituteSchool of MedicineShenzhen UniversityShenzhenChina
| |
Collapse
|
12
|
Rayas M, Gastaldelli A, Honka H, Pezzica S, Carli F, Peterson R, DeFronzo R, Salehi MS. GLP-1 enhances beta-cell response to protein ingestion and bariatric surgery amplifies it. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.22.23297377. [PMID: 37961500 PMCID: PMC10635165 DOI: 10.1101/2023.10.22.23297377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
OBJECTIVE Protein ingestion stimulates β-cell secretion and alters glucose flux. Enhanced action of glucagon-like peptide 1 (GLP-1) and increased plasma glucose excursion contribute to prandial hyperinsulinemia after gastric bypass surgery (GB) and sleeve gastrectomy (SG). We examined the contribution of endogenous GLP-1 to glucose kinetics and β-cell response to protein ingestion under basal glucose concentrations in humans, and whether these responses are affected by rerouted gut after GB or SG. DESIGN Glucose fluxes, insulin secretion rate (ISR), and incretin responses to a 50-gram oral protein load were compared between 10 non-diabetic individuals with GB, 9 matched subjects with SG and 7 non-operated controls (CN) with and without intravenous infusion of exendin-(9- 39) [Ex-9), a specific GLP-1 receptor (GLP-1R) antagonist. RESULTS Blocking GLP-1R increased the plasma glucose concentration before and after protein ingestion in all 3 groups (p<0.05) and decreased β-cell sensitivity to glucose in the first 30 minutes of protein ingestion (p<0.05). Reduction in the prandial ISR3h by Ex-9 infusion, however, only was observed in GB and SG (p<0.05 for interaction) and not in controls. Also, GLP-1R blockade increased post-protein insulin action in GB and SG, but not CN (p=0.09 for interaction). Endogenous glucose production (EGP) during the first 60 minutes after protein ingestion was increased in all 3 groups but EGP3h only was accentuated in GB by Ex-9 infusion (p<0.05 for interaction). CONCLUSION These findings are consistent with both a pancreatic and extrapancreatic role for GLP-1 during protein ingestion in humans, and GLP-1 actions are exaggerated by bariatric surgery.
Collapse
|
13
|
Mashayekhi M, Nian H, Mayfield D, Devin JK, Gamboa JL, Yu C, Silver HJ, Niswender K, Luther JM, Brown NJ. Weight Loss-Independent Effect of Liraglutide on Insulin Sensitivity in Individuals With Obesity and Prediabetes. Diabetes 2024; 73:38-50. [PMID: 37874653 PMCID: PMC10784656 DOI: 10.2337/db23-0356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023]
Abstract
Metabolic effects of glucagon-like peptide 1 (GLP-1) receptor agonists are confounded by weight loss and not fully recapitulated by increasing endogenous GLP-1. We tested the hypothesis that GLP-1 receptor (GLP-1R) agonists exert weight loss-independent, GLP-1R-dependent effects that differ from effects of increasing endogenous GLP-1. Individuals with obesity and prediabetes were randomized to receive for 14 weeks the GLP-1R agonist liraglutide, a hypocaloric diet, or the dipeptidyl peptidase 4 (DPP-4) inhibitor sitagliptin. The GLP-1R antagonist exendin(9-39) and placebo were administered in a two-by-two crossover study during mixed-meal tests. Liraglutide and diet, but not sitagliptin, caused weight loss. Liraglutide improved insulin sensitivity measured by HOMA for insulin resistance (HOMA-IR), the updated HOMA model (HOMA2), and the Matsuda index after 2 weeks, prior to weight loss. Liraglutide decreased fasting and postprandial glucose levels, and decreased insulin, C-peptide, and fasting glucagon levels. In contrast, diet-induced weight loss improved insulin sensitivity by HOMA-IR and HOMA2, but not the Matsuda index, and did not decrease glucose levels. Sitagliptin increased endogenous GLP-1 and GIP values without altering insulin sensitivity or fasting glucose levels, but decreased postprandial glucose and glucagon levels. Notably, sitagliptin increased GIP without altering weight. Acute GLP-1R antagonism increased glucose levels in all groups, increased the Matsuda index and fasting glucagon level during liraglutide treatment, and increased endogenous GLP-1 values during liraglutide and sitagliptin treatments. Thus, liraglutide exerts rapid, weight loss-independent, GLP-1R-dependent effects on insulin sensitivity that are not achieved by increasing endogenous GLP-1. ARTICLE HIGHLIGHTS Metabolic benefits of glucagon-like peptide 1 (GLP-1) receptor agonists are confounded by weight loss and are not fully achieved by increasing endogenous GLP-1 through dipeptidyl peptidase 4 (DPP-4) inhibition. We investigated weight loss-independent, GLP-1 receptor (GLP-1R)-dependent metabolic effects of liraglutide versus a hypocaloric diet or the DPP-4 inhibitor sitagliptin. GLP-1R antagonism with exendin(9-39) was used to assess GLP-1R-dependent effects during mixed meals. Liraglutide improved insulin sensitivity and decreased fasting and postprandial glucose prior to weight loss, and these benefits were reversed by exendin(9-39). GLP-1R agonists exert rapid, weight loss-independent, GLP-1R-dependent effects on insulin sensitivity not achieved by increasing endogenous GLP-1.
Collapse
Affiliation(s)
- Mona Mashayekhi
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | - Hui Nian
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Dustin Mayfield
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Jessica K. Devin
- UCHealth Endocrinology, Yampa Valley Medical Center, Steamboat Springs, CO
| | - Jorge L. Gamboa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Chang Yu
- Department of Population Health, NYU Grossman School of Medicine, New York, NY
| | - Heidi J. Silver
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| | - Kevin Niswender
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | - James M. Luther
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN
| | - Nancy J. Brown
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| |
Collapse
|
14
|
Yamamoto Y, Togawa T, Sekine O, Ozamoto Y, Fuse J, Azuma C, Ito-Kobayashi J, Oe Y, Hagiwara A, Kobayashi M, Kitamura T, Iwanishi M, Shimatsu A, Kashiwagi A. The oral disposition index calculated from a meal tolerance test is a crucial indicator for evaluating differential normalization of postprandial glucose and triglyceride excursions in morbidly obese patients after laparoscopic sleeve gastrectomy. Endocr J 2023; 70:1141-1157. [PMID: 37853621 DOI: 10.1507/endocrj.ej23-0241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2023] Open
Abstract
To determine the normalization of postprandial blood glucose (PG) and triglyceride (TG) excursions in 30 morbidly obese patients with or without diabetes mellitus (DM) 1-year after they underwent a laparoscopic sleeve gastrectomy (LSG) vs. their pre-surgery data, we administered the 75-g oral glucose tolerance test (OGTT) and a meal tolerance test (MTT) using a 75-g glucose-equivalent carbohydrate- and fat-containing meal. The results were as follows; (i) Postoperative body-weight reduction was associated with DM remission and reduced multiple cardiometabolic risks. (ii) OGTT data showing postprandial hyper-insulinemic hypoglycemia in many post-surgery patients were associated with overdiagnosis of improved glucose tolerance. However, postoperative MTT data without hypoglycemia showed no improvement in the glucose tolerance vs. pre-surgery data. (iii) The disposition index (DI) i.e., [Matsuda index] × (Glucose-induced insulin secretion) was progressively worsened from normal glucose tolerance to DM patients after LSG. These post-surgery DI values measured by the MTT were correlated with 2h-plasma glucose levels and were not normalized in DM patients. (iv) The baseline, 2h-TG, and an increase in 2h-TG values above baseline were correlated with the insulin resistance index, DI, or HbA1c; These TG values were normalized post-LSG. In conclusion, the glucose tolerance curve measured by the MTT was not normalized in T2DM patients, which was associated with impaired normalization of the DI values in those patients 1-year after the LSG. However, the baseline TG and a fat-induced 2h-TG values were normalized postoperatively. The MTT can be used to assess normalization in postprandial glucose and TG excursions after LSG.
Collapse
Affiliation(s)
- Yukako Yamamoto
- Department of Diabetes and Endocrinology, Omi Medical Center, Shiga 525-8585, Japan
| | - Takeshi Togawa
- Department of Bariatric and Metabolic Surgery, Omi Medical Center, Shiga 525-8585, Japan
| | - Osamu Sekine
- Department of Diabetes and Endocrinology, Omi Medical Center, Shiga 525-8585, Japan
| | - Yuki Ozamoto
- Department of Bariatric and Metabolic Surgery, Omi Medical Center, Shiga 525-8585, Japan
| | - Junko Fuse
- Nutritional Department, Omi Medical Center, Shiga 525-8585, Japan
| | - Choka Azuma
- Department of Diabetes and Endocrinology, Omi Medical Center, Shiga 525-8585, Japan
| | - Jun Ito-Kobayashi
- Department of Diabetes and Endocrinology, Omi Medical Center, Shiga 525-8585, Japan
| | - Yasumitsu Oe
- Department of Bariatric and Metabolic Surgery, Omi Medical Center, Shiga 525-8585, Japan
| | - Akeo Hagiwara
- Department of Bariatric and Metabolic Surgery, Omi Medical Center, Shiga 525-8585, Japan
| | - Masaki Kobayashi
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
| | - Masanori Iwanishi
- Department of Diabetes and Endocrinology, Omi Medical Center, Shiga 525-8585, Japan
| | - Akira Shimatsu
- Department of Diabetes and Endocrinology, Omi Medical Center, Shiga 525-8585, Japan
| | - Atsunori Kashiwagi
- Department of Diabetes and Endocrinology, Omi Medical Center, Shiga 525-8585, Japan
| |
Collapse
|
15
|
Honka H, Gastaldelli A, Pezzica S, Peterson R, DeFronzo R, Salehi M. Endogenous glucagon-like peptide 1 diminishes prandial glucose counterregulatory response to hypoglycemia after gastric bypass surgery. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.20.23295840. [PMID: 37790563 PMCID: PMC10543055 DOI: 10.1101/2023.09.20.23295840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
We have previously shown that prandial endogenous glucose production (EGP) during insulin-induced hypoglycemia is smaller in non-diabetic subjects with gastric bypass (GB), where prandial glucagon-like peptide 1 (GLP-1) concentrations are 5-10 times higher than those in non-operated controls. Here, we sought to determine the effect of endogenous GLP-1 on prandial counterregulatory response to hypoglycemia after GB. Glucose fluxes, and islet-cell and gut hormone responses before and after mixed-meal ingestion were compared during a hyperinsulinemic hypoglycemic (~3.2 mmol/l) clamp with and without a GLP-1 receptor (GLP-1R) antagonist exendin-(9-39) (Ex-9) in non-diabetic subjects with prior GB compared to matched subjects with SG and non-surgical controls. In this setting, GLP-1R blockade had no effect on insulin secretion or insulin action, whereas prandial glucagon was enhanced in all 3 groups. Ex-9 infusion raised prandial EGP response to hypoglycemia in every GB subject but had no consistent effects on EGP among subjects with SG or non-operated controls (P < 0.05 for interaction). These results indicate that impaired post-meal glucose counterregulatory response to hypoglycemia after GB is partly mediated by endogenous GLP-1, highlighting a novel mechanism of action of GLP-1R antagonists for the treatment of prandial hypoglycemia in this population.
