1
|
Taneera J, Saber-Ayad MM. Preservation of β-Cells as a Therapeutic Strategy for Diabetes. Horm Metab Res 2024; 56:261-271. [PMID: 38387480 DOI: 10.1055/a-2239-2668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The preservation of pancreatic islet β-cells is crucial in diabetes mellitus, encompassing both type 1 and type 2 diabetes. β-cell dysfunction, reduced mass, and apoptosis are central to insufficient insulin secretion in both types. Research is focused on understanding β-cell characteristics and the factors regulating their function to develop novel therapeutic approaches. In type 1 diabetes (T1D), β-cell destruction by the immune system calls for exploring immunosuppressive therapies, non-steroidal anti-inflammatory drugs, and leukotriene antagonists. Islet transplantation, stem cell therapy, and xenogeneic transplantation offer promising strategies for type 1 diabetes treatment. For type 2 diabetes (T2D), lifestyle changes like weight loss and exercise enhance insulin sensitivity and maintain β-cell function. Additionally, various pharmacological approaches, such as cytokine inhibitors and protein kinase inhibitors, are being investigated to protect β-cells from inflammation and glucotoxicity. Bariatric surgery emerges as an effective treatment for obesity and T2D by promoting β-cell survival and function. It improves insulin sensitivity, modulates gut hormones, and expands β-cell mass, leading to diabetes remission and better glycemic control. In conclusion, preserving β-cells offers a promising approach to managing both types of diabetes. By combining lifestyle modifications, targeted pharmacological interventions, and advanced therapies like stem cell transplantation and bariatric surgery, we have a significant chance to preserve β-cell function and enhance glucose regulation in diabetic patients.
Collapse
Affiliation(s)
- Jalal Taneera
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha M Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Welch AA, Farahani RA, Egan AM, Laurenti MC, Zeini M, Vella M, Bailey KR, Cobelli C, Dalla Man C, Matveyenko A, Vella A. Glucagon-like peptide-1 receptor blockade impairs islet secretion and glucose metabolism in humans. J Clin Invest 2023; 133:e173495. [PMID: 37751301 PMCID: PMC10645389 DOI: 10.1172/jci173495] [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: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUNDProglucagon can be processed to glucagon-like peptide1 (GLP-1) within the islet, but its contribution to islet function in humans remains unknown. We sought to understand whether pancreatic GLP-1 alters islet function in humans and whether this is affected by type 2 diabetes.METHODSWe therefore studied individuals with and without type 2 diabetes on two occasions in random order. On one occasion, exendin 9-39, a competitive antagonist of the GLP-1 Receptor (GLP1R), was infused, while on the other, saline was infused. The tracer dilution technique ([3-3H] glucose) was used to measure glucose turnover during fasting and during a hyperglycemic clamp.RESULTSExendin 9-39 increased fasting glucose concentrations; fasting islet hormone concentrations were unchanged, but inappropriate for the higher fasting glucose observed. In people with type 2 diabetes, fasting glucagon concentrations were markedly elevated and persisted despite hyperglycemia. This impaired suppression of endogenous glucose production by hyperglycemia.CONCLUSIONThese data show that GLP1R blockade impairs islet function, implying that intra-islet GLP1R activation alters islet responses to glucose and does so to a greater degree in people with type 2 diabetes.TRIAL REGISTRATIONThis study was registered at ClinicalTrials.gov NCT04466618.FUNDINGThe study was primarily funded by NIH NIDDK DK126206. AV is supported by DK78646, DK116231 and DK126206. CDM was supported by MIUR (Italian Minister for Education) under the initiative "Departments of Excellence" (Law 232/2016).
Collapse
Affiliation(s)
- Andrew A. Welch
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Rahele A. Farahani
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Aoife M. Egan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Marcello C. Laurenti
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Maya Zeini
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Max Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Kent R. Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Salehi M, Peterson R, Tripathy D, Pezzica S, DeFronzo R, Gastaldelli A. Differential effect of gastric bypass versus sleeve gastrectomy on insulinotropic action of endogenous incretins. Obesity (Silver Spring) 2023; 31:2774-2785. [PMID: 37853989 PMCID: PMC10593483 DOI: 10.1002/oby.23872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVE Prandial hyperinsulinemia after Roux-en-Y gastric bypass surgery (GB), and to lesser degree after sleeve gastrectomy (SG), has been attributed to rapid glucose flux from the gut and increased insulinotropic gut hormones. However, β-cell sensitivity to exogenous incretin is reduced after GB. This study examines the effect of GB versus SG on prandial glycemia and β-cell response to increasing concentrations of endogenous incretins. METHODS Glucose kinetics, insulin secretion rate (ISR), and incretin responses to 50-g oral glucose ingestion were compared between ten nondiabetic participants with GB versus nine matched individuals with SG and seven nonoperated normal glucose tolerant control individuals (CN) with and without administration of 200 mg of sitagliptin. RESULTS Fasting glucose and hormonal levels were similar among three groups. Increasing plasma concentrations of endogenous incretins by two- to three-fold diminished prandial glycemia and increased β-cell secretion in all three groups (p < 0.05), but insulin secretion per insulin sensitivity (i.e., disposition index) was increased only in GB (p < 0.05 for interaction). However, plot of the slope of ISR (from premeal to peak values) versus plasma glucagon-like peptide-1 concentration was smaller after GB compared with SG and CN. CONCLUSIONS After GB, increasing incretin activity augments prandial β-cell response whereas the β-cell sensitivity to increasing plasma concentrations of endogenous incretin is diminished.
Collapse
Affiliation(s)
- Marzieh Salehi
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
- STVHCS, Audie Murphy Hospital, San Antonio, TX, United States
| | - Richard Peterson
- Department of Surgery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Devjit Tripathy
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
| | - Samantha Pezzica
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Ralph DeFronzo
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
| | - Amalia Gastaldelli
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| |
Collapse
|
6
|
Farahani RA, Egan AM, Welch AA, Laurenti MC, Cobelli C, Dalla Man C, Vella A. The Effect of Glucagon-Like Peptide 1 Receptor Blockade on Glucagon-Induced Stimulation of Insulin Secretion. Diabetes 2023; 72:449-454. [PMID: 36562995 PMCID: PMC10260388 DOI: 10.2337/db22-0709] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
Data from transgenic rodent models suggest that glucagon acts as an insulin secretagogue by signaling through the glucagon-like peptide 1 receptor (GLP-1R) present on β-cells. However, its net contribution to physiologic insulin secretion in humans is unknown. To address this question, we studied individuals without diabetes in two separate experiments. Each subject was studied on two occasions in random order. In the first experiment, during a hyperglycemic clamp, glucagon was infused at 0.4 ng/kg/min, increasing by 0.2 ng/kg/min every hour for 5 h. On one day, exendin-9,39 (300 pmol/kg/min) was infused to block GLP-1R, while on the other, saline was infused. The insulin secretion rate (ISR) was calculated by nonparametric deconvolution from plasma concentrations of C-peptide. Endogenous glucose production and glucose disappearance were measured using the tracer-dilution technique. Glucagon concentrations, by design, did not differ between study days. Integrated ISR was lower during exendin-9,39 infusion (213 ± 26 vs. 191 ± 22 nmol/5 h, saline vs. exendin-9,39, respectively; P = 0.02). In the separate experiment, exendin-9,39 infusion, compared with saline infusion, also decreased the β-cell secretory response to a 1-mg glucagon bolus. These data show that, in humans without diabetes, glucagon partially stimulates the β-cell through GLP-1R.
Collapse
Affiliation(s)
- Rahele A. Farahani
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN
| | - Aoife M. Egan
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN
| | - Andrew A. Welch
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN
| | - Marcello C. Laurenti
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
| | - Claudio Cobelli
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
7
|
Salehi M, Peterson R, Tripathy D, Pezzica S, DeFronzo R, Gastaldelli A. Insulinotropic effect of endogenous incretins is greater after gastric bypass than sleeve gastrectomy despite diminished beta-cell sensitivity to plasma incretins. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.28.23287755. [PMID: 37034666 PMCID: PMC10081422 DOI: 10.1101/2023.03.28.23287755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
BACKGROUND/AIMS Prandial hyperinsulinemia after Roux-en Y gastric bypass surgery (GB), and to lesser degree after sleeve gastrectomy (SG), has been attributed to rapid glucose flux from the gut and increased insulinotropic gut hormones. However, β-cell sensitivity to exogenous incretin is markedly reduced after GB. This study examines the effect of GB versus SG on prandial glycemia and β-cell response to increasing concentrations of endogenous incretins. METHODS Glucose kinetics, insulin secretion rate (ISR), and incretin responses to 50-gram oral glucose ingestion were compared between 10 non-diabetic subjects with GB versus 9 matched individuals with SG and 7 non-operated normal glucose tolerant controls (CN) on two days with and without administration of 200 mg sitagliptin. RESULTS Fasting glucose and hormonal levels were similar among 3 groups. Increasing plasma concentrations of endogenous incretins by 2-3-fold diminished post-OGTT glycemia and increased β-cell secretion in all 3 groups (p<0.05), but insulin secretion per insulin sensitivity (i.e., disposition index) was increased only in GB (p<0.05 for interaction). As a result, sitagliptin administration led to hypoglycemia in 3 of 10 GB. Yet, plot of the slope of ISR versus the increase in endogenous incretin concentration was smaller after GB compared to both SG and CN. CONCLUSION Augmented glycemic-induced β-cell response caused by enhanced incretin activity is unique to GB and not shared with SG. However, the β-cell sensitivity to increasing concentrations of endogenous incretin is smaller after bariatric surgery, particularly after GB, compared to non-operated controls, indicating a long-term adaptation of gut-pancreas axis after these procedures. HIGHLIGHTS What is known?: Glycemic effects of gastric bypass (GB) and sleeve gastrectomy (SG) is attributed to rapid nutrient flux and enhanced insulinotropic effects of gut hormones but β-cell sensitivity to exogenous GLP-1 or GIP is diminished after GB. What the present findings add?: Post-OGTT β-cell sensitivity to enhanced endogenous incretins by DPP4i is markedly reduced in bariatric subjects versus non-operated controls, and yet insulin secretory response (disposition index) is increased leading to hypoglycemia in GB and not SG. Significance?: Blunted sensitivity to GLP-1 may represent β-cell adaptation to massive elevation in GLP-1 secretion following bariatric surgery to protect against hypoglycemia.The differential effect of enhanced concentrations of incretins on post-OGTT insulin response (disposition index) among GB versus SG highlights a distinct adaptive process among the two procedures.Augmented insulinotropic effects of gut hormones on postprandial insulin secretory response after GB despite a reduced beta-cell sensitivity to plasma concentrations of GLP-1 makes a case for non-hormonal mechanisms of GLP-1 action after GB.Better understanding of long-term effects of bariatric surgery on gut-pancreas axis activity is critical in development of GLP-1-based strategies to address glucose abnormalities (both hyperglycemia and hypoglycemia) in these settings.
