1
|
Wilbrink JA, van Avesaat M, Nienhuijs SW, Stronkhorst A, Masclee AAM. Changes in gastrointestinal motility and gut hormone secretion after Roux-en-Y gastric bypass and sleeve gastrectomy for individuals with severe obesity. Clin Obes 2025; 15:e12721. [PMID: 39727180 PMCID: PMC11907097 DOI: 10.1111/cob.12721] [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: 11/09/2023] [Accepted: 10/23/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Bariatric surgery is very effective in long-term weight management. The present study was undertaken to investigate the short-term effects of sleeve gastrectomy (SG) and of Roux-en-Y gastric bypass (RYGB) on (a) gastrointestinal (GI) motility, that is gastric emptying and oro-cecal transit time and (b) secretion of regulatory gut peptides and (c) their interrelationship. METHODS Prospective single-centre study in which we assessed gastric emptying, oro-cecal transit time and gut peptide release in 28 severely obese individuals before and 2, respectively, 12 months after bariatric surgery (either SG or RYGB). Plasma PYY, GLP-1, ghrelin, insulin and glucose levels were measured fasting and after intake of a solid standard 459 kcal meal at each occasion. Gastric emptying was measured by 13 C octanoic acid breath testing, and oro-cecal transit time was measured by lactulose H2 breath testing. Satiation was measured using VAS scores. RESULTS After both RYGB and SG gastric emptying become significantly accelerated, and postprandial release of the distal gut peptides GLP-1 and PYY becomes significantly increased, pointing to ileal brake activation. Oro-cecal transit time becomes significantly accelerated after SG but not after RYGB. No significant correlations were observed between changes in distal gut peptide release, changes in GI motility and clinical parameters. CONCLUSION Both SG and RYGB resulted in significant weight loss and significantly affected GI motility and PYY and GLP-1 secretion. Subtle differences between both procedures were found in effect on oro-cecal transit time and patterns of peptide secretion.
Collapse
Affiliation(s)
- Jennifer A. Wilbrink
- Division of Gastroenterology‐HepatologyMaastricht University Medical Center, Maastricht, The Netherlands. NUTRIM—School for Nutrition and Translational Research in MetabolismMaastrichtthe Netherlands
- Department of Gastroenterology‐HepatologyCatharina HospitalEindhoventhe Netherlands
- Department of Gastroenterology‐HepatologyZuyderland Medical Centre Sittard‐GeleenBG Geleenthe Netherlands
| | - Mark van Avesaat
- Division of Gastroenterology‐HepatologyMaastricht University Medical Center, Maastricht, The Netherlands. NUTRIM—School for Nutrition and Translational Research in MetabolismMaastrichtthe Netherlands
- Department of Gastroenterology‐HepatologyZuyderland Medical Centre Sittard‐GeleenBG Geleenthe Netherlands
| | | | - Arnold Stronkhorst
- Department of Gastroenterology‐HepatologyCatharina HospitalEindhoventhe Netherlands
| | - Ad A. M. Masclee
- Division of Gastroenterology‐HepatologyMaastricht University Medical Center, Maastricht, The Netherlands. NUTRIM—School for Nutrition and Translational Research in MetabolismMaastrichtthe Netherlands
| |
Collapse
|
2
|
Yang K, Wu YT, He Y, Dai JX, Luo YL, Xie JH, Ding WJ. GLP-1 and IL-6 regulates obesity in the gut and brain. Life Sci 2025; 362:123339. [PMID: 39730038 DOI: 10.1016/j.lfs.2024.123339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Obesity is a chronic metabolic disease characterized by excessive nutrient intake leading to increased subcutaneous or visceral fat, resulting in pathological and physiological changes. The incidence rate of obesity, an important form of metabolic syndrome, is increasing worldwide. Excess appetite is a key pathogenesis of obesity, and the inflammatory response induced by obesity has received increasing attention. This review focuses on the role of appetite-regulating factor (Glucogan-like peptide 1) and inflammatory factor (Interleukin-6) in the gut and brain in individuals with obesity and draws insights from the current literature.
Collapse
Affiliation(s)
- Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ting Wu
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin-Xiu Dai
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yu-Lu Luo
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jing-Hui Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei-Jun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
3
|
Kupnicka P, Król M, Żychowska J, Łagowski R, Prajwos E, Surówka A, Chlubek D. GLP-1 Receptor Agonists: A Promising Therapy for Modern Lifestyle Diseases with Unforeseen Challenges. Pharmaceuticals (Basel) 2024; 17:1470. [PMID: 39598383 PMCID: PMC11597758 DOI: 10.3390/ph17111470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Modern lifestyle diseases remain a persistent challenge in healthcare. Currently, about 422 million people worldwide are affected by diabetes, while 1 in 8 people are living with obesity. The development of glucagon-like peptide 1 receptor agonists (GLP-1RAs) has marked a significant milestone in treating these conditions. Interest in GLP-1RAs has grown due to evidence that, beyond their established role in diabetes management, these drugs influence other metabolic disorders. This is attributed to the fact that GLP-1 receptors are found in various healthy human tissues. However, a potential cause for concern is the expression of GLP-1 receptors in certain cancers. This review focuses on the most recent findings concerning the actions of GLP-1RAs, detailing their documented impact on the thyroid gland and pancreas. It addresses concerns about the long-term use of GLP-1RAs in relation to the development of pancreatitis, pancreatic cancer, and thyroid neoplasms by exploring the mechanisms and long-term effects in different patient subgroups and including data not discussed previously. This review was conducted through an examination of the literature available in the MedLine (PubMed) database, covering publications from 1978 to 10 May 2024. The collected articles were selected based on their relevance to studies of GLP-1 agonists and their effects on the pancreas and thyroid and assessed to meet the established inclusion criteria. The revised papers suggest that prolonged use of GLP-1RA could contribute to the formation of thyroid tumors and may increase the risk of acute inflammatory conditions such as pancreatitis, particularly in high-risk patients. Therefore, physicians should advise patients on the need for more frequent and detailed follow-ups.