Collapse
|
16
|
Ferreira FP, Pereira SS, Costa MM, Guimarães M, Albrechtsen NJW, Holst JJ, Nora M, Monteiro MP. Individuals with type 2 diabetes have higher density of small intestinal neurotensin-expressing cells. Mol Cell Biochem 2023; 478:2779-2787. [PMID: 36920577 PMCID: PMC10627918 DOI: 10.1007/s11010-023-04698-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 03/01/2023] [Indexed: 03/16/2023]
Abstract
Neurotensin (NT) is a gastro-intestinal hormone involved in several pathways that regulate energy and glucose homeostasis. NT was hypothesized to act in synergy with incretin hormones to potentiate its anti-diabetic effects. Additionally, circulating NT levels were shown to rise after bariatric surgery-induced weight loss. Knowledge of NT-secreting cells distribution along the small intestine and its variation according to diabetes status could provide insights on NT role in mediating type 2 diabetes (T2D) improvement after bariatric surgery. So, our aims were to characterize NT-expressing cell distribution along the human small intestine and to compare the relative density of NT-expressing cells in the small intestine of individuals with and without T2D undergoing bariatric surgery for obesity treatment. Autopsy-derived small intestine fragments (n = 30) were obtained at every 20 cm along the entire intestinal length. Additionally, jejunum biopsies (n = 29) were obtained during elective gastric bypass interventions from patients with (n = 10) or without T2D (n = 18). NT-expressing cells were identified by immunohistochemistry and quantified via computerized morphometric analysis. NT-expressing cell density increased along the human small intestine. NT-expressing cell density was significantly higher from 200 cm distal to the duodenojejunal flexure onward, as well as in subjects with T2D when compared to those without T2D. NT-expressing cell density increases along the human small gut, and a higher density is found in individuals with T2D. This finding suggests a potential role for NT in the mechanisms of disease and T2D improvement observed after bariatric surgery.
Collapse
Affiliation(s)
- Filipa P Ferreira
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Sofia S Pereira
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal.
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Madalena M Costa
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Marta Guimarães
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre Douro E Vouga, Santa Maria da Feira, Portugal
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2100, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Mário Nora
- Department of General Surgery, Centro Hospitalar de Entre Douro E Vouga, Santa Maria da Feira, Portugal
| | - Mariana P Monteiro
- Department of Anatomy, UMIB-Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira 228, Building 1.3, 4050-313, Porto, Portugal
- ITR-Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| |
Collapse
|
17
|
Visentin R, Brodersen K, Richelsen B, Møller N, Dalla Man C, Pedersen AK, Abrahamsen J, Holst JJ, Nielsen MF. Increased Insulin Secretion and Glucose Effectiveness in Obese Patients with Type 2 Diabetes following Bariatric Surgery. J Diabetes Res 2023; 2023:7127426. [PMID: 38020201 PMCID: PMC10663093 DOI: 10.1155/2023/7127426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Background β-cell dysfunction and insulin resistance are the main mechanisms causing glucose intolerance in type 2 diabetes (T2D). Bariatric surgeries, i.e., sleeve gastrectomy (SG) and Roux-en-Y gastric bypass (RYGB), are procedures both known to induce weight loss, increase insulin action, and enhance β-cell function, but hepatic insulin extraction and glucose effectiveness may also play a role. Methods To determine the contribution of these regulators on glucose tolerance after bariatric surgery, an oral glucose tolerance test (OGTT) was performed before and 2 months after surgery in 9 RYGB and 7 SG subjects. Eight healthy subjects served as metabolic controls. Plasma glucose, insulin, C-peptide, GLP-1, and GIP were measured during each OGTT. Insulin sensitivity and secretion, glucose effectiveness, and glucose rate of appearance were determined via oral minimal models. Results RYGB and SG resulted in similar weight reductions (13%, RYGB (p < 0.01); 14%, SG (p < 0.05)). Two months after surgery, insulin secretion (p < 0.05) and glucose effectiveness both improved equally in the two groups (11%, RYGB (p < 0.01); 8%, SG (p > 0.05)), whereas insulin sensitivity remained virtually unaltered. Bariatric surgery resulted in a comparable increase in the GLP-1 response during the OGTT, whereas GIP concentrations remained unaltered. Following surgery, oral glucose intake resulted in a comparable increase in hepatic insulin extraction, the response in both RYGB and SG patients significantly exceeding the response observed in the control subjects. Conclusions These results demonstrate that the early improvement in glucose tolerance in obese T2D after RYGB and SG surgeries is attributable mainly to increased insulin secretion and glucose effectiveness, while insulin sensitivity seems to play only a minor role. This trial is registered with NCT02713555.
Collapse
Affiliation(s)
- Roberto Visentin
- Department of Information Engineering, University of Padova, Padova, Italy
| | | | - Bjørn Richelsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital & Clinical Medicine, Aarhus University, Denmark
| | - Niels Møller
- Steno Diabetes Center Aarhus, Aarhus University Hospital & Clinical Medicine, Aarhus University, Denmark
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | | | - Jan Abrahamsen
- Department of Radiology, Viborg General Hospital, Denmark
| | - Jens Juul Holst
- Novo Nordisk Foundation, Center of Basic Metabolic Research and Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Denmark
| | | |
Collapse
|
18
|
Hernández-Montoliu L, Rodríguez-Peña MM, Puig R, Astiarraga B, Guerrero-Pérez F, Virgili N, López-Urdiales R, Osorio J, Monseny R, Lazzara C, Sobrino L, Pérez-Maraver M, Pérez-Prieto M, Pellitero S, Fernández-Veledo S, Vendrell J, Vilarrasa N. A specific gut microbiota signature is associated with an enhanced GLP-1 and GLP-2 secretion and improved metabolic control in patients with type 2 diabetes after metabolic Roux-en-Y gastric bypass. Front Endocrinol (Lausanne) 2023; 14:1181744. [PMID: 37916149 PMCID: PMC10616869 DOI: 10.3389/fendo.2023.1181744] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/22/2023] [Indexed: 11/03/2023] Open
Abstract
Objective To determine changes in incretins, systemic inflammation, intestinal permeability and microbiome modifications 12 months after metabolic RYGB (mRYGB) in patients with type 2 diabetes (T2D) and their relationship with metabolic improvement. Materials and methods Prospective single-center non-randomized controlled study, including patients with class II-III obesity and T2D undergoing mRYGB. At baseline and one year after surgery we performed body composition measurements, biochemical analysis, a meal tolerance test (MTT) and lipid test (LT) with determination of the area under the curve (AUC) for insulin, C-peptide, GLP-1, GLP-2, and fasting determinations of succinate, zonulin, IL-6 and study of gut microbiota. Results Thirteen patients aged 52.6 ± 6.5 years, BMI 39.3 ± 1.4 kg/m2, HbA1c 7.62 ± 1.5% were evaluated. After mRYGB, zonulin decreased and an increase in AUC after MTT was observed for GLP-1 (pre 9371 ± 5973 vs post 15788 ± 8021 pM, P<0.05), GLP-2 (pre 732 ± 182 vs post 1190 ± 447 ng/ml, P<0.001) and C- peptide, as well as after LT. Species belonging to Streptococaceae, Akkermansiacea, Rickenellaceae, Sutterellaceae, Enterobacteriaceae, Oscillospiraceae, Veillonellaceae, Enterobacterales_uc, and Fusobacteriaceae families increased after intervention and correlated positively with AUC of GLP-1 and GLP-2, and negatively with glucose, HbA1c, triglycerides and adiposity markers. Clostridium perfringens and Roseburia sp. 40_7 behaved similarly. In contrast, some species belonging to Lachnospiraceae, Erysipelotricaceae, and Rumnicocaceae families decreased and showed opposite correlations. Higher initial C-peptide was the only predictor for T2D remission, which was achieved in 69% of patients. Conclusions Patients with obesity and T2D submitted to mRYGB show an enhanced incretin response, a reduced gut permeability and a metabolic improvement, associated with a specific microbiota signature.
Collapse
Affiliation(s)
- Laura Hernández-Montoliu
- Department of Endocrinology and Nutrition, Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - M-Mar Rodríguez-Peña
- Hospital Universitari Joan XXIII de Tarragona, Institut d’Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rocío Puig
- Department of Endocrinology and Nutrition Hospital de la Santa Creu i Sant Pau, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Brenno Astiarraga
- Hospital Universitari Joan XXIII de Tarragona, Institut d’Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Fernando Guerrero-Pérez
- Department of Endocrinology and Nutrition, Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Nuria Virgili
- Department of Endocrinology and Nutrition, Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael López-Urdiales
- Department of Endocrinology and Nutrition, Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Osorio
- Department of General and Gastrointestinal Surgery. Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Rosa Monseny
- Clinical Nutrition Unit. Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Claudio Lazzara
- Department of General and Gastrointestinal Surgery. Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Lucía Sobrino
- Department of General and Gastrointestinal Surgery. Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Manuel Pérez-Maraver
- Department of Endocrinology and Nutrition, Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - María Pérez-Prieto
- Department of Endocrinology and Nutrition, Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Silvia Pellitero
- Department of Endocrinology and Nutrition and Health Sciences Research Institute and University Hospital Germans Trias i Pujol, Badalona, Spain
| | - Sonia Fernández-Veledo
- Hospital Universitari Joan XXIII de Tarragona, Institut d’Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Joan Vendrell
- Hospital Universitari Joan XXIII de Tarragona, Institut d’Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), Reus, Spain
| | - Nuria Vilarrasa
- Department of Endocrinology and Nutrition, Bellvitge University Hospital-Institut d'Investigació Biomédica de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
19
|
Smith K, Taylor GS, Walker M, Brunsgaard LH, Bowden Davies KA, Stevenson EJ, West DJ. Pre-Meal Whey Protein Alters Postprandial Insulinemia by Enhancing β-Cell Function and Reducing Insulin Clearance in T2D. J Clin Endocrinol Metab 2023; 108:e603-e612. [PMID: 36734166 PMCID: PMC10807909 DOI: 10.1210/clinem/dgad069] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 02/01/2023] [Indexed: 02/04/2023]
Abstract
CONTEXT Treatments that reduce postprandial glycemia (PPG) independent of stimulating insulin secretion are appealing for the management of type 2 diabetes (T2D). Consuming pre-meal whey protein (WP) reduces PPG by delaying gastric emptying and increasing plasma insulin concentrations. However, its effects on β-cell function and insulin kinetics remains unclear. OBJECTIVE To examine the PPG-regulatory effects of pre-meal WP by modeling insulin secretion rates (ISR), insulin clearance, and β-cell function. METHODS This was a single-blind, randomized, placebo-controlled, crossover design study in 18 adults with T2D (HbA1c, 56.7 ± 8.8 mmol/mol) who underwent 2 240-minute mixed-meal tolerance tests. Participants consumed WP (15 g protein) or placebo (0 g protein) 10 minutes before a mixed-macronutrient breakfast meal. PPG, pancreatic islet, and incretin hormones were measured throughout. ISR was calculated by C-peptide deconvolution. Estimates of insulin clearance and β-cell function were modeled from glucose, insulin, and ISR. Changes in PPG incremental area under the curve (iAUC; prespecified) and insulin clearance (post hoc) were measured. RESULTS β-cell function was 40% greater after WP (P = .001) and was accompanied with a -22% reduction in postprandial insulin clearance vs placebo (P < .0001). Both the peak change and PPG iAUC were reduced by WP (-1.5 mmol/L and -16%, respectively; both P < .05). Pre-meal WP augmented a 5.9-fold increase in glucagon and glucagon-like peptide 1 iAUC (both P < .0001), and a 1.5-fold increase in insulin iAUC (P < .001). Although the plasma insulin response was greater following WP, ISR was unaffected (P = .133). CONCLUSION In adults with T2D, pre-meal WP reduced PPG by coordinating an enhancement in β-cell function with a reduction in insulin clearance. This enabled an efficient postprandial insulinemic profile to be achieved without requiring further β-cell stimulation.Trial registry ISRCTN ID: ISRCTN17563146 Website link: www.isrctn.com/ISRCTN17563146.