Collapse
|
8
|
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: 9] [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
|
9
|
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: 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: 04/12/2022] [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
|
10
|
Fries CM, Haange SB, Rolle-Kampczyk U, Till A, Lammert M, Grasser L, Medawar E, Dietrich A, Horstmann A, von Bergen M, Fenske WK. Metabolic Profile and Metabolite Analyses in Extreme Weight Responders to Gastric Bypass Surgery. Metabolites 2022; 12:metabo12050417. [PMID: 35629921 PMCID: PMC9147451 DOI: 10.3390/metabo12050417] [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: 03/31/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Roux-en-Y gastric bypass (RYGB) surgery belongs to the most frequently performed surgical therapeutic strategies against adiposity and its comorbidities. However, outcome is limited in a substantial cohort of patients with inadequate primary weight loss or considerable weight regain. In this study, gut microbiota composition and systemically released metabolites were analyzed in a cohort of extreme weight responders after RYGB. Methods: Patients (n = 23) were categorized based on excess weight loss (EWL) at a minimum of two years after RYGB in a good responder (EWL 93 ± 4.3%) or a bad responder group (EWL 19.5 ± 13.3%) for evaluation of differences in metabolic outcome, eating behavior and gut microbiota taxonomy and metabolic activity. Results: Mean BMI was 47.2 ± 6.4 kg/m2 in the bad vs. 26.6 ± 1.2 kg/m2 in the good responder group (p = 0.0001). We found no difference in hunger and satiety sensation, in fasting or postprandial gut hormone release, or in gut microbiota composition between both groups. Differences in weight loss did not reflect in metabolic outcome after RYGB. While fecal and circulating metabolite analyses showed higher levels of propionate (p = 0.0001) in good and valerate (p = 0.04) in bad responders, respectively, conjugated primary and secondary bile acids were higher in good responders in the fasted (p = 0.03) and postprandial state (GCA, p = 0.02; GCDCA, p = 0.02; TCA, p = 0.01; TCDCA, p = 0.02; GDCA, p = 0.05; GUDCA, p = 0.04; TLCA, p = 0.04). Conclusions: Heterogenous weight loss response to RYGB surgery separates from patients’ metabolic outcome, and is linked to unique serum metabolite signatures post intervention. These findings suggest that the level of adiposity reduction alone is insufficient to assess the metabolic success of RYGB surgery, and that longitudinal metabolite profiling may eventually help us to identify markers that could predict individual adiposity response to surgery and guide patient selection and counseling.
Collapse
Affiliation(s)
- Charlotte M. Fries
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; (A.T.); (W.K.F.)
- Correspondence:
| | - Sven-Bastiaan Haange
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318 Leipzig, Germany; (S.-B.H.); (U.R.-K.); (M.v.B.)
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318 Leipzig, Germany; (S.-B.H.); (U.R.-K.); (M.v.B.)
| | - Andreas Till
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; (A.T.); (W.K.F.)
| | - Mathis Lammert
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1a, 04103 Leipzig, Germany; (M.L.); (L.G.); (E.M.); (A.H.)
| | - Linda Grasser
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1a, 04103 Leipzig, Germany; (M.L.); (L.G.); (E.M.); (A.H.)
| | - Evelyn Medawar
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1a, 04103 Leipzig, Germany; (M.L.); (L.G.); (E.M.); (A.H.)
| | - Arne Dietrich
- Department of Visceral and Metabolic Surgery, University Hospital Leipzig, Liebigstraße 18, 04103 Leipzig, Germany;
| | - Annette Horstmann
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstraße 1a, 04103 Leipzig, Germany; (M.L.); (L.G.); (E.M.); (A.H.)
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, 00290 Helsinki, Finland
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research GmbH-UFZ, Permoserstraße 15, 04318 Leipzig, Germany; (S.-B.H.); (U.R.-K.); (M.v.B.)
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Puschstraße 4, 04103 Leipzig, Germany
- Faculty of Life Sciences, Institute of Biochemistry, University of Leipzig, Brüderstraße 34, 04103 Leipzig, Germany
| | - Wiebke K. Fenske
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; (A.T.); (W.K.F.)
| |
Collapse
|
11
|
Cifuentes L, Hurtado MD, Cortes TM, Gonzales K, Acosta A, Shah M, Collazo-Clavell ML. Evaluation and Management of Patients Referred for Post-Bariatric Surgery Hypoglycemia at a Tertiary Care Center. Obes Surg 2022; 32:1578-1585. [PMID: 35260971 PMCID: PMC10866015 DOI: 10.1007/s11695-022-05986-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Following bariatric surgery, patients can develop non-specific symptoms self-described as hypoglycemia. However, confirming hypoglycemia can be technically challenging, and therefore, these individuals are frequently treated empirically. This study aimed to describe what diagnostic evaluation and therapeutic interventions patients referred for post-bariatric surgery hypoglycemia undergo. METHODS Retrospective observational cohort study of patients with a history of bariatric surgery was evaluated for post-bariatric surgery hypoglycemia in a tertiary referral center from 2008 to 2017. We collected demographic and bariatric surgery information, clinical presentation of symptoms referred to as hypoglycemia, laboratory and imaging studies performed to evaluate these symptoms, and symptom management and outcomes. RESULTS A total of 60/2450 (2.4%) patients who underwent bariatric surgery were evaluated in the Department of Endocrinology for hypoglycemia-related symptoms. The majority were middle-aged women without type 2 diabetes who had undergone Roux-en-Y gastric bypass. Thirty-nine patients (65%) completed a biochemical assessment for hypoglycemia episodes. Six (10%) had confirmed hypoglycemia by Whipple's triad, and four (6.7%) met the criteria for post-bariatric surgery hypoglycemia based on clinical and biochemical criteria. All patients were recommended dietary modification as the initial line of treatment, and this intervention resulted in most patients reporting at least some improvement in their symptoms. Eight patients (13%) were prescribed pharmacotherapy, and two patients required additional interventions for symptom control. CONCLUSIONS In our experience, evaluation for hypoglycemia-related symptoms after bariatric surgery was rare. Hypoglycemia was confirmed in the minority of patients. Even without establishing a diagnosis of hypoglycemia, dietary changes were a helpful strategy for symptom management for most patients.
Collapse
Affiliation(s)
- Lizeth Cifuentes
- Precision Medicine for Obesity Program, Mayo Clinic, 200 1st SW, Rochester, MN, 55905, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, 200 1st SW, Rochester, MN, 55905, USA
| | - Maria D Hurtado
- Precision Medicine for Obesity Program, Mayo Clinic, 200 1st SW, Rochester, MN, 55905, USA
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic Health System, 700 West Av. South, La Crosse, WI, 64601, USA
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Tiffany M Cortes
- Division of Endocrinology, University of Texas Health, San Antonio, TX, 78229, USA
| | - Kristen Gonzales
- Division of Endocrinology, Diabetes, and Metabolism, University of New Mexico Health Sciences Center, Albuquerque, NM, 87106, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Mayo Clinic, 200 1st SW, Rochester, MN, 55905, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, 200 1st SW, Rochester, MN, 55905, USA
| | - Meera Shah
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Maria L Collazo-Clavell
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
12
|
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: 2.0] [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
|
13
|
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: 6] [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
|
14
|
Pérez-Arana GM, Gómez AD, de Los Reyes JB, Camacho-Ramírez A, Fernández-Vivero J, Ribelles-García A, Almorza-Gomar D, Carrasco-Molinillo C, Mateo-Gavira I, Prada-Oliveira JA. The long-term failure of RYGB surgery in improving T2DM is related to hyperinsulinism. Ann Anat 2021; 240:151855. [PMID: 34785322 DOI: 10.1016/j.aanat.2021.151855] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) is the gold standard method for bariatric surgery and leads to substantial improvements in Type 2 Diabetes mellitus. However, many patients experience relapses in diabetes five years after undergoing this aggressive surgical procedure. We focus on beta-cell population changes and absorptive intestinal consequences after RYGB in a healthy nonobese animal model after a long survival period. METHODS For our purpose, we use three groups of Wistar rats: RYGB-operated, surgical control (Sham) and fasting control. We measure alpha-, beta-cell mass; transcription (Arx, and Pdx-1) and proliferation (Ki67) factors; glucose tolerance and insulin release after oral glucose tests; histological adaptive changes in the jejunum; and intestinal glucose transporters. RESULTS Our results showed an early increase in insulin secretion after surgery, that decrease at the end of the study. The beta-cell mass reduces twenty-four weeks after RYGB, which coincides with decrease of Pdx-1 transcription promoter factor. These was coincident with an increase in alpha-mass and a high expression of Arx in RYGB group. CONCLUSIONS The analysis of all data showed beta-cell mass transdifferentiation into alpha-cell mass in RYGB rats. Due to long-term exhaustion of the beta-cell population by hyperinsulinism derived from digestive tract adaptation to surgery.