Collapse
Affiliation(s)
- Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Małgorzata Król
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Justyna Żychowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Ryszard Łagowski
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Eryk Prajwos
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Anna Surówka
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
4
|
Holst JJ. GLP-1 physiology in obesity and development of incretin-based drugs for chronic weight management. Nat Metab 2024; 6:1866-1885. [PMID: 39160334 DOI: 10.1038/s42255-024-01113-9] [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] [Received: 01/30/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024]
Abstract
The introduction of the highly potent incretin receptor agonists semaglutide and tirzepatide has marked a new era in the treatment of type 2 diabetes and obesity. With normalisation of glycated haemoglobin levels and weight losses around 15-25%, therapeutic goals that were previously unrealistic are now within reach, and clinical trials have documented that these effects are associated with reduced risk of cardiovascular events and premature mortality. Here, I review this remarkable development from the earliest observations of glucose lowering and modest weight losses with native glucagon-like peptide (GLP)-1 and short acting compounds, to the recent development of highly active formulations and new molecules. I will classify these agents as GLP-1-based therapies in the understanding that these compounds or combinations may have actions on other receptors as well. The physiology of GLP-1 is discussed as well as its mechanisms of actions in obesity, in particular, the role of sensory afferents and GLP-1 receptors in the brain. I provide details regarding the development of GLP-1 receptor agonists for anti-obesity therapy and discuss the possible mechanism behind their beneficial effects on adverse cardiovascular events. Finally, I highlight new pharmacological developments, including oral agents, and discuss important questions regarding maintenance therapy.
Collapse
Affiliation(s)
- Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences. Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
5
|
Dziakova J, Torres A, Odovic M, Esteban JM, Vázquez-Romero M, Castillo A, Sánchez-Pernaute A, Gagner M. Spanish Experience with Latero-Lateral Duodeno-Ileostomy + Sleeve Gastrectomy with Magnet Anastomosis System. Obes Surg 2024; 34:3569-3575. [PMID: 39093385 DOI: 10.1007/s11695-024-07432-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND The partial diversion of intestinal contents facilitates achieving and maintaining weight loss and improving glycemic control in patients with obesity and with or without T2DM. The purpose of this study is to report our experience and 1-year follow-up with novel modification of SADI-S. METHODS This study is a part of a multicentric trial of patients that underwent primary side-to-side duodeno-ileostomy and sleeve gastrectomy (SG) with GT metabolic solutions magnetic anastomosis system. Feasibility, safety, and initial efficacy were evaluated. RESULTS The mean age of the patients included was 48 ± 8.75 years and the preoperative BMI was 43.32 ± 2.82 kg/m2. The complications were present in 30% of patients. The anastomosis patency was confirmed by the passage of radiological contrast under fluoroscopy at a mean of 17 days (17-29 days), and the mean expulsion time was 42 days (32-62). The mean diameter of the anastomosis after the magnet expulsion was 13.8 × 11.4 mm. The percentage of total weight lost at 1 year was 38.68 ± 8.48% (p < 0.001). The percentage of excess weight loss 82.5 ± 18.44% (p < 0.001) and improvements in glucose profiles were observed. Mean baseline HbA1c 5.77 ± 0.31% was reduced to 5.31 ± 0.26% (p < 0.024). CONCLUSIONS Latero-lateral duodeno-ileostomy + SG with magnetic duodenal bipartition is afeasible and reasonably safe technique and induces weight loss in patients with obesity and improvement of glycemic control. This modification could be considered as an option to standard SADI-S or as a first step in two stages procedure. However, larger studies are needed. TRIAL REGISTRATION Clinicaltrials.gov Identifier: #NCT05322122.
Collapse
Affiliation(s)
- Jana Dziakova
- Department of Surgery, Hospital Clínico San Carlos, C/ Prof Martín Lago S/N 28040, Madrid, Spain.
- Department of Surgery, School of Medicine, Complutense University, Madrid, Spain.
- San Carlos Health Research Institute (IdISSC), Madrid, Spain.
| | - Antonio Torres
- Department of Surgery, Hospital Clínico San Carlos, C/ Prof Martín Lago S/N 28040, Madrid, Spain
- Department of Surgery, School of Medicine, Complutense University, Madrid, Spain
- San Carlos Health Research Institute (IdISSC), Madrid, Spain
| | - Maja Odovic
- Department of Surgery, Hospital Clínico San Carlos, C/ Prof Martín Lago S/N 28040, Madrid, Spain
| | | | | | - Andrea Castillo
- Department of Surgery, Hospital Clínico San Carlos, C/ Prof Martín Lago S/N 28040, Madrid, Spain
| | - Andrés Sánchez-Pernaute
- Department of Surgery, Hospital Clínico San Carlos, C/ Prof Martín Lago S/N 28040, Madrid, Spain
- Department of Surgery, School of Medicine, Complutense University, Madrid, Spain
- San Carlos Health Research Institute (IdISSC), Madrid, Spain
| | - Michel Gagner
- Department of Surgery, Hôpital du Sacre Coeur, Montreal, Canada
- Department of Surgery, Westmount Square Surgical Center, Westmount, Canada
| |
Collapse
|
6
|
Sahasrabudhe A, Rupprecht LE, Orguc S, Khudiyev T, Tanaka T, Sands J, Zhu W, Tabet A, Manthey M, Allen H, Loke G, Antonini MJ, Rosenfeld D, Park J, Garwood IC, Yan W, Niroui F, Fink Y, Chandrakasan A, Bohórquez DV, Anikeeva P. Multifunctional microelectronic fibers enable wireless modulation of gut and brain neural circuits. Nat Biotechnol 2024; 42:892-904. [PMID: 37349522 PMCID: PMC11180606 DOI: 10.1038/s41587-023-01833-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/23/2023] [Indexed: 06/24/2023]
Abstract
Progress in understanding brain-viscera interoceptive signaling is hindered by a dearth of implantable devices suitable for probing both brain and peripheral organ neurophysiology during behavior. Here we describe multifunctional neural interfaces that combine the scalability and mechanical versatility of thermally drawn polymer-based fibers with the sophistication of microelectronic chips for organs as diverse as the brain and the gut. Our approach uses meters-long continuous fibers that can integrate light sources, electrodes, thermal sensors and microfluidic channels in a miniature footprint. Paired with custom-fabricated control modules, the fibers wirelessly deliver light for optogenetics and transfer data for physiological recording. We validate this technology by modulating the mesolimbic reward pathway in the mouse brain. We then apply the fibers in the anatomically challenging intestinal lumen and demonstrate wireless control of sensory epithelial cells that guide feeding behaviors. Finally, we show that optogenetic stimulation of vagal afferents from the intestinal lumen is sufficient to evoke a reward phenotype in untethered mice.