Collapse
Affiliation(s)
- Kieran Smith
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Human Nutrition and Exercise Research Centre, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Guy S Taylor
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Human Nutrition and Exercise Research Centre, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Mark Walker
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Lise H Brunsgaard
- Health and Performance Nutrition, Arla Foods Ingredients Group P/S, Viby J 8260, Denmark
| | - Kelly A Bowden Davies
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M1 7EL, UK
| | - Emma J Stevenson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Human Nutrition and Exercise Research Centre, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Daniel J West
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Human Nutrition and Exercise Research Centre, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
20
|
Yadav J, Liang T, Qin T, Nathan N, Schwenger KJP, Pickel L, Xie L, Lei H, Winer DA, Maughan H, Robertson SJ, Woo M, Lou W, Banks K, Jackson T, Okrainec A, Hota SS, Poutanen SM, Sung HK, Allard JP, Philpott DJ, Gaisano HY. Gut microbiome modified by bariatric surgery improves insulin sensitivity and correlates with increased brown fat activity and energy expenditure. Cell Rep Med 2023; 4:101051. [PMID: 37196633 PMCID: PMC10213984 DOI: 10.1016/j.xcrm.2023.101051] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 12/20/2022] [Accepted: 04/21/2023] [Indexed: 05/19/2023]
Abstract
Alterations in the microbiome correlate with improved metabolism in patients following bariatric surgery. While fecal microbiota transplantation (FMT) from obese patients into germ-free (GF) mice has suggested a significant role of the gut microbiome in metabolic improvements following bariatric surgery, causality remains to be confirmed. Here, we perform paired FMT from the same obese patients (BMI > 40; four patients), pre- and 1 or 6 months post-Roux-en-Y gastric bypass (RYGB) surgery, into Western diet-fed GF mice. Mice colonized by FMT from patients' post-surgery stool exhibit significant changes in microbiota composition and metabolomic profiles and, most importantly, improved insulin sensitivity compared with pre-RYGB FMT mice. Mechanistically, mice harboring the post-RYGB microbiome show increased brown fat mass and activity and exhibit increased energy expenditure. Moreover, improvements in immune homeostasis within the white adipose tissue are also observed. Altogether, these findings point to a direct role for the gut microbiome in mediating improved metabolic health post-RYGB surgery.
Collapse
Affiliation(s)
- Jitender Yadav
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Tao Liang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tairan Qin
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nayanan Nathan
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Lauren Pickel
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Li Xie
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Helena Lei
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Daniel A Winer
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Susan J Robertson
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Minna Woo
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital, University Health Network, Toronto, ON, Canada; Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Wendy Lou
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Kate Banks
- Department of Comparative Medicine, University of Toronto, Toronto, ON, Canada
| | - Timothy Jackson
- Division of General Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Allan Okrainec
- Division of General Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Susy S Hota
- Department of Medicine, University of Toronto, Toronto, ON, Canada; Infection Prevention and Control, University Health Network, Toronto, ON, Canada
| | - Susan M Poutanen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Microbiology & Division of Infectious Diseases, University Health Network and Sinai Health, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Johane P Allard
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital, University Health Network, Toronto, ON, Canada.
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Herbert Y Gaisano
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Brzozowska MM, Isaacs M, Bliuc D, Baldock PA, Eisman JA, White CP, Greenfield JR, Center JR. Effects of bariatric surgery and dietary intervention on insulin resistance and appetite hormones over a 3 year period. Sci Rep 2023; 13:6032. [PMID: 37055514 PMCID: PMC10102182 DOI: 10.1038/s41598-023-33317-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/11/2023] [Indexed: 04/15/2023] Open
Abstract
To examine an impact of three types of bariatric surgery compared with dietary intervention (DIET), on concurrent changes in Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and appetite hormones over 3 years. Fifty-five adults were studied during phase of weight loss (0-12 months) and during weight stability (12-36 months) post intervention. Measurements of HOMA-IR, fasting and postprandial PYY and GLP1, adiponectin, CRP, RBP4, FGF21 hormones and dual-Xray absorptiometry were performed throughout the study. All surgical groups achieved significant reductions in HOMA-IR with greatest difference between Roux-en-Y gastric bypass and DIET (- 3.7; 95% CI - 5.4, - 2.1; p = 0.001) at 12-36 months. Initial (0-12 months) HOMA-IR values were no different to DIET after adjustment for the lost weight. During 12-36 months, after controlling for treatment procedure and weight, for every twofold increase in postprandial PYY and adiponectin, HOMA-IR decreased by 0.91 (95% CI - 1.71, - 0.11; p = 0.030) and by 0.59 (95% CI - 1.10, - 0.10; p = 0.023) respectively. Initial, non-sustained changes in RBP4 and FGF21 were not associated with HOMA-IR values. While initial rapid weight loss reduces insulin resistance, the enhanced secretions of PYY and adiponectin may contribute to weight-independent improvements in HOMA-IR during weight stability.Clinical trial registration: Australian New Zealand Clinical Trials Registry (ANZCTR): ACTRN12613000188730.
Collapse
Affiliation(s)
- Malgorzata M Brzozowska
- Endocrinology, The Sutherland Hospital, Caringbah, Australia.
- Faculty of Medicine, UNSW Sydney, Sydney, Australia.
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia.
| | - Michelle Isaacs
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Dana Bliuc
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
| | - Paul A Baldock
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| | - John A Eisman
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| | - Chris P White
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Prince of Wales Hospital, NSW Health Pathology, Randwick, Australia
- Endocrinology, Prince of Wales Hospital, Randwick, Australia
| | - Jerry R Greenfield
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - Jacqueline R Center
- Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan Institute of Medical Research, Healthy Ageing Theme, Darlinghurst, Australia
- Endocrinology, St Vincent's Hospital Sydney, Darlinghurst, Australia
- School of Medicine, The University of Notre Dame Australia, Darlinghurst, Australia
| |
Collapse
|
22
|
Hindsø M, Hedbäck N, Svane MS, Møller A, Martinussen C, Jørgensen NB, Dirksen C, Gasbjerg LS, Kristiansen VB, Hartmann B, Rosenkilde MM, Holst JJ, Madsbad S, Bojsen-Møller KN. The Importance of Endogenously Secreted GLP-1 and GIP for Postprandial Glucose Tolerance and β-Cell Function After Roux-en-Y Gastric Bypass and Sleeve Gastrectomy Surgery. Diabetes 2023; 72:336-347. [PMID: 36478039 DOI: 10.2337/db22-0568] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
Enhanced secretion of glucagon-like peptide 1 (GLP-1) seems to be essential for improved postprandial β-cell function after Roux-en-Y gastric bypass (RYGB) but is less studied after sleeve gastrectomy (SG). Moreover, the role of the other major incretin hormone, glucose-dependent insulinotropic polypeptide (GIP), is relatively unexplored after bariatric surgery. We studied the effects of separate and combined GLP-1 receptor (GLP-1R) and GIP receptor (GIPR) blockade during mixed-meal tests in unoperated (CON), SG-operated, and RYGB-operated people with no history of diabetes. Postprandial GLP-1 concentrations were highest after RYGB but also higher after SG compared with CON. In contrast, postprandial GIP concentrations were lowest after RYGB. The effect of GLP-1R versus GIPR blockade differed between groups. GLP-1R blockade reduced β-cell glucose sensitivity and increased or tended to increase postprandial glucose responses in the surgical groups but had no effect in CON. GIPR blockade reduced β-cell glucose sensitivity and increased or tended to increase postprandial glucose responses in the CON and SG groups but had no effect in the RYGB group. Our results support that GIP is the most important incretin hormone in unoperated people, whereas GLP-1 and GIP are equally important after SG, and GLP-1 is the most important incretin hormone after RYGB.
Collapse
Affiliation(s)
- Morten Hindsø
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Nora Hedbäck
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Maria S Svane
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Andreas Møller
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | | | - Nils B Jørgensen
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Carsten Dirksen
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Lærke S Gasbjerg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Viggo B Kristiansen
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital, Hvidovre, Denmark
| | | |
Collapse
|
23
|
Liu T, Zou X, Ruze R, Xu Q. Bariatric Surgery: Targeting pancreatic β cells to treat type II diabetes. Front Endocrinol (Lausanne) 2023; 14:1031610. [PMID: 36875493 PMCID: PMC9975540 DOI: 10.3389/fendo.2023.1031610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/19/2023] [Indexed: 02/17/2023] Open
Abstract
Pancreatic β-cell function impairment and insulin resistance are central to the development of obesity-related type 2 diabetes mellitus (T2DM). Bariatric surgery (BS) is a practical treatment approach to treat morbid obesity and achieve lasting T2DM remission. Traditionally, sustained postoperative glycemic control was considered a direct result of decreased nutrient intake and weight loss. However, mounting evidence in recent years implicated a weight-independent mechanism that involves pancreatic islet reconstruction and improved β-cell function. In this article, we summarize the role of β-cell in the pathogenesis of T2DM, review recent research progress focusing on the impact of Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) on pancreatic β-cell pathophysiology, and finally discuss therapeutics that have the potential to assist in the treatment effect of surgery and prevent T2D relapse.