Collapse
Affiliation(s)
- Gonzalo-Martín Pérez-Arana
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cadiz, Spain; Asociación Gaditana de Apoyo al Investigador (AGAI) Cádiz, Spain; Institute for Bi omedical Science Research and Innovation (INIBICA) University of Cadiz, Spain.
| | | | | | | | - José Fernández-Vivero
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cadiz, Spain
| | | | - David Almorza-Gomar
- Complejo Hospitalario de Badajoz, Servicio Extremeño de Salud, Spain; Operative Statistic and Research Department, University of Cádiz, Spain
| | | | - Isabel Mateo-Gavira
- Institute for Bi omedical Science Research and Innovation (INIBICA) University of Cadiz, Spain; Endocrine and Metabolic Unit, Puerta del Mar Universitary Hospital, University of Cadiz, Spain
| | - José-Arturo Prada-Oliveira
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cadiz, Spain; Asociación Gaditana de Apoyo al Investigador (AGAI) Cádiz, Spain; Institute for Bi omedical Science Research and Innovation (INIBICA) University of Cadiz, Spain.
| |
Collapse
|
15
|
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: 4.3] [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
|
16
|
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
|
17
|
Jin ZL, Liu W. Progress in treatment of type 2 diabetes by bariatric surgery. World J Diabetes 2021; 12:1187-1199. [PMID: 34512886 PMCID: PMC8394224 DOI: 10.4239/wjd.v12.i8.1187] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/29/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
The incidence of type 2 diabetes (T2D) is increasing at an alarming rate worldwide. Bariatric surgical procedures, such as the vertical sleeve gastrectomy and Roux-en-Y gastric bypass, are the most efficient approaches to obtain substantial and durable remission of T2D. The benefits of bariatric surgery are realized through the consequent increased satiety and alterations in gastrointestinal hormones, bile acids, and the intestinal microbiota. A comprehensive understanding of the mechanisms by which various bariatric surgical procedures exert their benefits on T2D could contribute to the design of better non-surgical treatments for T2D. In this review, we describe the classification and evolution of bariatric surgery and explore the multiple mechanisms underlying the effect of bariatric surgery on insulin resistance. Based upon our summarization of the current knowledge on the underlying mechanisms, we speculate that the gut might act as a new target for improving T2D. Our ultimate goal with this review is to provide a better understanding of T2D pathophysiology in order to support development of T2D treatments that are less invasive and more scalable.
Collapse
Affiliation(s)
- Zhang-Liu Jin
- Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Wei Liu
- Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
18
|
Camacho-Ramírez A, Prada-Oliveira JA, Ribelles-García A, Almorza-Gomar D, Pérez-Arana GM. The Leading Role of Peptide Tyrosine Tyrosine in Glycemic Control After Roux-en-Y Gastric Bypass in Rats. Obes Surg 2021; 30:697-706. [PMID: 31701411 DOI: 10.1007/s11695-019-04239-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS Roux-en-Y gastric bypass (RYGB) is one of the most effective surgical therapies for the rapid resolution of type 2 diabetes. However, the mechanisms underlying the entero-hormonal response after surgery and the role of peptide tyrosine tyrosine (PYY) in the restoration of normoglycemia are still not clear. METHODS We reproduced the RYGB technique in Wistar and Goto-Kakizaki rats and performed serum hormonal, histological, and hormonal-infusion test. RESULTS Using the diabetic Goto-Kakizaki (GK) rat model, we demonstrated that PYY plasma levels showed a remarkable peak approximately 30 min earlier than GLP-1 or GIP after mixed-meal administration in RYGB-operated rats with PYY. The GLP-1 and GIP areas under the curve (AUCs) increased after RYGB in GK rats. Additionally, the findings suggested that PYY (3-36) infusion led to increased GLP-1 and GIP plasma levels close to those obtained after a meal. Finally, the number of GLP-1-positive cells appeared to increase in the three segments of the small intestine in GK-RYGB-operated rats beyond the early presence of nutrient stimulation in the ileum. Nevertheless, PYY-positive cell numbers appeared to increase only in the ileum. CONCLUSION At least in rats, these data demonstrate an earlier essential role for PYY in gut hormone regulation after RYGB. We understand that PYY contributes to GLP-1 and GIP release and there must be the existence of enteroendocrine communication routes between the distal and proximal small intestine.
Collapse
Affiliation(s)
- Alonso Camacho-Ramírez
- Puerta del Mar Hospital, University of Cadiz, Cadiz, Spain.,Asociación Gaditana de Apoyo al Investigador, Cadiz, Spain.,Biomedical Science Research and Innovation Institute (INIBICA), University of Cadiz, Cadiz, Spain
| | - J Arturo Prada-Oliveira
- Asociación Gaditana de Apoyo al Investigador, Cadiz, Spain.,Biomedical Science Research and Innovation Institute (INIBICA), University of Cadiz, Cadiz, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cadiz, Plaza Fragela s/n, 11003, Cadiz, Spain
| | - Antonio Ribelles-García
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cadiz, Plaza Fragela s/n, 11003, Cadiz, Spain.,Sustainable Social Development Research Institute (INDESS), University of Cadiz, Cadiz, Spain
| | - David Almorza-Gomar
- Biomedical Science Research and Innovation Institute (INIBICA), University of Cadiz, Cadiz, Spain.,Operative Statistic and Research Department, University of Cadiz, Cadiz, Spain
| | - Gonzalo M Pérez-Arana
- Asociación Gaditana de Apoyo al Investigador, Cadiz, Spain. .,Biomedical Science Research and Innovation Institute (INIBICA), University of Cadiz, Cadiz, Spain. .,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cadiz, Plaza Fragela s/n, 11003, Cadiz, Spain.
| |
Collapse
|
19
|
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: 30] [Impact Index Per Article: 10.0] [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
|
20
|
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: 16] [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
|
21
|
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
|
22
|
Metabolic Effect of the Hepatic Branch of the Vagal Nerve in One-Anastomosis Gastric Bypass (OAGB). World J Surg 2021; 44:1939-1944. [PMID: 32020329 DOI: 10.1007/s00268-020-05405-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Bariatric surgery is often performed not only to lose weight, but also to improve obesity-related comorbidities. A certain metabolic effect of the bariatric techniques has been demonstrated, as the improvement or even remission of comorbidities is patent, before having lost a relevant amount of weight. The autonomic innervation of diverse viscera by the vagus nerve has been hypothesized to participate in it. We aimed to evaluate the ponderal and metabolic impact of the preservation of the hepatic branch of the vagus in type 2 diabetic (T2D) patients undergoing one-anastomosis gastric bypass (OAGB). PATIENTS AND METHODS We conducted a prospective study on patients with a preoperative diagnosis of morbid obesity and T2D, who underwent an OAGB. Preservation of the hepatic branch of the vagus was carried out in the first 14 patients (Group 1), whereas in another 14 patients it was sectioned. Ponderal and metabolic outcomes were assessed 1 and 2 years after surgery. RESULTS The length of the biliary limb was 210 cm in both groups. Postoperative BMI or excess BMI loss was not significantly different between groups. The patients included in Group 1 showed a statistically greater improvement in glycemic and lipid variables. CONCLUSION The preservation of the hepatic branch of the vagus in an OAGB leads to more favorable postoperative glycemic and lipid profiles.
Collapse
|
23
|
Incretin Hormones in Obesity and Related Cardiometabolic Disorders: The Clinical Perspective. Nutrients 2021; 13:nu13020351. [PMID: 33503878 PMCID: PMC7910956 DOI: 10.3390/nu13020351] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of obesity continues to grow rapidly worldwide, posing many public health challenges of the 21st century. Obese subjects are at major risk for serious diet-related noncommunicable diseases, including type 2 diabetes mellitus, cardiovascular disease, and non-alcoholic fatty liver disease. Understanding the mechanisms underlying obesity pathogenesis is needed for the development of effective treatment strategies. Dysregulation of incretin secretion and actions has been observed in obesity and related metabolic disorders; therefore, incretin-based therapies have been developed to provide new therapeutic options. Incretin mimetics present glucose-lowering properties, together with a reduction of appetite and food intake, resulting in weight loss. In this review, we describe the physiology of two known incretins—glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), and their role in obesity and related cardiometabolic disorders. We also focus on the available and incoming incretin-based medications that can be used in the treatment of the above-mentioned conditions.
Collapse
|
24
|
Kittah E, Camilleri M, Jensen MD, Vella A. A Pilot Study Examining the Effects of GLP-1 Receptor Blockade Using Exendin-(9,39) on Gastric Emptying and Caloric Intake in Subjects With and Without Bariatric Surgery. Metab Syndr Relat Disord 2020; 18:406-412. [PMID: 32833560 DOI: 10.1089/met.2020.0049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: Obesity causes significant morbidity and mortality and continues to be a significant public health concern. Unfortunately, lifestyle modification and pharmacotherapy do not produce durable results. This has led to bariatric surgical procedures playing an increasingly prominent role in the management of medically complicated obesity. Roux-en-Y gastric bypass and sleeve gastrectomy are the most commonly performed bariatric surgeries in North America and produce mechanical restriction with accelerated gastrointestinal transit accompanied by increased postprandial secretion of glucagon-like peptide-1 (GLP-1). GLP-1 is a gastrointestinal hormone that delays gastric emptying and causes satiety and weight loss. This raises the possibility that the postprandial rise in GLP-1 might affect feeding behavior over and above the mechanical restriction produced by bariatric surgery. Methods: We, therefore, sought to determine the effects of GLP-1 receptor blockade using exendin-9,39-a competitive antagonist of the actions of GLP-1 at its receptor-on caloric intake and gastrointestinal transit in subjects after sleeve gastrectomy and after Roux-en-Y gastric bypass compared with weight-matched controls. Results: GLP-1 receptor blockade did not alter caloric intake in people after bariatric surgery. However, caloric intake was decreased in age-, weight- and sex-matched control subjects, and the mechanisms require further study. Conclusions: Given the known effects of GLP-1 on gastric accommodation, future studies should ascertain effects of GLP-1 receptor blockade on gastric accommodation, which might be a useful and novel strategy to decrease caloric intake in humans with an intact upper gastrointestinal tract. The Clinical Trial Resigtration number is NCT02779075.