Collapse
Affiliation(s)
- Atharva Sahasrabudhe
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura E Rupprecht
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Sirma Orguc
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tural Khudiyev
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tomo Tanaka
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Secure System Platform Research Laboratories, NEC Corporation, Kawasaki, Japan
| | - Joanna Sands
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Weikun Zhu
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony Tabet
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie Manthey
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Harrison Allen
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gabriel Loke
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marc-Joseph Antonini
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Dekel Rosenfeld
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jimin Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Indie C Garwood
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT Health Sciences and Technology Graduate Program, Cambridge, MA, USA
| | - Wei Yan
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Farnaz Niroui
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoel Fink
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anantha Chandrakasan
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Diego V Bohórquez
- Laboratory of Gut Brain Neurobiology, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
7
|
Gómez-Maqueo A, Ferreira-Lazarte A, Amirruddin NS, Lin AHM. Generating slow digestibility in cooked potatoes by modulating starch accessibility to α-amylase and mucosal α-glucosidase to different levels. Food Hydrocoll 2023. [DOI: 10.1016/j.foodhyd.2023.108718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
8
|
Conley TE, White KL, Bond A, Harrison S, McLaughlin J, Lal S. Emerging uses of glucagon-like peptide 1 (GLP-1) receptor agonists following ileal resection: literature review and case examples. Frontline Gastroenterol 2023; 14:521-526. [PMID: 37854779 PMCID: PMC10579548 DOI: 10.1136/flgastro-2023-102402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/18/2023] [Indexed: 10/20/2023] Open
Abstract
Following ileal resection, the combination of severe bile acid (BA) malabsorption, rapid small bowel transit and unrestricted upper gastrointestinal (GI) secretion results in severe diarrhoea that can prove refractory to pharmacological therapies. While established therapies, including BA sequestrants and antidiarrhoeal drugs seek to ameliorate symptoms, they do not target the underlying pathophysiological mechanisms in this patient group. Their use can also be limited by both intolerance and adverse effects. The novel use of glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) in these patients may allow restoration of the physiological negative feedback mechanisms lost in ileal resection and reduce diarrhoea by prolonging small bowel transit time, limiting upper GI secretions and perhaps by inhibiting hepatic BA synthesis. While recent evidence supports the use of GLP-1 RAs as a safe and effective therapy for bile acid diarrhoea (BAD), it remains uncertain whether those with severe BAD and subsequent short bowel syndrome secondary to extensive ileal resection will benefit. Here, we present three cases of severe diarrhoea secondary to extensive ileal resection in which the use of the GLP-1 RA, liraglutide, was well tolerated and resulted in an objective improvement in diarrhoeal symptoms. We further provide a narrative review of the emerging evidence base supporting the use of GLP therapies in this challenging condition.
Collapse
Affiliation(s)
| | | | - Ashley Bond
- Intestinal Failure Unit, Salford Royal NHS Foundation Trust, Salford, UK
| | - Simon Harrison
- Intestinal Failure Unit, Salford Royal NHS Foundation Trust, Salford, UK
| | - John McLaughlin
- Gastroenterology, Salford Royal Hospitals NHS Trust, Salford, UK
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Simon Lal
- Intestinal Failure Unit, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|
9
|
Becetti I, Bwenyi EL, de Araujo IE, Ard J, Cryan JF, Farooqi IS, Ferrario CR, Gluck ME, Holsen LM, Kenny PJ, Lawson EA, Lowell BB, Schur EA, Stanley TL, Tavakkoli A, Grinspoon SK, Singhal V. The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets: A Report from the 23rd Annual Harvard Nutrition Obesity Symposium. Am J Clin Nutr 2023; 118:314-328. [PMID: 37149092 PMCID: PMC10375463 DOI: 10.1016/j.ajcnut.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023] Open
Abstract
Obesity is increasing at an alarming rate. The effectiveness of currently available strategies for the treatment of obesity (including pharmacologic, surgical, and behavioral interventions) is limited. Understanding the neurobiology of appetite and the important drivers of energy intake (EI) can lead to the development of more effective strategies for the prevention and treatment of obesity. Appetite regulation is complex and is influenced by genetic, social, and environmental factors. It is intricately regulated by a complex interplay of endocrine, gastrointestinal, and neural systems. Hormonal and neural signals generated in response to the energy state of the organism and the quality of food eaten are communicated by paracrine, endocrine, and gastrointestinal signals to the nervous system. The central nervous system integrates homeostatic and hedonic signals to regulate appetite. Although there has been an enormous amount of research over many decades regarding the regulation of EI and body weight, research is only now yielding potentially effective treatment strategies for obesity. The purpose of this article is to summarize the key findings presented in June 2022 at the 23rd annual Harvard Nutrition Obesity Symposium entitled "The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets." Findings presented at the symposium, sponsored by NIH P30 Nutrition Obesity Research Center at Harvard, enhance our current understanding of appetite biology, including innovative techniques used to assess and systematically manipulate critical hedonic processes, which will shape future research and the development of therapeutics for obesity prevention and treatment.