Collapse
Affiliation(s)
- Tiantong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Xi Zou
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
24
|
Shah A, Prasad M, Mark V, Holst JJ, Laferrère B. Glucagon-like peptide-1 effect on β-cell function varies according to diabetes remission status after Roux-en-Y gastric bypass. Diabetes Obes Metab 2022; 24:2081-2089. [PMID: 35676799 PMCID: PMC9595602 DOI: 10.1111/dom.14793] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/20/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
AIMS The contribution of endogenous glucagon-like peptide (GLP)-1 to β-cell function after Roux-en-Y gastric bypass surgery (RYGB) is well established in normoglycaemic individuals, but not in those with postoperative hyperglycaemia. We, therefore, studied the effect of GLP-1 on β-cell function in individuals with varying degrees of type 2 diabetes mellitus (T2D) control after RYGB. MATERIALS AND METHODS Glucose, insulin secretion rates, β-cell glucose sensitivity and glucagon were measured during an oral glucose tolerance test before (saline only) and at 3, 12 and 24 months after RYGB with and without infusion of the GLP-1 receptor blocker exendin9-39 (EX9). The cohort was retrospectively classified based on T2D remission (REM) status at the latest study time point: REM (n = 5), persistent T2D (n = 8), or impaired glucose tolerance (n = 16). RESULTS EX9 blunted the increase in β-cell glucose sensitivity at 3 months (-44.1%, p < .001) and 12 months (-43.3%, p < .001), but not at 24 months (-12.4%, p = .243). EX9 enhanced postprandial glucagon concentrations by 62.0% at 3 months (p = .008), 46.5% at 12 months (p = .055), and 30.4% at 24 months (p = .017). EX9 counterintuitively decreased glucose concentrations at 3 months in the entire cohort (p < .001) but had no effect on glycaemia at 12 and 24 months in persistent T2D and impaired glucose tolerance; it minimally worsened glycaemia in REM at 12 months. CONCLUSIONS GLP-1 blockade reversed the improvement in β-cell function observed after RYGB, but this effect varied temporally and by REM status. GLP-1 blockade transiently and minimally worsened glycaemia only in REM, and lowered postprandial glucose values at 3 months, regardless of REM status.
Collapse
Affiliation(s)
- Ankit Shah
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Malini Prasad
- New York Obesity Nutrition Research Center, Division of Endocrinology. Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Victoria Mark
- New York Obesity Nutrition Research Center, Division of Endocrinology. Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blandine Laferrère
- Division of Endocrinology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
25
|
Beneficial effects of SGLT2 inhibitor on metabolic inflexibility and visceral fat amount in animal model of obese type 2 diabetes. Heliyon 2022; 8:e11012. [PMID: 36281369 PMCID: PMC9587290 DOI: 10.1016/j.heliyon.2022.e11012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/24/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022] Open
Abstract
Background Obesity and type 2 diabetes mellitus (T2DM) are often accompanied with a disrupted diurnal rhythm of eating and sustained anabolic state, leading to metabolic inflexibility. In the present study, we plan to investigate effects of a sodium glucose co-transporter 2 (SGLT2) inhibitor, canagliflozin (CANA), on such a metabolic inflexibility, especially on fat metabolism, in the obese type 2 diabetic rats. Materials and methods Five-week-old male SDT (Spontaneously Diabetic Torii) fatty rats as a model of obesity and T2DM and Sprague-Dawley (SD) rats were treated by either CANA (10 mg/kg) or saline (vehicle) orally for 14 days. Then, after the measurement of respiratory quotient (RQ) and visceral and subcutaneous fat volumes, rats were euthanized and blood and tissue samples were collected. Results The treatment by CANA significantly enhanced β-ketone concentration in the blood during light period in the SDT fatty rats with no effect on blood glucose concentration. The CANA treatment significantly reduced visceral fat volume in the SDT fatty rats. A diurnal rhythm of RQ was severely disrupted and persistently high throughout the day in the vehicle-treated SDT fatty rats. By the administration of CANA clearly restored the disrupted diurnal rhythm of RQ with a revival of the nadir during light period. Quantitative real-time RT-PCR revealed a significant increase of AMP-activated protein kinase and decrease of acetyl-CoA carboxylase-1 expression in the liver, and a significant increase of hormone sensitive lipase and uncoupling protein-2 expression in the white adipose tissue by the treatment of CANA in the SDT fatty rats. Conclusion CANA as a SGLT2i reduced visceral fat amount via the enhancement of fat oxidation during the light period, leading to an amelioration of metabolic inflexibility in an obese diabetic model. A novel mechanism of CANA prompts the possibility that this new class of anti-diabetic agent could be a promising anti-obesity agent as well. SGLT2i reduced visceral fat independent of its effect on urinary glucose excretion. SGLT2i increased blood ketone concentration during light period. Concomitantly, RQ during light period was decreased, reviving a diurnal RQ rhythm. Changes in relevant molecules were observed towards fat oxidation. SGLT2i effectively corrected metabolic inflexibility in an obese diabetic model.
Collapse
|
26
|
Akalestou E, Lopez-Noriega L, Christakis I, Hu M, Miras AD, Leclerc I, Rutter GA. Vertical sleeve gastrectomy normalizes circulating glucocorticoid levels and lowers glucocorticoid action tissue-selectively in mice. Front Endocrinol (Lausanne) 2022; 13:1020576. [PMID: 36246869 PMCID: PMC9556837 DOI: 10.3389/fendo.2022.1020576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Glucocorticoids produced by the adrenal cortex are essential for the maintenance of metabolic homeostasis. Glucocorticoid activation is catalysed by 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1). Excess glucocorticoids are associated with insulin resistance and hyperglycaemia. A small number of studies have demonstrated effects on glucocorticoid metabolism of bariatric surgery, a group of gastrointestinal procedures known to improve insulin sensitivity and secretion, which were assumed to result from weight loss. In this study, we hypothesize that a reduction in glucocorticoid action following bariatric surgery contributes to the widely observed euglycemic effects of the treatment. Methods Glucose and insulin tolerance tests were performed at ten weeks post operatively and circulating corticosterone was measured. Liver and adipose tissues were harvested from fed mice and 11β-HSD1 levels were measured by quantitative RT-PCR or Western (immuno-) blotting, respectively. 11β-HSD1 null mice (Hsd11b1 -/-) were generated using CRISPR/Cas9 genome editing. Wild type and littermate Hsd11b1 -/- mice underwent Vertical Sleeve Gastrectomy (VSG) or sham surgery. Results Under the conditions used, no differences in weight loss were observed between VSG treated and sham operated mice. However, both lean and obese WT VSG mice displayed significantly improved glucose clearance and insulin sensitivity. Remarkably, VSG restored physiological corticosterone production in HFD mice and reduced 11β-HSD1 expression in liver and adipose tissue post-surgery. Elimination of the 11β-HSD1/Hsd11b1 gene by CRISPR/Cas9 mimicked the effects of VSG on body weight and tolerance to 1g/kg glucose challenge. However, at higher glucose loads, the euglycemic effect of VSG was superior to Hsd11b1 elimination. Conclusions Bariatric surgery improves insulin sensitivity and reduces glucocorticoid activation at the tissular level, under physiological and pathophysiological (obesity) conditions, irrespective of weight loss. These findings point towards a physiologically relevant gut-glucocorticoid axis, and suggest that lowered glucocorticoid exposure may represent an additional contribution to the health benefits of bariatric surgery.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Livia Lopez-Noriega
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Ioannis Christakis
- Endocrine and General Surgery, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alexander D. Miras
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
27
|
Sridhar A, Khan D, Abdelaal M, Elliott JA, Naughton V, Flatt PR, Le Roux CW, Docherty NG, Moffett CR. Differential effects of RYGB surgery and best medical treatment for obesity-diabetes on intestinal and islet adaptations in obese-diabetic ZDSD rats. PLoS One 2022; 17:e0274788. [PMID: 36137097 PMCID: PMC9499270 DOI: 10.1371/journal.pone.0274788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Modification of gut-islet secretions after Roux-En-Y gastric bypass (RYBG) surgery contributes to its metabolic and anti-diabetic benefits. However, there is limited knowledge on tissue-specific hormone distribution post-RYGB surgery and how this compares with best medical treatment (BMT). In the present study, pancreatic and ileal tissues were excised from male Zucker-Diabetic Sprague Dawley (ZDSD) rats 8-weeks after RYGB, BMT (daily oral dosing with metformin 300mg/kg, fenofibrate 100mg/kg, ramipril 1mg/kg, rosuvastatin 10mg/kg and subcutaneous liraglutide 0.2mg/kg) or sham operation (laparotomy). Insulin, glucagon, somatostatin, PYY, GLP-1 and GIP expression patterns were assessed using immunocytochemistry and analyzed using ImageJ. After RYGB and BMT, body weight and plasma glucose were decreased. Intestinal morphometry was unaltered by RYGB, but crypt depth was decreased by BMT. Intestinal PYY cells were increased by both interventions. GLP-1- and GIP-cell counts were unchanged by RYGB but BMT increased ileal GLP-1-cells and decreased those expressing GIP. The intestinal contents of PYY and GLP-1 were significantly enhanced by RYGB, whereas BMT decreased ileal GLP-1. No changes of islet and beta-cell area or proliferation were observed, but the extent of beta-cell apoptosis and islet integrity calculated using circularity index were improved by both treatments. Significantly decreased islet alpha-cell areas were observed in both groups, while beta- and PYY-cell areas were unchanged. RYGB also induced a decrease in islet delta-cell area. PYY and GLP-1 colocalization with glucagon in islets was significantly decreased in both groups, while co-staining of PYY with glucagon was decreased and that with somatostatin increased. These data characterize significant cellular islet and intestinal adaptations following RYGB and BMT associated with amelioration of obesity-diabetes in ZDSD rats. The differential responses observed and particularly those within islets, may provide important clues to the unique ability of RYGB to cause diabetes remission.