Collapse
Affiliation(s)
- Esi Kittah
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael D Jensen
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Adrian Vella
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
25
|
Zhou LY, Deng MQ, Xiao XH. Potential contribution of the gut microbiota to hypoglycemia after gastric bypass surgery. Chin Med J (Engl) 2020; 133:1834-1843. [PMID: 32649508 PMCID: PMC7470015 DOI: 10.1097/cm9.0000000000000932] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity has become a global health problem. Lifestyle modification and medical treatment only appear to yield short-term weight loss. Roux-en-Y gastric bypass (RYGB) is the most popular bariatric procedure, and it sustains weight reduction and results in the remission of obesity-associated comorbidities for obese individuals. However, patients who undergo this surgery may develop hypoglycemia. To date, the diagnosis is challenging and the prevalence of post-RYGB hypoglycemia (PRH) is unclear. RYGB alters the anatomy of the upper gastrointestinal tract and has a combined effect of caloric intake restriction and nutrient malabsorption. Nevertheless, the physiologic changes after RYGB are complex. Although hyperinsulinemia, incretin effects, dysfunction of β-cells and α-cells, and some other factors have been widely investigated and are reported to be possible mediators of PRH, the pathogenesis is still not completely understood. In light of the important role of the gut microbiome in metabolism, we hypothesized that the gut microbiome might also be a critical link between RYGB and hypoglycemia. In this review, we mainly highlight the current possible factors predisposing individuals to PRH, particularly related to the gut microbiota, which may yield significant insights into the intestinal regulation of glucose metabolic homeostasis and provide novel clues to improve the treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Li-Yuan Zhou
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | | | | |
Collapse
|
26
|
Abstract
As the prevalence of obesity has increased, bariatric surgery has become more common because of its proven efficacy at promoting weight loss and improving weight-related medical comorbidities. Although generally successful, bariatric surgery may also lead to complications and negatively affect health. This article highlights some common and rare complications of bariatric surgery. Specifically, it discusses the risk of nutrient deficiencies (which is influenced by surgery type) and their downstream effects, including ill-effects on bone health. Rarer complications, such as postbypass hypoglycemia and alcohol use disorder, are also discussed.
Collapse
Affiliation(s)
- Maria L Collazo-Clavell
- Division of Endocrinology, Diabetes and Nutrition, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA
| | - Meera Shah
- Division of Endocrinology, Diabetes and Nutrition, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
27
|
Egan AM, Vella A. Endoscopic Treatments for Obesity: The Good, the Bad, and the Ugly. Endocrinol Metab Clin North Am 2020; 49:315-328. [PMID: 32418593 DOI: 10.1016/j.ecl.2020.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the current setting of an obesity pandemic, there is an urgent need for minimally invasive, safe, and effective interventions for weight loss. Endoscopic bariatric procedures have been developed as an alternative to more traditional medical and surgical therapies. Multiple options are undergoing evaluation or are already available for clinical use. This review aims to describe these treatments, including their mechanisms of action, efficacy, safety and the knowledge gaps regarding their use.
Collapse
Affiliation(s)
- Aoife M Egan
- Division of Endocrinology and Diabetes, Department of Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Adrian Vella
- Division of Endocrinology and Diabetes, Department of Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
28
|
Zheng J, Liu J, Hong BS, Ke W, Huang M, Li Y. Circulating betatrophin/ANGPTL8 levels correlate with body fat distribution in individuals with normal glucose tolerance but not those with glucose disorders. BMC Endocr Disord 2020; 20:51. [PMID: 32299395 PMCID: PMC7161171 DOI: 10.1186/s12902-020-0531-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/01/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The relationship between betatrophin/ANGPTL8 and obesity has been investigated using body mass index (BMI); however, since BMI reflects overall adiposity rather than body fat distribution, it remains unclear whether fat deposition in different areas of the body affects betatrophin expression. Here, we investigated the correlation between circulating betatrophin levels and body fat distribution in patients with different glucose tolerance. METHODS We performed a cross-sectional study in 128 participants with impaired glucose tolerance (IGT; n = 64) or normal glucose tolerance (NGT; n = 64). Circulating betatrophin levels were detected by enzyme-linked immunosorbent assay (ELISA). Body fat distribution (subcutaneous, visceral, and limb fat) was measured by magnetic resonance imaging (MRI) and a body fat meter. RESULTS After controlling for age, sex, and BMI, betatrophin was correlated positively with visceral adipose tissue-to-subcutaneous adipose tissue ratio (VAT/SAT ratio; r = 0.339, p = 0.009) and negatively with body fat ratio (BFR; r = - 0.275, p = 0.035), left lower limb fat ratio (LLR; r = - 0.330, p = 0.011), and right lower limb fat ratio (RLR; r = - 0.288, p = 0.027) in the NGT group, with these correlations remaining after controlling for triglycerides. VAT/SAT ratio (standardized β = 0.419, p = 0.001) was independently associated with serum betatrophin levels; however, betatrophin was not associated with body fat distribution variables in the IGT group. CONCLUSIONS Circulating betatrophin levels correlated positively with VAT/SAT ratio and negatively with lower limb fat, but not with subcutaneous or upper limb fat, in individuals with normal glucose tolerance. Thus, betatrophin may be a potential biomarker for body fat distribution in individuals without glucose disorders.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Endocrinology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Juan Liu
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Beverly S Hong
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weijian Ke
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Minmin Huang
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanbing Li
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
29
|
Korakas E, Kountouri A, Raptis A, Kokkinos A, Lambadiari V. Bariatric Surgery and Type 1 Diabetes: Unanswered Questions. Front Endocrinol (Lausanne) 2020; 11:525909. [PMID: 33071965 PMCID: PMC7531037 DOI: 10.3389/fendo.2020.525909] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 08/20/2020] [Indexed: 01/19/2023] Open
Abstract
In recent decades there has been an alarming increase in the prevalence of obesity in patients with type 1 diabetes leading to the development of insulin resistance and cardiometabolic complications, with mechanisms poorly clarified. While bariatric surgery has long been considered an effective treatment option for patients with type 2 diabetes, the evidence regarding its benefits on weight loss and the prevention of complications in T1DM patients is scarce, with controversial outcomes. Bariatric surgery has been associated with a significant reduction in daily insulin requirement, along with a considerable reduction in body mass index, results which were sustained in the long term. Furthermore, studies suggest that bariatric surgery in type 1 diabetes results in the improvement of comorbidities related to obesity including hypertension and dyslipidemia. However, regarding glycemic control, the reduction of mean glycosylated hemoglobin was modest or statistically insignificant in most studies. The reasons for these results are yet to be elucidated; possible explanations include preservation of beta cell mass and increased residual function post-surgery, improvement in insulin action, altered GLP-1 function, timing of surgery, and association with residual islet cell mass. A number of concerns regarding safety issues have arisen due to the reporting of peri-operative and post-operative adverse events. The most significant complications are metabolic and include diabetic ketoacidosis, severe hypoglycemia and glucose fluctuations. Further prospective clinical studies are required to provide evidence for the effect of bariatric surgery on T1DM patients. The results may offer a better knowledge for the selection of people living with diabetes who will benefit more from a metabolic surgery.
Collapse
Affiliation(s)
- Emmanouil Korakas
- Second Department of Internal Medicine, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Kountouri
- Second Department of Internal Medicine, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Raptis
- Second Department of Internal Medicine, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Medicine, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
- *Correspondence: Vaia Lambadiari
| |
Collapse
|
30
|
El Khoury L, Chouillard E, Chahine E, Saikaly E, Debs T, Kassir R. Metabolic Surgery and Diabesity: a Systematic Review. Obes Surg 2019; 28:2069-2077. [PMID: 29679334 DOI: 10.1007/s11695-018-3252-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bariatric surgery is used to induce weight loss (baros = weight). Evidence has shown that bariatric surgery improves the comorbid conditions associated with obesity such as hypertension, hyperlipidemia, and type 2 diabetes mellitus T2DM. Hence, shifting towards using metabolic surgery instead of bariatric surgery is currently more appropriate in certain subset of patients. Endocrine changes resulting from operative manipulation of the gastrointestinal tract after metabolic surgery translate into metabolic benefits with respect to the comorbid conditions. Other changes include bacterial flora rearrangement, bile acids secretion, and adipose tissue effect. The aim of this systematic review is to examine clinical trials regarding long-term effects of bariatric and metabolic surgery on patients with T2DM and to evaluate the potential mechanisms leading to the improvement in the glycaemic control.
Collapse
Affiliation(s)
- Lionel El Khoury
- Department of Digestive and Minimally Invasive Surgery, Saint-Germain-en-Laye Medical Center, Poissy, France
| | - Elie Chouillard
- Department of Digestive and Minimally Invasive Surgery, Saint-Germain-en-Laye Medical Center, Poissy, France
| | - Elias Chahine
- Department of Digestive and Minimally Invasive Surgery, Saint-Germain-en-Laye Medical Center, Poissy, France
| | - Elias Saikaly
- Saint Georges Hospital University Medical Center, University of Balamand, Beirut, Lebanon
| | - Tarek Debs
- Department of General Surgery, CHU Archet, Nice, France
| | - Radwan Kassir
- Departement of Digestive Surgery, CHU Félix Guyon, Saint Denis, La Réunion, France.