Collapse
Affiliation(s)
- Imen Becetti
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States.
| | - Esther L Bwenyi
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Ivan E de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Jamy Ard
- Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Bariatric and Weight Management Center, Wake Forest Baptist Health, Winston-Salem, NC, United States; Center on Diabetes, Obesity, and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Hypertension and Vascular Research Center, Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Maya Angelou Center for Healthy Equity, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ismaa Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom; Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom; Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Carrie R Ferrario
- Department of Pharmacology, Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI, United States
| | - Marci E Gluck
- National Institutes of Health, Phoenix, AZ, United States; National Institute of Diabetes and Digestive and Kidney Disease, Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, Phoenix, AZ, United States
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, United States; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Elizabeth A Lawson
- Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Ellen A Schur
- Division of General Internal Medicine, University of Washington, Seattle, WA, United States; Univeristy of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States; Univeristy of Washington Nutrition and Obesity Research Center, University of Washington, Seattle, WA, United States; Clinical and Translational Research Services Core, University of Washington, Seattle, WA, United States
| | - Takara L Stanley
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Ali Tavakkoli
- Division of General and Gastrointestinal (GI) Surgery, Center for Weight Management and Wellness, Advanced Minimally Invasive Fellowship, Harvard Medical School, Boston, MA, United States
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Vibha Singhal
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Pediatric Endocrinology and Obesity Medicine, Massachusetts General Hospital, Boston, MA, United States; Pediatric Program MGH Weight Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
10
|
Kamakura R, Raza GS, Sodum N, Lehto V, Kovalainen M, Herzig K. Colonic Delivery of Nutrients for Sustained and Prolonged Release of Gut Peptides: A Novel Strategy for Appetite Management. Mol Nutr Food Res 2022; 66:e2200192. [PMID: 35938221 PMCID: PMC9787473 DOI: 10.1002/mnfr.202200192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/17/2022] [Indexed: 12/30/2022]
Abstract
Obesity is one of the major global threats to human health and risk factors for cardiometabolic diseases and certain cancers. Glucagon-like peptide-1 (GLP-1) plays a major role in appetite and glucose homeostasis and recently the USFDA approved GLP-1 agonists for the treatment of obesity and type 2 diabetes. GLP-1 is secreted from enteroendocrine L-cells in the distal part of the gastrointestinal (GI) tract in response to nutrient ingestion. Endogenously released GLP-1 has a very short half-life of <2 min and most of the nutrients are absorbed before reaching the distal GI tract and colon, which hinders the use of nutritional compounds for appetite regulation. The review article focuses on nutrients that endogenously stimulate GLP-1 and peptide YY (PYY) secretion via their receptors in order to decrease appetite as preventive action. In addition, various delivery technologies such as pH-sensitive, mucoadhesive, time-dependent, and enzyme-sensitive systems for colonic targeting of nutrients delivery are described. Sustained colonic delivery of nutritional compounds could be one of the most promising approaches to prevent obesity and associated metabolic diseases by, e.g., sustained GLP-1 release.
Collapse
Affiliation(s)
- Remi Kamakura
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Ghulam Shere Raza
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Nalini Sodum
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Vesa‐Pekka Lehto
- Department of Applied PhysicsFaculty of Science and ForestryUniversity of Eastern FinlandKuopioFI‐70211Finland
| | - Miia Kovalainen
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
| | - Karl‐Heinz Herzig
- Research Unit of BiomedicineFaculty of Medicine, and Medical Research CenterUniversity of Oulu and Oulu University HospitalOulu90220Finland
- Department of Pediatric Gastroenterology and Metabolic DiseasesPediatric InstitutePoznan University of Medical SciencesPoznań60–572Poland
| |
Collapse
|
11
|
Allard C, Cota D. Paracrine actions of glucagon-like peptide 1 in the gut unraveled. LIFE METABOLISM 2022; 1:6-7. [PMID: 39872692 PMCID: PMC11749695 DOI: 10.1093/lifemeta/loac010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 01/30/2025]
Affiliation(s)
- Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215 Bordeaux, France
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215 Bordeaux, France
| |
Collapse
|
12
|
Zhang T, Perkins MH, Chang H, Han W, de Araujo IE. An inter-organ neural circuit for appetite suppression. Cell 2022; 185:2478-2494.e28. [PMID: 35662413 PMCID: PMC9433108 DOI: 10.1016/j.cell.2022.05.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/31/2022] [Accepted: 05/09/2022] [Indexed: 02/03/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) is a signal peptide released from enteroendocrine cells of the lower intestine. GLP-1 exerts anorectic and antimotility actions that protect the body against nutrient malabsorption. However, little is known about how intestinal GLP-1 affects distant organs despite rapid enzymatic inactivation. We show that intestinal GLP-1 inhibits gastric emptying and eating via intestinofugal neurons, a subclass of myenteric neurons that project to abdominal sympathetic ganglia. Remarkably, cell-specific ablation of intestinofugal neurons eliminated intestinal GLP-1 effects, and their chemical activation functioned as a GLP-1 mimetic. GLP-1 sensing by intestinofugal neurons then engaged a sympatho-gastro-spinal-reticular-hypothalamic pathway that links abnormal stomach distension to craniofacial programs for food rejection. Within this pathway, cell-specific activation of discrete neuronal populations caused systemic GLP-1-like effects. These molecularly identified, delimited enteric circuits may be targeted to ameliorate the abdominal bloating and loss of appetite typical of gastric motility disorders.