Collapse
Affiliation(s)
- Ananyaa Sridhar
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
- * E-mail:
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Jessie A. Elliott
- Department of Surgery, Trinity Centre for Health Sciences and St. James’s Hospital, Dublin, Ireland
| | - Violetta Naughton
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Peter R. Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Carel W. Le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Charlotte R. Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
28
|
Gabe MBN, Gasbjerg LS, Gadgaard S, Lindquist P, Holst JJ, Rosenkilde MM. N-terminal alterations turn the gut hormone GLP-2 into an antagonist with gradual loss of GLP-2 receptor selectivity towards more GLP-1 receptor interaction. Br J Pharmacol 2022; 179:4473-4485. [PMID: 35523760 PMCID: PMC9541843 DOI: 10.1111/bph.15866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/21/2022] [Accepted: 03/17/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE To fully elucidate the regulatory role of the GLP-2 system in the gut and the bones, potent and selective GLP-2 receptor (GLP-2R) antagonists are needed. Searching for antagonist activity, we performed systematic N-terminal truncations of human GLP-2(1-33). EXPERIMENTAL APPROACH COS-7 cells were transfected with the human GLP-2R and assessed for cAMP accumulation or competition binding using 125 I-GLP-2(1-33)[M10Y]. To examine selectivity, human GLP-1 or GIP receptor expressing COS-7 cells were assessed for cAMP accumulation. KEY RESULTS The affinity for the GLP-2R of the N-terminally truncated GLP-2 peptides decreased with reduced N-terminal peptide length (Ki 6.5-871 nM), while increasing antagonism appeared with inhibitory potencies (IC50 ) values from 79 to 204 nM for truncation up to GLP-2(4-33) and then declined. In contrast, truncation-dependent increases in intrinsic activity were observed from an Emax of only 20% for GLP-(2-33) up to 46% for GLP-2(6-33) at 1 μM, followed by a decline. GLP-2(9-33) had the highest intrinsic efficacy (Emax 65%) and no antagonistic properties. Moreover, with truncations up to GLP-2(8-33) a gradual loss in selectivity for the GLP-2R appeared with increasing GLP-1 receptor (GLP-1R) inhibition (up to 73% at 1 μM). Lipidation of the peptides improved antagonism (IC50 down to 7.9 nM) for both the GLP-2R and the GLP-1R. CONCLUSION AND IMPLICATIONS The N-terminus of GLP-2 is crucial for GLP-2R activity and selectivity. Our observations form the basis for the development of tool compounds for further characterization of the GLP-2 system.
Collapse
Affiliation(s)
- Maria Buur Nordskov Gabe
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Laerke Smidt Gasbjerg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Sarina Gadgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Peter Lindquist
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
29
|
Ibacache-Saavedra P, Jerez-Mayorga D, Carretero-Ruiz A, Miranda-Fuentes C, Cano-Cappellacci M, Artero EG. Effects of bariatric surgery on cardiorespiratory fitness: A systematic review and meta-analysis. Obes Rev 2022; 23:e13408. [PMID: 34927337 DOI: 10.1111/obr.13408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/20/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022]
Abstract
Although bariatric surgery (BS) is recognized as an effective strategy for body weight loss, its impact on cardiorespiratory fitness (CRF) is still unclear. We aimed to examine postoperative changes in CRF (VO2max/peak ) and its relationship with weight loss among adults undergoing BS. We systematically searched the WoS, PubMed, MEDLINE, and Scopus databases. Observational and intervention studies were selected reporting the presurgery and postsurgery CRF, measured by breath-by-breath VO2 or its estimation. Eleven articles (312 patients) revealed that BS leads to a reduction in absolute VO2max/peak in the short term (effect size, ES = -0.539; 95%CI = -0.708, -0.369; p < 0.001), and those patients who suffered a more significant decrease in BMI after BS also had a greater loss of absolute VO2max/peak . However, VO2max/peak relative to body weight increased after surgery (ES = 0.658; 95%CI = 0.473, 0.842; p < 0.001). An insufficient number of studies were found investigating medium and long-term changes in CRF after BS. This study provides moderate-quality evidence that the weight loss induced by BS can reduce CRF in the short term, which represents a therapeutic target to optimize BS outcomes. More high-quality studies are needed to evaluate the impact of BS on VO2max/peak in the short, medium, and long term including normalized values for fat-free mass.
Collapse
Affiliation(s)
- Paulina Ibacache-Saavedra
- Exercise and Rehabilitation Sciences Laboratory, School of Physical Therapy, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago, Chile
| | - Daniel Jerez-Mayorga
- Exercise and Rehabilitation Sciences Laboratory, School of Physical Therapy, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago, Chile
| | - Alejandro Carretero-Ruiz
- Department of Education and SPORT Research Group (CTS-1024), CERNEP Research Center, University of Almería, Almeria, Spain
| | - Claudia Miranda-Fuentes
- Exercise and Rehabilitation Sciences Laboratory, School of Physical Therapy, Faculty of Rehabilitation Sciences, Universidad Andres Bello, Santiago, Chile
| | | | - Enrique G Artero
- Department of Education and SPORT Research Group (CTS-1024), CERNEP Research Center, University of Almería, Almeria, Spain
| |
Collapse
|
30
|
Prasad M, Mark V, Ligon C, Dutia R, Nair N, Shah A, Laferrère B. Role of the Gut in the Temporal Changes of β-Cell Function After Gastric Bypass in Individuals With and Without Diabetes Remission. Diabetes Care 2022; 45:469-476. [PMID: 34857533 PMCID: PMC8914419 DOI: 10.2337/dc21-1270] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/10/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The role of the gut in diabetes remission after Roux-en-Y gastric bypass (RYGB) is incompletely understood. We assessed the temporal change in insulin secretory capacity after RYGB, using oral and intravenous (IV) glucose, in individuals with type 2 diabetes. RESEARCH DESIGN AND METHODS Longitudinal, prospective measures of β-cell function were assessed after oral glucose intake and graded glucose infusion in individuals with severe obesity and diabetes studied at 0, 3 (n = 29), 12 (n = 24), and 24 (n = 20) months after RYGB. Data were collected between 2015 and 2019 in an academic clinical research center. RESULTS The decreases in body weight, fat mass, waist circumference, and insulin resistance after surgery (all P < 0.001 at 12 and 24 months) did not differ according to diabetes remission status. In contrast, both the magnitude and temporal changes in β-cell glucose sensitivity after oral glucose intake differed by remission status (P = 0.04): greater (6.5-fold; P < 0.01) and sustained in those in full remission, moderate and not sustained past 12 months in those with partial remission (3.3-fold; P < 0.001), and minimal in those not experiencing remission (2.7-fold; P = not significant). The improvement in β-cell function after IV glucose administration was not apparent until 12 months, significant only in those in full remission, and only ∼33% of that observed after oral glucose intake. Preintervention β-cell function and its change after surgery predicted remission; weight loss and insulin sensitivity did not. CONCLUSIONS Our data show the time course of changes in β-cell function after RYGB. The improvement in β-cell function after RYGB, but not changes in weight loss or insulin sensitivity, drives diabetes remission.
Collapse
Affiliation(s)
- Malini Prasad
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY
| | - Victoria Mark
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY
| | - Chanel Ligon
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Roxanne Dutia
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY
| | - Nandini Nair
- Division of Endocrinology, Columbia University Irving Medical Center, New York, NY
| | - Ankit Shah
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| | - Blandine Laferrère
- New York Nutrition Obesity Research Center, Columbia University Irving Medical Center, New York, NY.,Department of Medicine, Columbia University Irving Medical Center, New York, NY.,Division of Endocrinology, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
31
|
Fatima F, Hjelmesæth J, Birkeland KI, Gulseth HL, Hertel JK, Svanevik M, Sandbu R, Småstuen MC, Hartmann B, Holst JJ, Hofsø D. Gastrointestinal Hormones and β-Cell Function After Gastric Bypass and Sleeve Gastrectomy: A Randomized Controlled Trial (Oseberg). J Clin Endocrinol Metab 2022; 107:e756-e766. [PMID: 34463768 DOI: 10.1210/clinem/dgab643] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Indexed: 02/04/2023]
Abstract
CONTEXT Whether Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) differentially affect postprandial gastrointestinal hormones and β-cell function in type 2 diabetes remains unclear. OBJECTIVE We aimed to compare gastrointestinal hormones and β-cell function, assessed by an oral glucose tolerance test (OGTT) 5 weeks and 1 year after surgery, hypothesizing higher glucagon-like peptide-1 (GLP-1) levels and greater β-cell response to glucose after RYGB than after SG. METHODS This study was a randomized, triple-blind, single-center trial at a tertiary care center in Norway. The primary outcomes were diabetes remission and IVGTT-derived β-cell function. Participants with obesity and type 2 diabetes were allocated (1:1) to RYGB or SG. We measured gastrointestinal hormone profiles and insulin secretion as β-cell glucose sensitivity (β-GS) derived from 180-minute OGTTs. RESULTS Participants were 106 patients (67% women), mean (SD) age 48 (10) years. Diabetes remission rates at 1 year were higher after RYGB than after SG (77% vs 48%; P = 0.002). Incremental area under the curve (iAUC0-180) GLP-1 and β-GS increased more after RYGB than after SG, with 1-year between-group difference 1173 pmol/L*min (95% CI, 569-1776; P = 0.0010) and 0.45 pmol/kg/min/mmol (95% CI, 0.15-0.75; P = 0.0032), respectively. After surgery, fasting and postprandial ghrelin levels were higher and decremental AUC0-180 ghrelin, iAUC0-180 glucose-dependent insulinotropic polypeptide, and iAUC0-60 glucagon were greater after RYGB than after SG. Diabetes remission at 1 year was associated with higher β-GS and higher GLP-1 secretion. CONCLUSION RYGB was associated with greater improvement in β-cell function and higher postprandial GLP-1 levels than SG.
Collapse
Affiliation(s)
- Farhat Fatima
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
| | - Jøran Hjelmesæth
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Kåre Inge Birkeland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
- Department of Transplantation, Oslo University Hospital, 0424 Oslo, Norway
| | - Hanne Løvdal Gulseth
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, 0213 Oslo, Norway
| | | | - Marius Svanevik
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Surgery, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| | - Rune Sandbu
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Surgery, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| | - Milada Cvancarova Småstuen
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
- Department of Nutrition and Management, Oslo Metropolitan University, 0130 Oslo, Norway
| | - Bolette Hartmann
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens Juul Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Dag Hofsø
- Morbid Obesity Centre, Vestfold Hospital Trust, 3103 Tønsberg, Norway
| |
Collapse
|
32
|
Evans RM, Wei Z. Interorgan crosstalk in pancreatic islet function and pathology. FEBS Lett 2022; 596:607-619. [PMID: 35014695 DOI: 10.1002/1873-3468.14282] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022]
Abstract
Pancreatic β cells secrete insulin in response to glucose, a process that is regulated at multiple levels, including a network of input signals from other organ systems. Impaired islet function contributes to the pathogenesis of type 2 diabetes mellitus (T2DM), and targeting inter-organ communications, such as GLP-1 signalling, to enhance β-cell function has been proven to be a successful therapeutic strategy in the last decade. In this review, we will discuss recent advances in inter-organ communication from the metabolic, immune and neural system to pancreatic islets, their biological implication in normal pancreas endocrine function and their role in the (mal)adaptive responses of islet to nutrition-induced stress.