- Department of Bariatric Surgery, CHU Félix Guyon, Saint Denis, La Réunion, France.
| |
Collapse
|
31
|
Douros JD, Tong J, D’Alessio DA. The Effects of Bariatric Surgery on Islet Function, Insulin Secretion, and Glucose Control. Endocr Rev 2019; 40:1394-1423. [PMID: 31241742 PMCID: PMC6749890 DOI: 10.1210/er.2018-00183] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 04/23/2019] [Indexed: 01/19/2023]
Abstract
Although bariatric surgery was developed primarily to treat morbid obesity, evidence from the earliest clinical observations to the most recent clinical trials consistently demonstrates that these procedures have substantial effects on glucose metabolism. A large base of research indicates that bariatric surgeries such as Roux-en-Y gastric bypass (RYGB), vertical sleeve gastrectomy (VSG), and biliopancreatic diversion (BPD) improve diabetes in most patients, with effects frequently evident prior to substantial weight reduction. There is now unequivocal evidence from randomized controlled trials that the efficacy of surgery is superior to intensive life-style/medical management. Despite advances in the clinical understanding and application of bariatric surgery, there remains only limited knowledge of the mechanisms by which these procedures confer such large changes to metabolic physiology. The improvement of insulin sensitivity that occurs with weight loss (e.g., the result of diet, illness, physical training) also accompanies bariatric surgery. However, there is evidence to support specific effects of surgery on insulin clearance, hepatic glucose production, and islet function. Understanding the mechanisms by which surgery affects these parameters of glucose regulation has the potential to identify new targets for therapeutic discovery. Studies to distinguish among bariatric surgeries on key parameters of glucose metabolism are limited but would be of considerable value to assist clinicians in selecting specific procedures and investigators in delineating the resulting physiology. This review is based on literature related to factors governing glucose metabolism and insulin secretion after the commonly used RYGB and VSG, and the less frequently used BPD and adjustable gastric banding.
Collapse
Affiliation(s)
- Jonathan D Douros
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Jenny Tong
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - David A D’Alessio
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| |
Collapse
|
32
|
Salehi M, Gastaldelli A, D'Alessio DA. Beta-cell sensitivity to insulinotropic gut hormones is reduced after gastric bypass surgery. Gut 2019; 68:1838-1845. [PMID: 30772836 DOI: 10.1136/gutjnl-2018-317760] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Postprandial hyperinsulinaemia after Roux-en Y gastric bypass (GB) has been attributed to rapid nutrient flux from the gut, and an enhanced incretin effect. However, it is unclear whether surgery changes islet cell responsiveness to regulatory factors. This study tested the hypothesis that β-cell sensitivity to glucagon like-peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) is attenuated after GB. DESIGN Ten non-diabetic subjects with GB, and 9 body mass index (BMI)-matched and age-matched non-surgical controls (CN) with normal glucose tolerance had blood glucose clamped at ~7.8 mM on three separate days. Stepwise incremental infusions of GLP-1 (15, 30, 60, 120 and 300 ng/LBkg/h), GIP (75, 150, 300, 600 and 1200 ng/LBkg/h) or saline were administered from 90 to 240 min and insulin secretion measured. RESULTS GB subjects had similar fasting glucose levels but lower fasting insulin compared with CN, likely due to increased insulin clearance. The average insulin secretion rates (ISRs) to 7.8 mM glucose were ~30% lower in GB relative to CN subjects. However, incretin-stimulated ISRs, adjusted for insulin sensitivity and glucose-stimulated insulin secretion, were even more attenuated in the GB subjects, by threefold to fourfold (AUCISR(90-240 min) during GLP-1 and GIP: 47±8 and 44±12 nmol in GB and 116±16 and 161±44 in CN; p<0.01). CONCLUSION After GB, the sensitivity of insulin secretion to both glucose and incretins is diminished.
Collapse
Affiliation(s)
- Marzieh Salehi
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Department of Medicine, University of Texas at San Antonio, San Antonio, Texas, USA.,Bartter Research Unit, South Texas Veterans Health Care System, Audie Murphy Hospital, San Antonio, Texas, USA
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| | - David A D'Alessio
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
33
|
Chakraborty S, Desai A, Halland M, Burton D, Camilleri M, Zinsmeister AR, Bharucha AE. Relationship between symptoms during a gastric emptying study and intestinal chemosensitivity with daily symptoms. Neurogastroenterol Motil 2019; 31:e13686. [PMID: 31328363 PMCID: PMC6746577 DOI: 10.1111/nmo.13686] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/25/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Non-ulcer dyspepsia (NUD) is a heterogeneous disorder, which is characterized by upper gastrointestinal symptoms and sensorimotor disturbances, including abnormal gastric emptying (GE) and increased intestinal chemosensitivity, and associated with greater plasma glucagon-like peptide-1 (GLP-1) levels during duodenal lipid infusion. However, the relationship(s) between these disturbances and daily symptoms in NUD is variable. We hypothesize that abnormal GE and symptoms during a GE study and during duodenal lipid infusion are associated with the severity of daily symptoms and that GLP-1 mediates symptoms during duodenal lipid infusion in NUD. METHODS Gastric emptying of solids, symptoms during the GE study and duodenal lipid infusion, and daily gastrointestinal symptoms (2 week diary) were measured in 24 healthy controls and 40 NUD patients. During duodenal lipid infusion, participants received the GLP-1 antagonist exendin 9-39 or placebo. KEY RESULTS In controls and patients, GE of solids was normal in 75% and 75%, delayed in 8% and 12.5%, or rapid in 17% and 12.5%, respectively. No controls but 26 patients (65%) had severe symptoms during the GE study. During lipid infusion, gastrointestinal symptoms were greater (P = .001) in patients and not affected by exendin. Symptoms during GE study and lipid infusion accounted for respectively 62% and 37% of variance in daily symptom severity. CONCLUSIONS In NUD, symptoms during a GE study and to a lesser extent during lipid infusion explain the variance in daily symptoms. Intestinal chemosensitivity is not reduced by GLP-1 antagonist. Assessment of symptoms during a GE study may provide a useful biomarker for NUD in research and clinical practice.
Collapse
Affiliation(s)
| | - Anshuman Desai
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester Minnesota
| | - Magnus Halland
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester Minnesota
| | - Duane Burton
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester Minnesota
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester Minnesota
| | - Alan R. Zinsmeister
- Division of Biomedical Statistics and Informatics Mayo Clinic Rochester Minnesota
| | - Adil E. Bharucha
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester Minnesota
| |
Collapse
|
34
|
Remission of Type 2 Diabetes Mellitus after Bariatric Surgery: Fact or Fiction? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16173171. [PMID: 31480306 PMCID: PMC6747427 DOI: 10.3390/ijerph16173171] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/20/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Although type 2 diabetes mellitus (T2DM) has been traditionally viewed as an intractable chronic medical condition, accumulating evidence points towards the notion that a complete remission of T2DM is feasible following a choice of medical and/or surgical interventions. This has been paralleled by increasing interest in the establishment of a universal definition for T2DM remission which, under given circumstances, could be considered equivalent to a “cure”. The efficacy of bariatric surgery in particular for achieving glycemic control has highlighted surgery as a candidate curative intervention for T2DM. Herein, available evidence regarding available surgical modalities and the mechanisms that drive metabolic amelioration after bariatric surgery are reviewed. Furthermore, reports from observational and randomized studies with regard to T2DM remission are reviewed, along with concepts relevant to the variety of definitions used for T2DM remission and other potential sources of discrepancy in success rates among different studies.
Collapse
|
35
|
Boland BB, Mumphrey MB, Hao Z, Townsend RL, Gill B, Oldham S, Will S, Morrison CD, Yu S, Münzberg H, Rhodes CJ, Trevaskis JL, Berthoud HR. Combined loss of GLP-1R and Y2R does not alter progression of high-fat diet-induced obesity or response to RYGB surgery in mice. Mol Metab 2019; 25:64-72. [PMID: 31126840 PMCID: PMC6600699 DOI: 10.1016/j.molmet.2019.05.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/30/2019] [Accepted: 05/04/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Understanding the mechanisms underlying the remarkable beneficial effects of gastric bypass surgery is important for the development of non-surgical therapies or less invasive surgeries in the fight against obesity and metabolic disease. Although the intestinal L-cell hormones glucagon-like peptide-1 (GLP-1) and peptide tyrosine-tyrosine (PYY) have attracted the most attention, direct tests in humans and rodents with pharmacological blockade or genetic deletion of either the GLP1-receptor (GLP1R) or the Y2-receptor (Y2R) were unable to confirm their critical roles in the beneficial effects gastric bypass surgery on body weight and glucose homeostasis. However, new awareness of the power of combinatorial therapies in the treatment of metabolic disease would suggest that combined blockade of more than one signaling pathway may be necessary to reverse the beneficial effects of bariatric surgery. METHODS The metabolic effects of high-fat diet and the ability of Roux-en-Y gastric bypass surgery to lower food intake and body weight, as well as improve glucose handling, was tested in GLP1R and Y2R-double knockout (GLP1RKO/Y2RKO) and C57BL6J wildtype (WT) mice. RESULTS GLP1RKO/Y2RKO and WT mice responded similarly for up to 20 weeks on high-fat diet and 16 weeks after RYGB. There were no significant differences in loss of body and liver weight, fat mass, reduced food intake, relative increase in energy expenditure, improved fasting insulin, glucose tolerance, and insulin tolerance between WT and GLP1RKO/Y2RKO mice after RYGB. CONCLUSIONS Combined loss of GLP1R and Y2R-signaling was not able to negate or attenuate the beneficial effects of RYGB on body weight and glucose homeostasis in mice, suggesting that a larger number of signaling pathways is involved or that the critical pathway has not yet been identified.