Collapse
Affiliation(s)
- Tong Zhang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA,Department of Colorectal Surgery, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong 510180, China,Jinan University, Guangzhou, Guangdong 510632, China
| | - Matthew H. Perkins
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Hao Chang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Wenfei Han
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA,Correspondence: (W.H.), (I.E.d.A.)
| | - Ivan E. de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA,Artificial Intelligence and Emerging Technologies in Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA,Lead contact,Correspondence: (W.H.), (I.E.d.A.)
| |
Collapse
|
13
|
Activation of gastrointestinal ileal brake response with dietary slowly digestible carbohydrates, with no observed effect on subjective appetite, in an acute randomized, double-blind, crossover trial. Eur J Nutr 2022; 61:1965-1980. [PMID: 35079895 DOI: 10.1007/s00394-021-02770-2] [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: 05/07/2021] [Accepted: 12/03/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE To test the hypothesis that oral ingestion of slowly digestible carbohydrates (SDCs) that reach the ileum triggers the ileal brake as indicated by delayed gastric emptying, reduced glycemic response, and decreased subjective appetite. METHODS The study was a five-arm, randomized, double-blind, crossover trial with a 1-week washout period between treatments (n = 20; 9 females, 11 males). Five treatments consisted of three SDC ingredients [raw corn starch, isomaltooligosaccharide (IMO), sucromalt], and an IMO/sucromalt combination, shown in vitro to have slow and extended digestion profiles, and a rapidly digestible carbohydrate control (maltodextrin). Carbohydrates (26 g) were incorporated into yogurt [300 g total; carbohydrate (~ 77 g), fat (~ 0.2 g), and protein (~ 9 g)] with closely matched energy content (346 kcal) and viscosity (~ 30,000 cP). Outcomes were measured in a 4 h postprandial period. RESULTS Mean gastric half-emptying times were moderately though significantly increased for the raw corn starch and IMO treatments (P < 0.05), but they could be sub-divided into larger effect responder (n = 11) and non-responder groups (n = 9). Longer time for glycemic response to return to baseline was associated with increased gastric half-emptying time in an exploratory subset of data removing gastric half-emptying times > 3.5 h (P = 0.02). No significant differences in appetite ratings were observed. CONCLUSION SDCs caused slower gastric emptying rate through activation of the ileal brake, as closely matched semi-solid yogurts were used and only rate of carbohydrate digestion differed. Extending glycemic response through consumption of SDCs was associated with triggering the ileal brake. TRIAL REGISTRATION ClinicalTrials.gov NCT03630445, August 2018, retrospectively registered.
Collapse
|
14
|
Janani L, Bamehr H, Tanha K, Mirzabeigi P, Montazeri H, Tarighi P. Effects of Sitagliptin as Monotherapy and Add-On to Metformin on Weight Loss among Overweight and Obese Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis. Drug Res (Stuttg) 2021; 71:477-488. [PMID: 34388848 DOI: 10.1055/a-1555-2797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Sitagliptin is known as an antidiabetic agent inhibiting the dipeptidyl peptidase-4. Although sitagliptin may influence weight, controversial results have been reported, and there is no general agreement on this issue. Therefore, this study assessed the effect of sitagliptin as monotherapy and add-on therapy to metformin on weight reduction in overweight or obese cases with type 2 diabetes. METHODS We reviewed the following databases to identify all relevant papers published until 1st April 2021: Web of Science, MEDLINE, Embase, Scopus, Cochrane Central Register of Controlled Trials Cochrane Library, and Google Scholar. The research included all clinical trials investigating the effect of sitagliptin in obese or overweight adult patients with type 2 diabetes without any language restriction. RESULTS In total, eighteen randomized controlled trials with 2009 participants were included in our meta-analysis. Results showed supplementation of sitagliptin has led to weight loss for sitagliptin treated (MD -0.99; 95% CI; (-1.87, -0.12); p=0.026)) and sitagliptin+metformin treated groups (MD -1.09; 95% CI; (-1.69, -0.49); p<0.001)). Also, the intervention has influenced body mass index in sitagliptin treated (MD -0.23; 95% CI; (-0.45, 0.02); p=0.033)) and sitagliptin+metformin treated groups (MD -0.52; 95% CI; (-0.96, 0.08); p=0.020)) comparing to placebo. CONCLUSION Our results demonstrated that sitagliptin administration with or without metformin might reduce the body weight and body mass index if these drugs are taken for more than 6 months.
Collapse
Affiliation(s)
- Leila Janani
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Hadi Bamehr
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Kiarash Tanha
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Parastoo Mirzabeigi
- Department of Clinical Pharmacy, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Montazeri
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Van Liefferinge E, Müller M, Van Noten N, Degroote J, Niknafs S, Roura E, Michiels J. Cinnamaldehyde Induces Release of Cholecystokinin and Glucagon-Like Peptide 1 by Interacting with Transient Receptor Potential Ankyrin 1 in a Porcine Ex-Vivo Intestinal Segment Model. Animals (Basel) 2021; 11:2262. [PMID: 34438718 PMCID: PMC8388503 DOI: 10.3390/ani11082262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/17/2022] Open
Abstract
Cinnamaldehyde and capsaicin have been reported to exert effects on the gastric function, mediated by the interaction with transient receptor potential ankyrin channel 1 (TRPA1) and transient receptor potential vanilloid channel 1 (TRPV1), respectively. This study examined whether these compounds could trigger the release of cholecystokinin (CCK) and/or glucagon-like peptide 1 (GLP-1) in the pig's gut in a porcine ex-vivo intestinal segment model. Furthermore, it was verified whether this response was mediated by TRPA1 or TRPV1 by using the channel's antagonist. These gut peptides play a key role in the "intestinal brake", a feedback mechanism that influences the function of proximal parts of the gut. Structural analogues of cinnamaldehyde were screened as well, to explore structure-dependent activation. Results showed a significant effect of capsaicin on GLP-1 release in the proximal small intestine, TRPV1 independent. TRPA1 showed to be strongly activated by cinnamaldehyde, both in proximal and distal small intestine, evidenced by the release of CCK and GLP-1, respectively. Out of all structural derivates, cinnamaldehyde showed the highest affinity for TRPA1, which elucidates the importance of the α,β-unsaturated aldehyde moiety. In conclusion, cinnamaldehyde as a TRPA1 agonist, is a promising candidate to modulate gastric function, by activating intestinal brake mechanisms.