Collapse
Affiliation(s)
- Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zong Wei
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, USA
| |
Collapse
|
33
|
Miedzybrodzka EL, Gribble FM, Reimann F. Targeting the Enteroendocrine System for Treatment of Obesity. Handb Exp Pharmacol 2022; 274:487-513. [PMID: 35419620 DOI: 10.1007/164_2022_583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mimetics of the anorexigenic gut hormone glucagon-like peptide 1 (GLP-1) were originally developed as insulinotropic anti-diabetic drugs but also evoke significant weight loss, leading to their recent approval as obesity therapeutics. Co-activation of receptors for GLP-1 and other gut hormones which reduce food intake - peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP) - is now being explored clinically to enhance efficacy. An alternative approach involves pharmacologically stimulating endogenous secretion of these hormones from enteroendocrine cells (EECs) to recapitulate the metabolic consequences of bariatric surgery, where highly elevated postprandial levels of GLP-1 and PYY3-36 are thought to contribute to improved glycaemia and weight loss.
Collapse
Affiliation(s)
- Emily L Miedzybrodzka
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
34
|
Gasbjerg LS, Bari EJ, Christensen M, Knop FK. Exendin(9-39)NH 2 : Recommendations for clinical use based on a systematic literature review. Diabetes Obes Metab 2021; 23:2419-2436. [PMID: 34351033 DOI: 10.1111/dom.14507] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/25/2022]
Abstract
AIM To present an overview of exendin(9-39)NH2 usage as a scientific tool in humans and provide recommendations for dosage and infusion regimes. METHODS We systematically searched the literature on exendin(9-39)NH2 and included for review 44 clinical studies reporting use of exendin(9-39)NH2 in humans. RESULTS Exendin(9-39)NH2 binds to the orthosteric binding site of the glucagon-like peptide-1 (GLP-1) receptor with high affinity. The plasma elimination half-life of exendin(9-39)NH2 after intravenous administration is ~30 minutes, requiring ~2.5 hours of constant infusion before steady-state plasma concentrations can be expected. Studies utilizing infusions with exendin(9-39)NH2 in humans have applied varying regimens (priming with a bolus or constant infusion) and dosages (continuous infusion rate range 30-900 pmol/kg/min) with subsequent differences in effects. Administration of exendin(9-39)NH2 in healthy individuals, patients with diabetes, obese patients, and patients who have undergone bariatric surgery significantly increases fasting and postprandial levels of glucose and glucagon, but has inconsistent effects on circulating concentrations of insulin and C-peptide, gastric emptying, appetite sensations, and food intake. Importantly, exendin(9-39)NH2 induces secretion of all L cell products (ie, in addition to GLP-1, also peptide YY, glucagon-like peptide-2, oxyntomodulin, and glicentin) complicating use of exendin(9-39)NH2 as a tool to study the isolated effect of GLP-1. CONCLUSIONS Exendin(9-39)NH2 is selective for the GLP-1 receptor, with numerous and complex whole-body effects. To obtain GLP-1 receptor blockade in humans, we recommend an initial high-dose infusion, followed by a continuous infusion rate aiming at a ratio of exendin(9-39)NH2 to GLP-1 of 2000:1. Highlights Exendin(9-39)NH2 is a competitive antagonist of the human GLP-1 receptor. Exendin(9-39)NH2 has been used as a tool to delineate human GLP-1 physiology since 1998. Exendin(9-39)NH2 induces secretion of GLP-1 and other L cell products. Reported effects of exendin(9-39)NH2 on insulin levels and food intake are inconsistent. Here, we provide recommendations for the use of exendin(9-39)NH2 in clinical studies.
Collapse
Affiliation(s)
- Laerke Smidt Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Johanning Bari
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Mikkel Christensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip Krag Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Centre Copenhagen, Gentofte, Denmark
| |
Collapse
|
35
|
GLP-1: 10-year follow-up after Roux-en-Y gastric bypass. Langenbecks Arch Surg 2021; 407:559-568. [PMID: 34651238 DOI: 10.1007/s00423-021-02341-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 09/23/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE Glucagon-like peptide-1 (GLP-1) is a hormone widely studied in the short-term postoperative follow-up of Roux-en-Y gastric bypass due to its elevation and association with improvement of the glucose metabolism, but there are few studies in 10 years after RYGB follow-up with the same patient. METHODS Twenty morbidity obesity patients were submitted to RYGB; these patients were divided into two groups: normal glucose-tolerant morbidly obese patients (NGT) 11 patients and abnormal glucose metabolism morbidly obese patients (AGM) 9 patients. Oral glucose tolerance test (OGTT) was done during four different periods: T1 (first evaluation), T2 (pre-surgery), T3 (9 months after surgery) and T4 (10 years after surgery). RESULTS Groups were matched for age and gender, and as NGT and AGM had BMI of 46.31 ± 5.03 kg/m2 and 50.87 ± 10.31 kg/m2. After 10 years of RYGB, they were obesity grade I with BMI for NGT 32.45 ± 4.99 kg/m2 and AGM 34.85 ± 4.46 kg/m2. Plasma glucose levels decreased NGT group at T4 period had a significant reduction at 120 min after OGTT for NGT 55.49 ± 17.15 mg/dL (p˂0.001). Insulin levels changed from T1 to T4 for the NGT group. GLP-1 curves were statistically different between the NGT and AGM groups. The AGM group had a higher mean for GLP-1 secretion at T4 period and at 30 min of OGTT 63.85 ± 37.98 pmol/L when compared to NGT 50.73 ± 24.82 pmol/L with AGM > NGT with p˂0.001. CONCLUSION Evaluation of the same patient during 4 different periods shows that, even with weight regain, after 10-years of RYGB high levels of GLP-1 remained which can be associated with metabolic improvement especially at the NGT group.
Collapse
|
36
|
A pilot study about the development and characterization of a Roux en Y gastric bypass model in obese Yucatan minipigs. Sci Rep 2021; 11:20190. [PMID: 34642370 PMCID: PMC8511153 DOI: 10.1038/s41598-021-98575-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 07/21/2021] [Indexed: 11/30/2022] Open
Abstract
Performing the Roux-en-Y gastric bypass (RYGBP) in obese Yucatan minipigs provides an opportunity to explore the mechanisms behind the effects of this surgery in controlled environmental and nutritional conditions. We hypothesized that RYGBP in these minipigs would induce changes at multiple levels, as in obese humans. We sought to characterize RYGBP in a diet-induced obese minipig model, compared with a pair-fed sham group. After inducing obesity with an ad libitum high-fat/high-sugar diet, we performed RYGBP (n = 7) or sham surgery (n = 6). Oral glucose tolerance tests (OGTT) were performed before and after surgery. Histological analyses were conducted to compare the alimentary limb at sacrifice with tissue sampled during RYGBP surgery. One death occurred in the RYGBP group at postoperative day (POD) 3. Before sacrifice, weight loss was the same across groups. GLP-1 secretion (OGTT) was significantly higher at 15, 30 and 60 min at POD 7, and at 30 and 60 min at POD 30 in the RYGBP group. Incremental insulin area under the curve increased significantly after RYGBP (p = 0.02). RYGBP induced extensive remodeling of the alimentary limb. Results show that RYGBP can be safely performed in obese minipigs, and changes mimic those observed in humans.
Collapse
|
37
|
Martinussen C, Svane MS, Bojsen-Møller KN, Jensen CZ, Kristiansen VB, Bookout AL, Jørgensen SB, Holst JJ, Wewer Albrechtsen NJ, Madsbad S, Kuhre RE. Plasma GDF15 levels are similar between subjects after bariatric surgery and matched controls and are unaffected by meals. Am J Physiol Endocrinol Metab 2021; 321:E443-E452. [PMID: 34370594 DOI: 10.1152/ajpendo.00190.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Growth differentiating factor 15 (GDF15) is expressed in the intestine and is one of the most recently identified satiety peptides. The mechanisms controlling its secretion are unclear. The present study investigated whether plasma GDF15 concentrations are meal-related and if potential responses depend on macronutrient type or are affected by previous bariatric surgery. The study included 1) volunteers ingesting rapidly vs. slowly digested carbohydrates (sucrose vs. isomaltose; n = 10), 2) volunteers who had undergone Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG) surgery and unoperated matched controls ingesting a liquid mixed meal (n = 9-10 in each group), and 3) individuals with previous RYGB compared with unoperated controls ingesting isocaloric glucose, fat, or protein (n = 6 in each group). Plasma was collected after an overnight fast and up to 6 h after ingestion (≥12 time points). In cohort 1, fasting GDF15 concentrations were ∼480 pg/mL. Concentrations after sucrose or isomaltose intake did not differ from baseline (P = 0.26 to P > 0.99) and total area under the curves (tAUCs were similar between groups (P = 0.77). In cohort 2, fasting GDF15 concentrations were as follows (pg/mL): RYGB = 540 ± 41.4, SG = 477 ± 36.4, and controls = 590 ± 41.8, with no between-group differences (P = 0.73). Concentrations did not increase at any postprandial time point (over all time factor: P = 0.10) and tAUCs were similar between groups (P = 0.73). In cohort 3, fasting plasma GDF15 was similar among the groups (P > 0.99) and neither glucose, fat, nor protein intake consistently increased the concentrations. In conclusion, we find that plasma GDF15 was not stimulated by meal intake and that fasting concentrations did not differ between RYGB-, SG-, and body mass index (BMI)-matched controls when investigated during the weight stable phase after RYGB and SG.NEW & NOTEWORTHY Our combined data show that GDF15 does not increase in response to a liquid meal. Moreover, we show for the first time that ingestion of sucrose, isomaltose, glucose, fat, or protein also does not increase plasma GDF15 concentrations, questioning the role of GDF15 in regulation of food source preference. Finally, we find that neither fasting nor postprandial plasma GDF15 concentrations are increased in individuals with previous bariatric surgery compared with unoperated body mass index (BMI)-matched controls.