Collapse
Affiliation(s)
- Brandon B Boland
- Cardiovascular, Renal and Metabolic Diseases, MedImmune LLC, Gaithersburg, MD, USA
| | | | - Zheng Hao
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | - Benji Gill
- Cardiovascular, Renal and Metabolic Diseases, MedImmune LLC, Gaithersburg, MD, USA
| | - Stephanie Oldham
- Cardiovascular, Renal and Metabolic Diseases, MedImmune LLC, Gaithersburg, MD, USA
| | - Sarah Will
- Cardiovascular, Renal and Metabolic Diseases, MedImmune LLC, Gaithersburg, MD, USA
| | | | - Sangho Yu
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Heike Münzberg
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Christopher J Rhodes
- Cardiovascular, Renal and Metabolic Diseases, MedImmune LLC, Gaithersburg, MD, USA
| | - James L Trevaskis
- Cardiovascular, Renal and Metabolic Diseases, MedImmune LLC, Gaithersburg, MD, USA
| | | |
Collapse
|
36
|
Chakraborty S, Halland M, Burton D, Desai A, Neja B, Low P, Singer W, Camilleri M, Zinsmeister AR, Bharucha AE. GI Dysfunctions in Diabetic Gastroenteropathy, Their Relationships With Symptoms, and Effects of a GLP-1 Antagonist. J Clin Endocrinol Metab 2019; 104:1967-1977. [PMID: 30358871 PMCID: PMC6467444 DOI: 10.1210/jc.2018-01623] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
Abstract
CONTEXT Delayed gastric emptying (GE) is common but often asymptomatic in diabetes. The relationship between symptoms, glycemia, and neurohormonal functions, including glucagonlike peptide 1 (GLP-1), are unclear. OBJECTIVES To assess whether GE disturbances, symptoms during a GE study, and symptoms during enteral lipid infusion explain daily symptoms and whether GLP-1 mediates symptoms during enteral lipid infusion. DESIGN In this randomized controlled trial, GE, enteral lipid infusion, gastrointestinal (GI) symptoms during these assessments, autonomic functions, glycosylated hemoglobin (HbA1c), and daily GI symptoms (2-week Gastroparesis Cardinal Symptom Index diary) were evaluated. During enteral lipid infusion, participants received the GLP-1 antagonist exendin 9-39 or placebo. SETTING Single tertiary referral center. PARTICIPANTS 24 healthy controls and 40 patients with diabetic gastroenteropathy. MAIN OUTCOME MEASURES GE, symptoms during enteral lipid infusion, and the effect of exendin 9-39 on the latter. RESULTS In patients, GE was normal (55%), delayed (33%), or rapid (12%). During lipid infusion, GI symptoms tended to be greater (P = 0.06) in patients with diabetes mellitus (DM) than controls; exendin 9-39 did not affect symptoms. The HbA1c was inversely correlated with the mean symptom score during the GE study (r = -0.46, P = 0.003) and lipid infusion (r = -0.47, P < 0.01). GE and symptoms during GE study accounted for 40% and 32%, respectively, of the variance in daily symptom severity and quality of life. CONCLUSIONS In DM gastroenteropathy, GE and symptoms during a GE study explain daily symptoms. Symptoms during enteral lipid infusion were borderline increased but not reduced by a GLP-1 antagonist.
Collapse
Affiliation(s)
| | - Magnus Halland
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Duane Burton
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Anshuman Desai
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Bridget Neja
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Phillip Low
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Alan R Zinsmeister
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Adil E Bharucha
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
37
|
Gasbjerg LS, Helsted MM, Hartmann B, Jensen MH, Gabe MBN, Sparre-Ulrich AH, Veedfald S, Stensen S, Lanng AR, Bergmann NC, Christensen MB, Vilsbøll T, Holst JJ, Rosenkilde MM, Knop FK. Separate and Combined Glucometabolic Effects of Endogenous Glucose-Dependent Insulinotropic Polypeptide and Glucagon-like Peptide 1 in Healthy Individuals. Diabetes 2019; 68:906-917. [PMID: 30626611 DOI: 10.2337/db18-1123] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/05/2019] [Indexed: 11/13/2022]
Abstract
The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) are secreted postprandially and contribute importantly to postprandial glucose tolerance. In this study, we assessed the individual and combined contributions of endogenous GIP and GLP-1 to the postprandial changes in glucose and glucoregulatory hormones using the novel GIP receptor antagonist GIP(3-30)NH2 and the well-established GLP-1 receptor antagonist exendin(9-39)NH2 During 4-h oral glucose tolerance tests (75 g) combined with an ad libitum meal test, 18 healthy men received on four separate days in randomized, double-blinded order intravenous infusions of A) GIP(3-30)NH2 (800 pmol/kg/min) plus exendin(9-39)NH2 (0-20 min: 1,000 pmol/kg/min; 20-240 min: 450 pmol/kg/min), B) GIP(3-30)NH2, C) exendin(9-39)NH2, and D) saline, respectively. Glucose excursions were significantly higher during A than during B, C, and D, while glucose excursions during B were higher than during C and D. Insulin secretion (assessed by C-peptide/glucose ratio) was reduced by 37 ± 16% (A), 30 ± 17% (B), and 8.6 ± 16% (C) compared with D (mean ± SD). A and C resulted in higher glucagon levels and faster gastric emptying. In conclusion, endogenous GIP affects postprandial plasma glucose excursions and insulin secretion more than endogenous GLP-1, but the hormones contribute additively to postprandial glucose regulation in healthy individuals.
Collapse
Affiliation(s)
- Lærke S Gasbjerg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mads M Helsted
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Bolette Hartmann
- 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
| | - Mette H Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Antag Therapeutics ApS, Copenhagen, Denmark
| | - Maria B N Gabe
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alexander H Sparre-Ulrich
- 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
- Antag Therapeutics ApS, Copenhagen, Denmark
| | - Simon Veedfald
- 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
| | - Signe Stensen
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amalie R Lanng
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Natasha C Bergmann
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Zealand Pharma A/S, Glostrup, Denmark
| | - Mikkel B Christensen
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
- Department of Clinical Medicine, 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
| | - Mette M Rosenkilde
- 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
| | - Filip K Knop
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Debédat J, Amouyal C, Aron-Wisnewsky J, Clément K. Impact of bariatric surgery on type 2 diabetes: contribution of inflammation and gut microbiome? Semin Immunopathol 2019; 41:461-475. [DOI: 10.1007/s00281-019-00738-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/15/2019] [Indexed: 02/06/2023]
|
39
|
Shah M, Laurenti MC, Dalla Man C, Ma J, Cobelli C, Rizza RA, Vella A. Contribution of endogenous glucagon-like peptide-1 to changes in glucose metabolism and islet function in people with type 2 diabetes four weeks after Roux-en-Y gastric bypass (RYGB). Metabolism 2019; 93:10-17. [PMID: 30586575 PMCID: PMC6401231 DOI: 10.1016/j.metabol.2018.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/07/2018] [Accepted: 12/20/2018] [Indexed: 01/03/2023]
Abstract
UNLABELLED Glucagon-Like Peptide-1 (GLP-1) is an insulin secretagogue which is elevated after Roux-en-Y Gastric Bypass (RYGB). However, its contribution to glucose metabolism after RYGB remains uncertain. AIMS We tested the hypothesis that GLP-1 lowers postprandial glucose concentrations and improves β-cell function after RYGB. MATERIALS AND METHODS To address these questions we used a labeled mixed meal to assess glucose metabolism and islet function in 12 obese subjects with type 2 diabetes studied before and four weeks after RYGB. During the post-RYGB study subjects were randomly assigned to receive an infusion of either saline or Exendin-9,39 a competitive antagonist of GLP-1 at its receptor. Exendin-9,39 was infused at 300 pmol/kg/min for 6 h. All subjects underwent RYGB for medically-complicated obesity. RESULTS Exendin-9,39 resulted in increased integrated incremental postprandial glucose concentrations (181 ± 154 vs. 582 ± 129 mmol per 6 h, p = 0.02). In contrast, this was unchanged in the presence of saline (275 ± 88 vs. 315 ± 66 mmol per 6 h, p = 0.56) after RYGB. Exendin-9,39 also impaired β-cell responsivity to glucose but did not alter Disposition Index (DI). CONCLUSIONS These data indicate that the elevated GLP-1 concentrations that occur early after RYGB improve postprandial glucose tolerance by enhancing postprandial insulin secretion.
Collapse
Affiliation(s)
- Meera Shah
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| | - Marcello C Laurenti
- Department of Information Engineering, University of Padua, Via Gradenigo 6A, Padua 35131, Italy.