Collapse
Affiliation(s)
- Elout Van Liefferinge
- Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Ghent University, 9000 Ghent, Belgium; (N.V.N.); (J.D.); (J.M.)
| | - Maximiliano Müller
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.M.); (S.N.); (E.R.)
| | - Noémie Van Noten
- Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Ghent University, 9000 Ghent, Belgium; (N.V.N.); (J.D.); (J.M.)
| | - Jeroen Degroote
- Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Ghent University, 9000 Ghent, Belgium; (N.V.N.); (J.D.); (J.M.)
| | - Shahram Niknafs
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.M.); (S.N.); (E.R.)
| | - Eugeni Roura
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St. Lucia, QLD 4072, Australia; (M.M.); (S.N.); (E.R.)
| | - Joris Michiels
- Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Ghent University, 9000 Ghent, Belgium; (N.V.N.); (J.D.); (J.M.)
| |
Collapse
|
16
|
von Molitor E, Riedel K, Krohn M, Hafner M, Rudolf R, Cesetti T. Sweet Taste Is Complex: Signaling Cascades and Circuits Involved in Sweet Sensation. Front Hum Neurosci 2021; 15:667709. [PMID: 34239428 PMCID: PMC8258107 DOI: 10.3389/fnhum.2021.667709] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Sweetness is the preferred taste of humans and many animals, likely because sugars are a primary source of energy. In many mammals, sweet compounds are sensed in the tongue by the gustatory organ, the taste buds. Here, a group of taste bud cells expresses a canonical sweet taste receptor, whose activation induces Ca2+ rise, cell depolarization and ATP release to communicate with afferent gustatory nerves. The discovery of the sweet taste receptor, 20 years ago, was a milestone in the understanding of sweet signal transduction and is described here from a historical perspective. Our review briefly summarizes the major findings of the canonical sweet taste pathway, and then focuses on molecular details, about the related downstream signaling, that are still elusive or have been neglected. In this context, we discuss evidence supporting the existence of an alternative pathway, independent of the sweet taste receptor, to sense sugars and its proposed role in glucose homeostasis. Further, given that sweet taste receptor expression has been reported in many other organs, the physiological role of these extraoral receptors is addressed. Finally, and along these lines, we expand on the multiple direct and indirect effects of sugars on the brain. In summary, the review tries to stimulate a comprehensive understanding of how sweet compounds signal to the brain upon taste bud cells activation, and how this gustatory process is integrated with gastro-intestinal sugar sensing to create a hedonic and metabolic representation of sugars, which finally drives our behavior. Understanding of this is indeed a crucial step in developing new strategies to prevent obesity and associated diseases.
Collapse
Affiliation(s)
- Elena von Molitor
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| | | | | | - Mathias Hafner
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Tiziana Cesetti
- Institute of Molecular and Cell Biology, Hochschule Mannheim, Mannheim, Germany
| |
Collapse
|
17
|
Maselli DB, Camilleri M. Effects of GLP-1 and Its Analogs on Gastric Physiology in Diabetes Mellitus and Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1307:171-192. [PMID: 32077010 DOI: 10.1007/5584_2020_496] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The processing of proglucagon in intestinal L cells results in the formation of glucagon, GLP-1, and GLP-2. The GLP-1 molecule becomes active through the effect of proconvertase 1, and it is inactivated by dipeptidyl peptidase IV (DPP-IV), so that the half-life of endogenous GLP-1 is 2-3 min. GLP-1 stimulates insulin secretion from β cells in the islets of Langerhans. Human studies show that infusion of GLP-1 results in slowing of gastric emptying and increased fasting and postprandial gastric volumes. Retardation of gastric emptying reduces postprandial glycemia. Exendin-4 is a peptide agonist of the GLP-1 receptor that promotes insulin secretion. Chemical modifications of exendin-4 and GLP-1 molecules have been accomplished to prolong the half-life of GLP-1 agonists or analogs. This chapter reviews the effects of GLP-1-related drugs used in treatment of diabetes or obesity on gastric motor functions, chiefly gastric emptying. The literature shows that diverse methods have been used to measure effects of the GLP-1-related drugs on gastric emptying, with most studies using the acetaminophen absorption test which essentially measures gastric emptying of liquids during the first hour and capacity to absorb the drug over 4-6 h, expressed as AUC. The most valid measurements by scintigraphy (solids or liquids) and acetaminophen absorption at 30 or 60 min show that GLP-1-related drugs used in diabetes or obesity retard gastric emptying, and this is associated with reduced glycemia and variable effects on food intake and appetite. GLP-1 agonists and analogs are integral to the management of patients with type 2 diabetes mellitus and obesity. The effects on gastric emptying are reduced with long-acting preparations or long-term use of short-acting preparations as a result of tachyphylaxis. The dual agonists targeting GLP-1 and another receptor (GIP) do not retard gastric emptying, based on reports to date. In summary, GLP-1 agonists and analogs are integral to the management of patients with type 2 diabetes mellitus and obesity, and their effects are mediated, at least in part, by retardation of gastric emptying.
Collapse
Affiliation(s)
- Daniel B Maselli
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
An alternative pathway for sweet sensation: possible mechanisms and physiological relevance. Pflugers Arch 2020; 472:1667-1691. [PMID: 33030576 DOI: 10.1007/s00424-020-02467-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
Abstract
Sweet substances are detected by taste-bud cells upon binding to the sweet-taste receptor, a T1R2/T1R3 heterodimeric G protein-coupled receptor. In addition, experiments with mouse models lacking the sweet-taste receptor or its downstream signaling components led to the proposal of a parallel "alternative pathway" that may serve as metabolic sensor and energy regulator. Indeed, these mice showed residual nerve responses and behavioral attraction to sugars and oligosaccharides but not to artificial sweeteners. In analogy to pancreatic β cells, such alternative mechanism, to sense glucose in sweet-sensitive taste cells, might involve glucose transporters and KATP channels. Their activation may induce depolarization-dependent Ca2+ signals and release of GLP-1, which binds to its receptors on intragemmal nerve fibers. Via unknown neuronal and/or endocrine mechanisms, this pathway may contribute to both, behavioral attraction and/or induction of cephalic-phase insulin release upon oral sweet stimulation. Here, we critically review the evidence for a parallel sweet-sensitive pathway, involved signaling mechanisms, neural processing, interactions with endocrine hormonal mechanisms, and its sensitivity to different stimuli. Finally, we propose its physiological role in detecting the energy content of food and preparing for digestion.