Collapse
Affiliation(s)
- Christoffer Martinussen
- Department of Endocrinology, Hvidovre Hospital, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Saur Svane
- Department of Endocrinology, Hvidovre Hospital, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department for Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rune Ehrenreich Kuhre
- Obesity Pharmacology, Novo Nordisk, Måløv, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Smith KR, Moran TH. Gastrointestinal peptides in eating-related disorders. Physiol Behav 2021; 238:113456. [PMID: 33989649 PMCID: PMC8462672 DOI: 10.1016/j.physbeh.2021.113456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/19/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Food intake is tightly controlled by homeostatic signals sensitive to metabolic need for the regulation of body weight. This review focuses on the peripherally-secreted gastrointestinal peptides (i.e., ghrelin, cholecystokinin, glucagon-like peptide 1, and peptide tyrosine tyrosine) that contribute to the control of appetite and discusses how these peptides or the signals arising from their release are disrupted in eating-related disorders across the weight spectrum, namely anorexia nervosa, bulimia nervosa, and obesity, and whether they are normalized following weight restoration or weight loss treatment. Further, the role of gut peptides in the pathogenesis and treatment response in human weight conditions as identified by rodent models are discussed. Lastly, we review the incretin- and hormone-based pharmacotherapies available for the treatment of obesity and eating-related disorders.
Collapse
Affiliation(s)
- Kimberly R Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States.
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
39
|
Akalestou E, Suba K, Lopez-Noriega L, Georgiadou E, Chabosseau P, Gallie A, Wretlind A, Legido-Quigley C, Leclerc I, Salem V, Rutter GA. Intravital imaging of islet Ca 2+ dynamics reveals enhanced β cell connectivity after bariatric surgery in mice. Nat Commun 2021; 12:5165. [PMID: 34453049 PMCID: PMC8397709 DOI: 10.1038/s41467-021-25423-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/06/2021] [Indexed: 11/25/2022] Open
Abstract
Bariatric surgery improves both insulin sensitivity and secretion and can induce diabetes remission. However, the mechanisms and time courses of these changes, particularly the impact on β cell function, are difficult to monitor directly. In this study, we investigated the effect of Vertical Sleeve Gastrectomy (VSG) on β cell function in vivo by imaging Ca2+ dynamics in islets engrafted into the anterior eye chamber. Mirroring its clinical utility, VSG in mice results in significantly improved glucose tolerance, and enhanced insulin secretion. We reveal that these benefits are underpinned by augmented β cell function and coordinated activity across the islet. These effects involve changes in circulating GLP-1 levels which may act both directly and indirectly on the β cell, in the latter case through changes in body weight. Thus, bariatric surgery leads to time-dependent increases in β cell function and intra-islet connectivity which are likely to contribute to diabetes remission.
Collapse
Affiliation(s)
- Elina Akalestou
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Kinga Suba
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Livia Lopez-Noriega
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Eleni Georgiadou
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Pauline Chabosseau
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Alasdair Gallie
- grid.413629.b0000 0001 0705 4923Central Biological Services (CBS) Hammersmith Hospital Campus, London, UK
| | - Asger Wretlind
- grid.419658.70000 0004 0646 7285Systems Medicine, Steno Diabetes Center, Gentofte, Copenhagen, Denmark
| | - Cristina Legido-Quigley
- grid.419658.70000 0004 0646 7285Systems Medicine, Steno Diabetes Center, Gentofte, Copenhagen, Denmark
| | - Isabelle Leclerc
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Victoria Salem
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK ,grid.413629.b0000 0001 0705 4923Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Guy A. Rutter
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK ,grid.59025.3b0000 0001 2224 0361Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore, Singapore ,grid.14848.310000 0001 2292 3357Centre de Recherches du CHUM, University of Montreal, Montreal, QC Canada
| |
Collapse
|
40
|
Akkus G, Tetiker T. Which predictors could effect on remission of type 2 diabetes mellitus after the metabolic surgery: A general perspective of current studies? World J Diabetes 2021; 12:1312-1324. [PMID: 34512896 PMCID: PMC8394232 DOI: 10.4239/wjd.v12.i8.1312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/18/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The alarming rise in the worldwide prevalence of obesity is paralleled by an increasing burden of type 2 diabetes mellitus (T2DM). Metabolic surgery is the most effective means of obtaining substantial and durable weight loss in individual obese patients with T2DM. There are randomized trials that justify the inclusion of metabolic surgery into the treatment algorithm for patients with T2DM, but remission rates of T2DM after metabolic surgery can display great variability. AIM To discuss the most commonly used surgical options including vertical sleeve gastrectomy, adjustable gastric banding, Roux-en-Y gastric bypass, and biliopancreatic diversion with duodenal switch. METHODS We also report from observational and randomized controlled studies on rate of remission of T2DM after the surgical procedures. RESULTS In light of the recent findings, metabolic surgery is a safe and effective treatment option for obese patient with T2DM, but further studies are needed to clarify better the rate of diabetes remission. CONCLUSION In light of the recent findings, metabolic surgery is a safe and effective treatment option for obese patients with T2DM, but further studies are needed to clarify better the rate of diabetes remission.
Collapse
Affiliation(s)
- Gamze Akkus
- Department of Internal Medicine, Cukurova University Medical Faculty, Adana 33170, Turkey
- Department of Endocrinology, Cukurova University, Adana 33170, Turkey
| | - Tamer Tetiker
- Department of Internal Medicine, Cukurova University Medical Faculty, Adana 33170, Turkey
- Department of Endocrinology, Cukurova University, Adana 33170, Turkey
| |
Collapse
|
41
|
Holst JJ, Gasbjerg LS, Rosenkilde MM. The Role of Incretins on Insulin Function and Glucose Homeostasis. Endocrinology 2021; 162:6199910. [PMID: 33782700 PMCID: PMC8168943 DOI: 10.1210/endocr/bqab065] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Indexed: 12/14/2022]
Abstract
The incretin effect-the amplification of insulin secretion after oral vs intravenous administration of glucose as a mean to improve glucose tolerance-was suspected even before insulin was discovered, and today we know that the effect is due to the secretion of 2 insulinotropic peptides, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). But how important is it? Physiological experiments have shown that, because of the incretin effect, we can ingest increasing amounts of amounts of glucose (carbohydrates) without increasing postprandial glucose excursions, which otherwise might have severe consequences. The mechanism behind this is incretin-stimulated insulin secretion. The availability of antagonists for GLP-1 and most recently also for GIP has made it possible to directly estimate the individual contributions to postprandial insulin secretion of a) glucose itself: 26%; b) GIP: 45%; and c) GLP-1: 29%. Thus, in healthy individuals, GIP is the champion. When the action of both incretins is prevented, glucose tolerance is pathologically impaired. Thus, after 100 years of research, we now know that insulinotropic hormones from the gut are indispensable for normal glucose tolerance. The loss of the incretin effect in type 2 diabetes, therefore, contributes greatly to the impaired postprandial glucose control.
Collapse
Affiliation(s)
- Jens Juul Holst
- Department of Biomedical Sciences and the NovoNordisk Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, The Panum Institute, Copenhagen N, DK-2200 Denmark
- Correspondence: Jens Juul Holst, MD, University of Copenhagen, Department of Biomedical Sciences, The Panum Institute, 3 Blegdamsvej, Copenhagen, DK-2200 Denmark.
| | - Lærke Smidt Gasbjerg
- Department of Biomedical Sciences and the NovoNordisk Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, The Panum Institute, Copenhagen N, DK-2200 Denmark
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences and the NovoNordisk Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, The Panum Institute, Copenhagen N, DK-2200 Denmark
| |
Collapse
|
42
|
Arora T, Vanslette AM, Hjorth SA, Bäckhed F. Microbial regulation of enteroendocrine cells. MED 2021; 2:553-570. [DOI: 10.1016/j.medj.2021.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 02/08/2023]
|
43
|
Abstract
PURPOSE OF REVIEW Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease in the United States and increasing globally. The progressive form of NAFLD, nonalcoholic steatohepatitis (NASH), can lead to cirrhosis and complications of end-stage liver disease. No FDA-approved therapy for NAFLD/NASH exists. Treatment of NAFLD/NASH includes effective and sustained life-style modification and weight loss. This review reports on the recent findings of bariatric surgery in the management of NASH. RECENT FINDINGS NAFLD, at all stages, is common in those who meet indication for bariatric surgery. Bariatric surgery resolves NAFLD/NASH and reverses early stages of fibrosis. Although randomized controlled trials of bariatric surgery in NASH are infeasible, studies defining the metabolic changes induced by bariatric surgery, and their effect on NASH, provide insight for plausible pharmacologic targets for the nonsurgical treatment of NASH. SUMMARY Resolution of NASH and fibrosis regression can occur after bariatric surgery. Although the exact mechanism(s) underlying the improvement of NASH and hepatic fibrosis following bariatric surgery is not fully elucidated, emerging data on this topic is vitally important for lending insight into the pharmacotherapies for NASH for patients who are not otherwise suitable candidates for bariatric surgery.
Collapse
|
44
|
Lu VB, Gribble FM, Reimann F. Nutrient-Induced Cellular Mechanisms of Gut Hormone Secretion. Nutrients 2021; 13:nu13030883. [PMID: 33803183 PMCID: PMC8000029 DOI: 10.3390/nu13030883] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/27/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract can assess the nutrient composition of ingested food. The nutrient-sensing mechanisms in specialised epithelial cells lining the gastrointestinal tract, the enteroendocrine cells, trigger the release of gut hormones that provide important local and central feedback signals to regulate nutrient utilisation and feeding behaviour. The evidence for nutrient-stimulated secretion of two of the most studied gut hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), along with the known cellular mechanisms in enteroendocrine cells recruited by nutrients, will be the focus of this review. The mechanisms involved range from electrogenic transporters, ion channel modulation and nutrient-activated G-protein coupled receptors that converge on the release machinery controlling hormone secretion. Elucidation of these mechanisms will provide much needed insight into postprandial physiology and identify tractable dietary approaches to potentially manage nutrition and satiety by altering the secreted gut hormone profile.
Collapse
|
45
|
Do Gut Hormones Contribute to Weight Loss and Glycaemic Outcomes after Bariatric Surgery? Nutrients 2021; 13:nu13030762. [PMID: 33652862 PMCID: PMC7996890 DOI: 10.3390/nu13030762] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery is an effective intervention for management of obesity through treating dysregulated appetite and achieving long-term weight loss maintenance. Moreover, significant changes in glucose homeostasis are observed after bariatric surgery including, in some cases, type 2 diabetes remission from the early postoperative period and postprandial hypoglycaemia. Levels of a number of gut hormones are dramatically increased from the early period after Roux-en-Y gastric bypass and sleeve gastrectomy—the two most commonly performed bariatric procedures—and they have been suggested as important mediators of the observed changes in eating behaviour and glucose homeostasis postoperatively. In this review, we summarise the current evidence from human studies on the alterations of gut hormones after bariatric surgery and their impact on clinical outcomes postoperatively. Studies which assess the role of gut hormones after bariatric surgery on food intake, hunger, satiety and glucose homeostasis through octreotide use (a non-specific inhibitor of gut hormone secretion) as well as with exendin 9–39 (a specific glucagon-like peptide-1 receptor antagonist) are reviewed. The potential use of gut hormones as biomarkers of successful outcomes of bariatric surgery is also evaluated.