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padua, Via Gradenigo 6A, Padua 35131, Italy.
| | - Jing Ma
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| | - Claudio Cobelli
- Department of Information Engineering, University of Padua, Via Gradenigo 6A, Padua 35131, Italy.
| | - Robert A Rizza
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW This review examines the hormonal regulation of gastric emptying, a topic of increasing relevance, given the fact that medications that are analogs of some of these hormones or act as agonists at the hormonal receptors, are used in clinical practice for optimizing metabolic control in the treatment of type 2 diabetes and in obesity. RECENT FINDINGS The major effects on gastric emptying result from actions of incretins, particularly gastric inhibitory polypeptide, glucagon-like peptide-1, and peptide tyrosine-tyrosine, the duodenal and pancreatic hormones, motilin, glucagon, and amylin, and the gastric orexigenic hormones, ghrelin and motilin. All of these hormones delay gastric emptying, except for ghrelin and motilin which accelerate gastric emptying. These effects on gastric emptying parallel the effects of the hormones on satiation (by those retarding emptying) and increase appetite by those that accelerate emptying. Indeed, in addition to the effects of these hormones on hypothalamic appetite centers and glycemic control, there is evidence that some of their biological effects are mediated through actions on the stomach, particularly with the glucagon-like peptide-1 analogs or agonists used in treating obesity. SUMMARY Effects of gastrointestinal hormones on gastric emptying are increasingly recognized as important mediators of satiation and postprandial glycemic control.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
41
|
Adams JD, Treiber G, Hurtado MD, Laurenti MC, Dalla Man C, Cobelli C, Rizza RA, Vella A. Increased Rates of Meal Absorption Do Not Explain Elevated 1-Hour Glucose in Subjects With Normal Glucose Tolerance. J Endocr Soc 2018; 3:135-145. [PMID: 30591957 PMCID: PMC6302905 DOI: 10.1210/js.2018-00222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/16/2018] [Indexed: 01/12/2023] Open
Abstract
Context In subjects with normal fasting glucose (NFG) and normal glucose tolerance (NGT), glucose concentrations >155 mg/dL 1 hour after 75 g of oral glucose predict increased risk of progression to diabetes. Recently, it has been suggested that the mechanism underlying this abnormality is increased gut absorption of glucose. Objective We sought to determine the rate of systemic appearance of meal-derived glucose in subjects classified by their 1-hour glucose after a 75-g oral glucose challenge. Design This was a cross-sectional study. Participating subjects underwent a 75-g oral glucose challenge and a labeled mixed meal test. Setting An inpatient clinical research unit at an academic medical center. Participants Thirty-six subjects with NFG/NGT participated in this study. Interventions Subjects underwent an oral glucose tolerance test. Subsequently, they underwent a labeled mixed meal to measure fasting and postprandial glucose metabolism. Main Outcome Measures We examined β-cell function and the rate of meal appearance (Meal Ra) in NFG/NGT subjects. Subsequently, we examined the relationship of peak postchallenge glucose with Meal Ra and indices of β-cell function. Results Peak glucose concentrations correlated inversely with β-cell function. No relationship of Meal Ra with peak postchallenge glucose concentrations was observed. Conclusion In subjects with NFG/NGT, elevated 1-hour peak postchallenge glucose concentrations reflect impaired β-cell function rather than increased systemic meal appearance.
Collapse
Affiliation(s)
- J D Adams
- Division of Endocrinology, Diabetes, and Metabolism, Mayo Clinic, Rochester, Minnesota
| | - Gerlies Treiber
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Maria Daniela Hurtado
- Division of Endocrinology, Diabetes, and Metabolism, Mayo Clinic, Rochester, Minnesota
| | - Marcello C Laurenti
- Department of Information Engineering, Università di Padova, 36131 Padova, Italy
| | - Chiara Dalla Man
- Department of Information Engineering, Università di Padova, 36131 Padova, Italy
| | - Claudio Cobelli
- Department of Information Engineering, Università di Padova, 36131 Padova, Italy
| | - Robert A Rizza
- Division of Endocrinology, Diabetes, and Metabolism, Mayo Clinic, Rochester, Minnesota
| | - Adrian Vella
- Division of Endocrinology, Diabetes, and Metabolism, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
42
|
He R, Yin Y, Yin W, Li Y, Zhao J, Zhang W. Prevention of pancreatic acinar cell carcinoma by Roux-en-Y Gastric Bypass Surgery. Nat Commun 2018; 9:4183. [PMID: 30305629 PMCID: PMC6180124 DOI: 10.1038/s41467-018-06571-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 09/06/2018] [Indexed: 02/08/2023] Open
Abstract
Roux-en-Y Gastric Bypass Surgery (RYGB) prevents the occurrence of pancreatic cell acinar carcinoma (ACC) in male and female Ngn3-Tsc1-/- mice. Ngn3 directed Cre deletion of Tsc1 gene induced the development of pancreatic ACC. The transgenic mice with sham surgery demonstrated a cancer incidence of 96.7 ± 3.35% and survival rate of 67.0 ± 1.4% at the age of 300 days. Metastasis to liver and kidney was observed in 69.7 ± 9.7% and 44.3 ± 8.01% of these animals, respectively. All animals with RYGB performed at the age of 16 weeks survived free of pancreatic ACC up to the age of 300 days. RYGB significantly attenuated the activation of mTORC1 signaling and inhibition of tumor suppressor genes: p21, p27, and p53 in pancreatic ACC. Our studies demonstrate that bariatric surgery may limit the occurrence and growth of pancreatic ACC through the suppression of mTORC1 signaling in pancreas. RYGB shows promise for intervention of both metabolic dysfunction and organ cancer.
Collapse
Affiliation(s)
- Rui He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Key Laboratory of Reproduction and Genetic of Ningxia Hui Autonomous Region, School of Basic Medicine, and People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Shengli Street No.1160, Yinchuan, 750004, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Wenzhen Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Yin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Jing Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109-0346, USA.
| |
Collapse
|
43
|
Laferrère B, Pattou F. Weight-Independent Mechanisms of Glucose Control After Roux-en-Y Gastric Bypass. Front Endocrinol (Lausanne) 2018; 9:530. [PMID: 30250454 PMCID: PMC6140402 DOI: 10.3389/fendo.2018.00530] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/22/2018] [Indexed: 12/14/2022] Open
Abstract
Roux-en-Y gastric bypass results in large and sustained weight loss and resolution of type 2 diabetes in 60% of cases at 1-2 years. In addition to calorie restriction and weight loss, various gastro-intestinal mediated mechanisms, independent of weight loss, also contribute to glucose control. The anatomical re-arrangement of the small intestine after gastric bypass results in accelerated nutrient transit, enhances the release of post-prandial gut hormones incretins and of insulin, alters the metabolism and the entero-hepatic cycle of bile acids, modifies intestinal glucose uptake and metabolism, and alters the composition and function of the microbiome. The amelioration of beta cell function after gastric bypass in individuals with type 2 diabetes requires enteric stimulation. However, beta cell function in response to intravenous glucose stimulus remains severely impaired, even in individuals in full clinical diabetes remission. The permanent impairment of the beta cell may explain diabetes relapse years after surgery.
Collapse
Affiliation(s)
- Blandine Laferrère
- Division of Endocrinology, New York Obesity Nutrition Research Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - François Pattou
- Translational Research on Diabetes, UMR 1190, Inserm, Université Lille, Lille, France
- Endocrine and Metabolic Surgery, CHU Lille, Lille, France
| |
Collapse
|
44
|
Salehi M, Vella A, McLaughlin T, Patti ME. Hypoglycemia After Gastric Bypass Surgery: Current Concepts and Controversies. J Clin Endocrinol Metab 2018; 103:2815-2826. [PMID: 30101281 PMCID: PMC6692713 DOI: 10.1210/jc.2018-00528] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/01/2018] [Indexed: 12/12/2022]
Abstract
CONTEXT Hypoglycemia, occurring after bariatric and other forms of upper gastrointestinal surgery, is increasingly encountered by clinical endocrinologists. The true frequency of this condition remains uncertain, due, in part, to differences in the diagnostic criteria and in the affected populations, as well as relative lack of patient and physician awareness and understanding of this condition. Postbariatric hypoglycemia can be severe and disabling for some patients, with neuroglycopenia (altered cognition, seizures, and loss of consciousness) leading to falls, motor vehicle accidents, and job and income loss. Moreover, repeated episodes of hypoglycemia can result in hypoglycemia unawareness, further impairing safety and requiring the assistance of others to treat hypoglycemia. OBJECTIVE In this review, we summarize and integrate data from studies of patients affected by hypoglycemia after Roux-en-Y gastric bypass (RYGB) surgery, obtained from PubMed searches (1990 to 2017) and reference searches of relevant retrieved articles. Whereas hypoglycemia can also be observed after sleeve gastrectomy and fundoplication, this review is focused on post-RYGB, given the greater body of published clinical studies at present. OUTCOME MEASURES Data addressing specific aspects of diagnosis, pathophysiology, and treatment were reviewed by the authors; when not available, the authors have provided opinions based on clinical experience with this challenging condition. CONCLUSIONS Hypoglycemia, occurring after gastric bypass surgery, is challenging for patients and physicians alike. This review provides a systematic approach to diagnosis and treatment based on the underlying pathophysiology.
Collapse
Affiliation(s)
- Marzieh Salehi
- Diabetes Division, University of Texas Health at San Antonio, San Antonio, Texas
| | | | | | - Mary-Elizabeth Patti
- Research and Clinic Divisions, Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts
- Correspondence and Reprint Requests: Mary-Elizabeth Patti, MD, Research and Clinic Divisions, Joslin Diabetes Center and Harvard Medical School, 1 Joslin Place, Boston, Massachusetts 02215. E-mail:
| |
Collapse
|
45
|
Douros JD, Lewis AG, Smith EP, Niu J, Capozzi M, Wittmann A, Campbell J, Tong J, Wagner C, Mahbod P, Seeley R, D'Alessio DA. Enhanced Glucose Control Following Vertical Sleeve Gastrectomy Does Not Require a β-Cell Glucagon-Like Peptide 1 Receptor. Diabetes 2018; 67:1504-1511. [PMID: 29759973 PMCID: PMC6054432 DOI: 10.2337/db18-0081] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/04/2018] [Indexed: 12/12/2022]
Abstract
Bariatric surgeries, including vertical sleeve gastrectomy (VSG), resolve diabetes in 40-50% of patients. Studies examining the molecular mechanisms underlying this effect have centered on the role of the insulinotropic glucagon-like peptide 1 (GLP-1), in great part because of the ∼10-fold rise in its circulating levels after surgery. However, there is currently debate over the role of direct β-cell signaling by GLP-1 to mediate improved glucose tolerance following surgery. In order to assess the importance of β-cell GLP-1 receptor (GLP-1R) for improving glucose control after VSG, a mouse model of this procedure was developed and combined with a genetically modified mouse line allowing an inducible, β-cell-specific Glp1r knockdown (Glp1rβ-cell-ko). Mice with VSG lost ∼20% of body weight over 30 days compared with sham-operated controls and had a ∼60% improvement in glucose tolerance. Isolated islets from VSG mice had significantly greater insulin responses to glucose than controls. Glp1r knockdown in β-cells caused glucose intolerance in diet-induced obese mice compared with obese controls, but VSG improved glycemic profiles to similar levels during oral and intraperitoneal glucose challenges in Glp1rβ-cell-ko and Glp1rWT mice. Therefore, even though the β-cell GLP-1R seems to be important for maintaining glucose tolerance in obese mice, in these experiments it is dispensable for the improvement in glucose tolerance after VSG. Moreover, the metabolic physiology activated by VSG can overcome the deficits in glucose regulation caused by lack of β-cell GLP-1 signaling in obesity.