Collapse
|
19
|
van Erp RJJ, de Vries S, van Kempen TATG, Den Hartog LA, Gerrits WJJ. Feed intake patterns nor growth rates of pigs are affected by dietary resistant starch, despite marked differences in digestion. Animal 2020; 14:1402-1412. [PMID: 31852553 DOI: 10.1017/s1751731119002945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Current feed evaluation systems often assume that fermented starch (i.e. resistant starch (RS)) yields less energy than digested starch. However, growth rates of pigs fed low and high RS diets are often the same when feed is available ad libitum. This may be explained by its effect on digestive processes changing feeding behavior, and consequently energy utilization. This study aims to investigate the effect of RS on nutrient digestion and digesta passage rate in pigs, in combination with its effect on feeding behavior and growth performance under ad libitum conditions. In experiment 1, 20 male pigs (40 ± 2.82 kg) were fed diets containing either 50% waxy maize starch (low in RS (LRS)) or high-amylose maize starch (high in RS (HRS)), and soluble and insoluble indigestible markers. After 14 days of adaptation to the diets, pigs were fed hourly to reach steady state (6 h), dissected, and digesta were collected from eight segments. From the collected samples, nutrient digestion and passage rate of the solid and liquid digesta fraction were determined. In experiment 2, 288 pigs (80 ± 0.48 kg; sex ratio per pen 1 : 1; boar : gilt) were housed in groups of 6. Pigs were ad libitum-fed one of the experimental diets, and slaughtered at approximately 115 kg. Feed intake, growth and carcass parameters were measured. Ileal starch digestibility was greater for LRS-fed than for HRS-fed pigs (98.0% v. 74.0%; P < 0.001), where the additional undigested starch in HRS-fed pigs was fermented in the large intestine. No effects of RS on digesta passage rate of the solid or liquid digesta fraction and on feeding behavior were observed. Growth rate and feed intake did not differ between diets, whereas feed efficiency of HRS-fed pigs was 1%-unit higher than that of LRS-fed pigs (P = 0.041). The efficiency of feed used for carcass gain did not differ between diets indicating that the difference in feed efficiency was determined by the non-carcass fraction. Despite a 30% greater RS intake (of total starch) with HRS than with LRS, carcass gain and feed efficiency used for carcass gain were unaffected. RS did not affect digesta passage rate nor feeding behavior suggesting that the difference in energy intake between fermented and digested starch is compensated for post-absorptively. Our results indicate that the net energy value of fermented starch currently used in pig feed evaluation systems is underestimated and should be reconsidered.
Collapse
Affiliation(s)
- R J J van Erp
- Department of Research & Development, Trouw Nutrition, Stationsstraat 77, 3800 AGAmersfoort, The Netherlands
- Animal Nutrition Group, Wageningen University, P.O. Box 338, 6700 AHWageningen, The Netherlands
| | - S de Vries
- Animal Nutrition Group, Wageningen University, P.O. Box 338, 6700 AHWageningen, The Netherlands
| | - T A T G van Kempen
- Department of Research & Development, Trouw Nutrition, Stationsstraat 77, 3800 AGAmersfoort, The Netherlands
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - L A Den Hartog
- Department of Research & Development, Trouw Nutrition, Stationsstraat 77, 3800 AGAmersfoort, The Netherlands
- Animal Nutrition Group, Wageningen University, P.O. Box 338, 6700 AHWageningen, The Netherlands
| | - W J J Gerrits
- Animal Nutrition Group, Wageningen University, P.O. Box 338, 6700 AHWageningen, The Netherlands
| |
Collapse
|
20
|
Hollanda Martins Da Rocha M, Lee ADW, Marin MLDM, Faintuch S, Mishaly A, Faintuch J. Treating short bowel syndrome with pharmacotherapy. Expert Opin Pharmacother 2020; 21:709-720. [DOI: 10.1080/14656566.2020.1724959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mariana Hollanda Martins Da Rocha
- Clinical nutrition multidisciplinary team (MDT), Hospital das Clinicas, Sao Paulo, Brazil
- Head, Short bowel syndrome MDT, Hospital das Clinicas, Sao Paulo, Brazil
| | - André Dong Won Lee
- Clinical nutrition multidisciplinary team (MDT), Hospital das Clinicas, Sao Paulo, Brazil
- Liver and Digestive Organs Transplantation Service, Hospital das Clinicas, Department of Gastroenterology, Hospital das Clinicas, Sao Paulo, Brazil
| | - Marcia Lucia De Mario Marin
- Clinical nutrition multidisciplinary team (MDT), Hospital das Clinicas, Sao Paulo, Brazil
- Research Unit, Central Pharmacy, Hospital das Clinicas, Sao Paulo, Brazil
| | - Salomao Faintuch
- Clinical Director, Vascular and Interventional Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Asher Mishaly
- Medical Student, Americas Faculty of Medicine, Sao Paulo, Brazil
| | - Joel Faintuch
- Department of Gastroenterology, Hospital das Clinicas and Sao Paulo University Medical School, Sao Paulo, Brazil
| |
Collapse
|
21
|
Herwig E, Schwean-Lardner K, Van Kessel A, Savary RK, Classen HL. Assessing the effect of starch digestion characteristics on ileal brake activation in broiler chickens. PLoS One 2020; 15:e0228647. [PMID: 32032378 PMCID: PMC7006927 DOI: 10.1371/journal.pone.0228647] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/20/2020] [Indexed: 01/10/2023] Open
Abstract
The objective of this research was to evaluate activation of the ileal brake in broiler chickens using diets containing semi-purified wheat (WS; rapidly and highly digested) and pea (PS; slowly and poorly digested) starch. Diets were formulated to contain six WS:PS ratios (100:0, 80:20, 60:40, 40:60, 20:80, 0:100) and each starch ratio was fed to 236 Ross 308 male broilers housed in 4 litter floor pens. At 28 d of age, the effect of PS concentration was assessed on starch digestion, digestive tract morphology, and digesta pH and short-chain fatty acid (SCFA) concentration. Glucagon-like peptide-1 (GLP-1) and peptide tyrosine-tyrosine (PYY) status were assessed in serum (ELISA) and via gene expression in jejunal and ileal tissue (proglucagon for GLP-1). Data were analyzed using regression analyses, and significance was accepted at P ≤ 0.05. Increasing dietary PS resulted in reduced starch digestibility in the small intestine, but had no effect in the colon. Crop content pH responded quadratically to PS level with an estimated minimum at 55% PS. Total SCFA increased linearly in the crop with PS level, but changed in a quadratic fashion in the ileum (estimated maximum at 62% PS). Ceacal SCFA concentrations were highest for the 80 and 100% PS levels. The relative empty weight (crop, small intestine, colon), length (small intestine) and content (crop jejunum, Ileum) of digestive tract sections increased linearly with increasing PS concentration. Dietary treatment did not affect serum GLP-1 or PYY or small intestine transcript abundance. In conclusion, feeding PS increased the presence of L-cell activators (starch, SCFA) and increased trophic development and content of the digestive tract, suggestive of L-cell activation. However, no direct evidence of ileal brake activation was found by measuring venous blood levels of GLP-1 or PYY or corresponding gene expression in small intestine tissue.