Collapse
|
46
|
Keshavjee SH, Schwenger KJP, Yadav J, Jackson TD, Okrainec A, Allard JP. Factors Affecting Metabolic Outcomes Post Bariatric Surgery: Role of Adipose Tissue. J Clin Med 2021; 10:714. [PMID: 33670215 PMCID: PMC7916950 DOI: 10.3390/jcm10040714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is an ever-growing public health crisis, and bariatric surgery (BS) has become a valuable tool in ameliorating obesity, along with comorbid conditions such as diabetes, dyslipidemia and hypertension. BS techniques have come a long way, leading to impressive improvements in the health of the majority of patients. Unfortunately, not every patient responds optimally to BS and there is no method that is sufficient to pre-operatively predict who will receive maximum benefit from this surgical intervention. This review focuses on the adipose tissue characteristics and related parameters that may affect outcomes, as well as the potential influences of insulin resistance, BMI, age, psychologic and genetic factors. Understanding the role of these factors may help predict who will benefit the most from BS.
Collapse
Affiliation(s)
- Sara H. Keshavjee
- Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA;
| | - Katherine J. P. Schwenger
- Division of Gastroenterology, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada;
| | - Jitender Yadav
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Timothy D. Jackson
- Division of General Surgery, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; (T.D.J.); (A.O.)
| | - Allan Okrainec
- Division of General Surgery, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; (T.D.J.); (A.O.)
| | - Johane P. Allard
- Division of Gastroenterology, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada;
| |
Collapse
|
47
|
Hindsø M, Svane MS, Hedbäck N, Holst JJ, Madsbad S, Bojsen-Møller KN. The role of GLP-1 in postprandial glucose metabolism after bariatric surgery: a narrative review of human GLP-1 receptor antagonist studies. Surg Obes Relat Dis 2021; 17:1383-1391. [PMID: 33771461 DOI: 10.1016/j.soard.2021.01.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/22/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022]
Abstract
The Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) bariatric procedures lead to remission or improvement of type 2 diabetes. A weight loss-independent augmentation of postprandial insulin secretion contributes to the improvement in glycemic control after RYGB and is associated with a ∼10-fold increase in plasma concentrations of the incretin hormone glucagon-like peptide-1 (GLP-1). However, the physiologic importance of the markedly increased postprandial GLP-1 secretion after RYGB has been much debated. The effect of GLP-1 receptor blockade after RYGB has been investigated in 12 studies. The studies indicate a shift toward a more prominent role for GLP-1 in postprandial β-cell function after RYGB. The effect of GLP-1 receptor antagonism on glucose tolerance after RYGB is more complex and is associated with important methodological challenges. The postprandial GLP-1 response is less enhanced after SG compared with RYGB. However, the effect of GLP-1 receptor blockade after SG has been examined in 1 study only and needs further investigation.
Collapse
Affiliation(s)
- Morten Hindsø
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.
| | - Maria S Svane
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Nora Hedbäck
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, University of Copenhagen and Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | | |
Collapse
|
48
|
Abstract
Glucagon like peptide-1 (GLP-1), a peptide hormone from the intestinal tract, plays a central role in the coordination of postprandial glucose homeostasis through actions on insulin secretion, food intake and gut motility. GLP-1 forms the basis for a variety of current drugs for the treatment of type 2 diabetes and obesity, as well as new agents currently being developed. Here, we provide a concise overview of the core physiology of GLP-1 secretion and action, and the role of the peptide in human health, disease and therapeutics.
Collapse
Affiliation(s)
- Fiona M Gribble
- Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - Frank Reimann
- Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
49
|
Miskelly MG, Shcherbina L, Thorén Fischer AH, Abels M, Lindqvist A, Wierup N. GK-rats respond to gastric bypass surgery with improved glycemia despite unaffected insulin secretion and beta cell mass. Peptides 2021; 136:170445. [PMID: 33197511 DOI: 10.1016/j.peptides.2020.170445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Roux-en-Y gastric bypass (RYGB) is the most effective treatment for morbid obesity and results in rapid remission of type 2 diabetes (T2D), before significant weight loss occurs. The underlying mechanisms for T2D remission are not fully understood. To gain insight into these mechanisms we used RYGB-operated diabetic GK-rats and Wistar control rats. Twelve adult male Wistar- and twelve adult male GK-rats were subjected to RYGB- or sham-operation. Oral glucose tolerance tests (OGTT) were performed six weeks after surgery. RYGB normalized fasting glucose levels in GK-rats, without affecting fasting insulin levels. In both rat strains, RYGB caused increased postprandial responses in glucose, GLP-1, and GIP. RYGB caused elevated postprandial insulin secretion in Wistar-rats, but had no effect on insulin secretion in GK-rats. In agreement with this, RYGB improved HOMA-IR in GK-rats, but had no effect on HOMA-β. RYGB-operated GK-rats had an increased number of GIP receptor and GLP-1 receptor immunoreactive islet cells, but RYGB had no major effect on beta or alpha cell mass. Furthermore, in RYGB-operated GK-rats, increased Slc5a1, Pck2 and Pfkfb1 and reduced Fasn hepatic mRNA expression was observed. In summary, our data shows that RYGB induces T2D remission and enhanced postprandial incretin hormone secretion in GK-rats, without affecting insulin secretion or beta cell mass. Thus our data question the dogmatic view of how T2D remission is achieved and instead point at improved insulin sensitivity as the main mechanism of remission.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/surgery
- Disease Models, Animal
- Gastric Bypass
- Gastric Inhibitory Polypeptide/genetics
- Glucagon-Like Peptide 1/genetics
- Glucose Tolerance Test
- Humans
- Insulin/genetics
- Insulin/metabolism
- Insulin Secretion/genetics
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Obesity, Morbid/genetics
- Obesity, Morbid/metabolism
- Obesity, Morbid/pathology
- Obesity, Morbid/surgery
- Rats
- Rats, Wistar
- Weight Loss/genetics
- Weight Loss/physiology
Collapse
Affiliation(s)
- Michael G Miskelly
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | - Liliya Shcherbina
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | | | - Mia Abels
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | - Andreas Lindqvist
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden
| | - Nils Wierup
- Neuroendocrine Cell Biology, Lund University Diabetes Centre, Malmö, Sweden.
| |
Collapse
|
50
|
van Duinkerken E, Bernardes G, van Bloemendaal L, Veltman DJ, Barkhof F, Mograbi DC, Gerdes VEA, Deacon CF, Holst JJ, Drent ML, Diamant M, ten Kulve J, Ijzerman RG. Cerebral effects of glucagon-like peptide-1 receptor blockade before and after Roux-en-Y gastric bypass surgery in obese women: A proof-of-concept resting-state functional MRI study. Diabetes Obes Metab 2021; 23:415-424. [PMID: 33084088 PMCID: PMC7821255 DOI: 10.1111/dom.14233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/27/2020] [Accepted: 10/18/2020] [Indexed: 12/31/2022]
Abstract
AIM To assess the effects of Roux-en-Y gastric bypass surgery (RYGB)-related changes in glucagon-like peptide-1 (GLP-1) on cerebral resting-state functioning in obese women. MATERIALS AND METHODS In nine obese females aged 40-54 years in the fasted state, we studied the effects of RYGB and GLP-1 on five a priori selected networks implicated in food- and reward-related processes as well as environment monitoring (default mode, right frontoparietal, basal ganglia, insula/anterior cingulate and anterior cingulate/orbitofrontal networks). RESULTS Before surgery, GLP-1 receptor blockade (using exendin9-39) was associated with increased right caudate nucleus (basal ganglia network) and decreased right middle frontal (right frontoparietal network) connectivity compared with placebo. RYGB resulted in decreased right orbitofrontal (insula/anterior cingulate network) connectivity. In the default mode network, after surgery, GLP-1 receptor blockade had a larger effect on connectivity in this region than GLP-1 receptor blockade before RYGB (all PFWE < .05). Results remained similar after correction for changes in body weight. Default mode and right frontoparietal network connectivity changes were related to changes in body mass index and food scores after RYGB. CONCLUSIONS These findings suggest GLP-1 involvement in resting-state networks related to food and reward processes and monitoring of the internal and external environment, pointing to a potential role for GLP-1-induced changes in resting-state connectivity in RYGB-mediated weight loss and appetite control.
Collapse
Affiliation(s)
- Eelco van Duinkerken
- Department of Medical PsychologyAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
- Amsterdam Diabetes Center/Department of Internal MedicineAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
- Department of NeurologyHospital Universitário Gaffrée e Guinle, Universidade Federal do Estado do Rio de JaneiroRio de JaneiroBrazil
- Center for EpilepsyInstituto Estadual do Cérebro Paulo NiemeyerRio de JaneiroBrazil
| | - Gabriel Bernardes
- Departament of PsychologyPontifícia Universidade Católica do Rio de JaneiroRio de JaneiroBrazil
| | - Liselotte van Bloemendaal
- Amsterdam Diabetes Center/Department of Internal MedicineAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
| | - Dick J. Veltman
- Department of PsychiatryAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear MedicineAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
- Institute of Neurology and Healthcare EngineeringUniversity College LondonLondonUK
| | - Daniel C. Mograbi
- Departament of PsychologyPontifícia Universidade Católica do Rio de JaneiroRio de JaneiroBrazil
- Institute of PsychiatryKing's College LondonLondonUK
| | | | - Carolyn F. Deacon
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum InstituteUniversity of CopenhagenCopenhagenDenmark
| | - Jens J. Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum InstituteUniversity of CopenhagenCopenhagenDenmark
| | - Madeleine L. Drent
- Amsterdam Diabetes Center/Department of Internal MedicineAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
- Section of Clinical Neuropsychology, Department of Clinical, Neuro‐ & Developmental PsychologyFaculty of Behavioral and Movement Sciences, Vrije UniversiteitAmsterdamthe Netherlands
| | - Michaela Diamant
- Amsterdam Diabetes Center/Department of Internal MedicineAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
| | - Jennifer ten Kulve
- Amsterdam Diabetes Center/Department of Internal MedicineAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
| | - Richard G. Ijzerman
- Amsterdam Diabetes Center/Department of Internal MedicineAmsterdam University Medical Centers, Vrije UniversiteitAmsterdamthe Netherlands
| |
Collapse
|