Collapse
Affiliation(s)
- Jonathan D Douros
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, NC
| | - Alfor G Lewis
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, and Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Eric P Smith
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, and Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
| | - JingJing Niu
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, NC
| | - Megan Capozzi
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, NC
| | - April Wittmann
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, NC
| | - Jonathan Campbell
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, NC
| | - Jenny Tong
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, NC
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, and Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University, Durham, NC
| | - Constance Wagner
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, and Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
| | - Parinaz Mahbod
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, and Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
| | - Randy Seeley
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, and Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - David A D'Alessio
- Duke Molecular Physiology Institute, Department of Medicine, Duke University, Durham, NC
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, and Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University, Durham, NC
| |
Collapse
|
46
|
Ma J, Vella A. What Has Bariatric Surgery Taught Us About the Role of the Upper Gastrointestinal Tract in the Regulation of Postprandial Glucose Metabolism? Front Endocrinol (Lausanne) 2018; 9:324. [PMID: 29997575 PMCID: PMC6028568 DOI: 10.3389/fendo.2018.00324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/31/2018] [Indexed: 02/06/2023] Open
Abstract
The interaction between the upper gastrointestinal tract and the endocrine system is important in the regulation of metabolism and of weight. The gastrointestinal tract has a heterogeneous cellular content and comprises a variety of cells that elaborate paracrine and endocrine mediators that collectively form the entero-endocrine system. The advent of therapy that utilizes these pathways as well as the association of bariatric surgery with diabetes remission has (re-)kindled interest in the role of the gastrointestinal tract in glucose homeostasis. In this review, we will use the changes wrought by bariatric surgery to provide insights into the various gut-pancreas interactions that maintain weight, regulate satiety, and limit glucose excursions after meal ingestion.
Collapse
Affiliation(s)
- Jing Ma
- Division of Endocrinology and Metabolism, Shanghai Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic College of Medicine, Rochester, NY, United States
| | - Adrian Vella
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic College of Medicine, Rochester, NY, United States
| |
Collapse
|
47
|
Smith EP, Polanco G, Yaqub A, Salehi M. Altered glucose metabolism after bariatric surgery: What's GLP-1 got to do with it? Metabolism 2018; 83:159-166. [PMID: 29113813 DOI: 10.1016/j.metabol.2017.10.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 10/20/2017] [Accepted: 10/25/2017] [Indexed: 01/20/2023]
Abstract
Bariatric surgery is an effective treatment for obesity. The two widely performed weight-loss procedures, Roux-en-Y gastric bypass (GB) and sleeve gastrectomy (SG), alter postprandial glucose pattern and enhance gut hormone secretion immediately after surgery before significant weight loss. This weight-loss independent glycemic effects of GB has been attributed to an accelerated nutrient transit from stomach pouch to the gut and enhanced secretion of insulinotropic gut factors; in particular, glucagon-like peptide-1 (GLP-1). Meal-induced GLP-1 secretion is as much as tenfold higher in patients after GB compared to non-surgical individuals and inhibition of GLP-1 action during meals reduces postprandial hyperinsulinemia after GB two to three times more than that in persons without surgery. Moreover, in a subgroup of patients with the late complication of postprandial hyperinsulinemic hypoglycemia after GB, GLP1R blockade reverses hypoglycemia by reducing meal stimulated insulin secretion. The role of enteroinsular axis activity after SG, an increasingly popular alternative to GB, is less understood but, similar to GB, SG accelerates nutrient delivery to the intestine, improves glucose tolerance, and increases postprandial GLP-1 secretion. This review will focus on the current evidence for and against the role of GLP-1 on glycemic effects of GB and will also highlight differences between GB and SG.
Collapse
Affiliation(s)
- Eric P Smith
- Division of Endocrinology, Diabetes & Metabolism, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| | - Georgina Polanco
- Division of Endocrinology, Diabetes & Metabolism, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Abid Yaqub
- Division of Endocrinology, Diabetes & Metabolism, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Marzieh Salehi
- Division of Endocrinology, Diabetes & Metabolism, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
48
|
Zhai H, Li Z, Peng M, Huang Z, Qin T, Chen L, Li H, Zhang H, Zhang W, Xu G. Takeda G Protein-Coupled Receptor 5-Mechanistic Target of Rapamycin Complex 1 Signaling Contributes to the Increment of Glucagon-Like Peptide-1 Production after Roux-en-Y Gastric Bypass. EBioMedicine 2018; 32:201-214. [PMID: 29859856 PMCID: PMC6020750 DOI: 10.1016/j.ebiom.2018.05.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The mechanism by which Roux-en-Y Gastric Bypass (RYGB) increases the secretion of glucagon-like peptide-1 (GLP-1) remains incompletely defined. Here we investigated whether TGR5-mTORC1 signaling mediates the RYGB-induced alteration in GLP-1 production in mice and human beings. METHODS Circulating bile acids, TGR5-mTORC1 signaling, GLP-1 synthesis and secretion were determined in lean or obese male C57BL/6 mice with or without RYGB operation, as well as in normal glycemic subjects, obese patients with type 2 diabetes before and after RYGB. RESULTS Positive relationships were observed among circulating bile acids, ileal mechanistic target of rapamycin complex 1 (mTORC1) signaling and GLP-1 during changes in energy status in the present study. RYGB increased circulating bile acids, ileal Takeda G protein-coupled receptor 5 (TGR5) and mTORC1 signaling activity, as well as GLP-1 production in both mice and human subjects. Inhibition of ileal mTORC1 signaling by rapamycin significantly attenuated the stimulation of bile acid secretion, TGR5 expression and GLP-1 synthesis induced by RYGB in lean and diet-induced obese mice. GLP-1 production and ileal TGR5-mTORC1 signaling were positively correlated with plasma deoxycholic acid (DCA) in mice. Treatment of STC-1 cells with DCA stimulated the production of GLP-1. This effect was associated with a significant enhancement of TGR5-mTORC1 signaling. siRNA knockdown of mTORC1 or TGR5 abolished the enhancement of GLP-1 synthesis induced by DCA. DCA increased interaction between mTOR-regulatory-associated protein of mechanistic target of rapamycin (Raptor) and TGR5 in STC-1 cells. INTERPRETATION Deoxycholic acid-TGR5-mTORC1 signaling contributes to the up-regulation of GLP-1 production after RYGB.
Collapse
Affiliation(s)
- Hening Zhai
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China; Endoscopy Center, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510630, China
| | - Zhi Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Miao Peng
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Zhaoqi Huang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Tingfeng Qin
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Linxi Chen
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Hanbing Li
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Heng Zhang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China
| | - Weizhen Zhang
- Shenzhen University Diabetes Center, Shenzhen University Health Science Center, Shenzhen, Guangdong 518060, China; Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA.
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
49
|
Shah M, Pham A, Gershuni V, Mundi MS. Curing Diabetes Through Bariatric Surgery: Evolution of Our Understanding. CURRENT SURGERY REPORTS 2018. [DOI: 10.1007/s40137-018-0209-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
50
|
Morrison DJ, Kowalski GM, Grespan E, Mari A, Bruce CR, Wadley GD. Measurement of postprandial glucose fluxes in response to acute and chronic endurance exercise in healthy humans. Am J Physiol Endocrinol Metab 2018; 314:E503-E511. [PMID: 29351488 DOI: 10.1152/ajpendo.00316.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The effect of endurance exercise on enhancing insulin sensitivity and glucose flux has been well established with techniques such as the hyperinsulinemic clamp. Although informative, such techniques do not emulate the physiological postprandial state, and it remains unclear how exercise improves postprandial glycaemia. Accordingly, combining mixed-meal tolerance testing and the triple-stable isotope glucose tracer approach, glucose fluxes [rates of meal glucose appearance (Ra), disposal (Rd), and endogenous glucose production (EGP)] were determined following acute endurance exercise (1 h cycling; ~70% V̇o2max) and 4 wk of endurance training (cycling 5 days/wk). Training was associated with a modest increase in V̇o2max (~7%, P < 0.001). Postprandial glucose and insulin responses were reduced to the same extent following acute and chronic training. Interestingly, this was not accompanied by changes to rates of meal Ra, Rd, or degree of EGP suppression. Glucose clearance (Rd relative to prevailing glucose) was, however, enhanced with acute and chronic exercise. Furthermore, the duration of EGP suppression was shorter with acute and chronic exercise, with EGP returning toward fasting levels more rapidly than pretraining conditions. These findings suggest that endurance exercise influences the efficiency of the glucoregulatory system, where pretraining rates of glucose disposal and production were achieved at lower glucose and insulin levels. Notably, there was no influence of chronic training over and above that of a single exercise bout, providing further evidence that glucoregulatory benefits of endurance exercise are largely attributed to the residual effects of the last exercise bout.
Collapse
Affiliation(s)
- Dale J Morrison
- Deakin University, Geelong, Australia, Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Burwood, Australia
| | - Greg M Kowalski
- Deakin University, Geelong, Australia, Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Burwood, Australia
| | | | - Andrea Mari
- CNR Institute of Neuroscience , Padua , Italy
| | - Clinton R Bruce
- Deakin University, Geelong, Australia, Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Burwood, Australia
| | - Glenn D Wadley
- Deakin University, Geelong, Australia, Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Burwood, Australia
| |
Collapse
|