Collapse
Affiliation(s)
- Eugenia Herwig
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Karen Schwean-Lardner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Andrew Van Kessel
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Rachel K. Savary
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Henry L. Classen
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
22
|
van Erp RJJ, de Vries S, van Kempen TATG, Gerrits WJJ. Pigs Ferment Enzymatically Digestible Starch when it Is Substituted for Resistant Starch. J Nutr 2019; 149:1346-1353. [PMID: 31162602 PMCID: PMC6686059 DOI: 10.1093/jn/nxz072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/18/2019] [Accepted: 03/25/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Feeding behavior is controlled by satiety mechanisms, which are affected by the extent of starch digestion, and thus resistant starch (RS) intake. Alterations in feeding behavior to changes in RS intake may depend on the adaptation of processes involved when shifting from starch digestion to fermentation or vice versa. OBJECTIVES The aim of this study was to investigate how growing pigs adapt their feeding behavior in response to increasing and decreasing dietary RS concentrations. METHODS Thirty-six groups of 6 pigs (25.4 ± 2.8 kg; Hypor Libra × Hypor Maxter; male:female, 1:1) were fed diets containing 50% high-amylose maize starch (high RS; HRS) or waxy maize starch (low RS; LRS). Over 28 d, diets were exchanged following a 5-step titration (25% per step) that was executed in the upward (LH) or downward direction (HL). Twelve groups received a control diet to correct for changes over time. Individual feeding behavior and total tract starch digestion and fermentation were evaluated. The response in each parameter to increasing dietary HRS inclusion was estimated through the use of linear regression procedures, and tested for titration direction and sex effects. RESULTS Complete substitution of LRS with HRS increased the proportion of starch fermented, which was greater in LH pigs than in HL pigs (17.6% compared with 8.18%; P < 0.001), and decreased the feed intake (106 g/d; P = 0.021) and meal size (12.6 g; P < 0.001) of LH pigs, but not of HL pigs. In LH pigs, the size of the starch fermentation response positively correlated with the size of the feed intake response (r = 0.90, P < 0.001). CONCLUSIONS The attenuated response in starch fermentation in HL pigs indicates that pigs adapt more slowly to dietary supply of digestible starch than to RS, consequently resulting in fermentation of enzymatically digestible starch. Feed intake and feeding behavior only changed in pigs poorly adapting to RS, indicating that adequacy of adaptation, rather than RS itself, drives feed intake. These findings stress the importance of diet history for nutrient digestion and feeding behavior.
Collapse
Affiliation(s)
- Rik J J van Erp
- Trouw Nutrition, Amersfoort, The Netherlands
- Animal Nutrition Group, Wageningen University, Wageningen, The Netherlands
| | - Sonja de Vries
- Animal Nutrition Group, Wageningen University, Wageningen, The Netherlands
| | - Theo A T G van Kempen
- Trouw Nutrition, Amersfoort, The Netherlands
- North Carolina State University, Raleigh, NC
| | - Walter J J Gerrits
- Animal Nutrition Group, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
23
|
Holst JJ. The incretin system in healthy humans: The role of GIP and GLP-1. Metabolism 2019; 96:46-55. [PMID: 31029770 DOI: 10.1016/j.metabol.2019.04.014] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 01/07/2023]
Abstract
The incretin effect, the amplification of insulin secretion occurring when glucose is taken in orally as compared to infused intravenously, is one of the factors that help the body to tolerate carbohydrate/glucose ingestion. These include 1) amount and type of carbohydrates; 2) gastric emptying rate; 3) digestion and absorption of the carbohydrates; 4) secretion and effect of the incretin hormones; 5) disposition of absorbed nutrients/glucose. The incretin effect can also be viewed as the fraction of the ingested glucose load handled via gastrointestinal mechanisms (including the incretin effect); it is calculated by comparison of the amount of glucose required to copy, by intravenous infusion, the oral load. Typically, for 75 g of oral glucose, about 25 g are required. This means that the GastroIntestinal Glucose Disposal (GIGD) is 66%. Both the GIGD and the incretin effect depend on the amount of glucose ingested: for higher doses the GIGD may amount to 80%, which shows that this effect is a major contributor to glucose tolerance. The main mechanism behind it is stimulation of insulin secretion by a proportional secretion of the insulinotropic hormones GIP and GLP-1. Recently it has become possible to estimate their contributions in healthy humans using specific and potent receptor antagonists. Both hormones act to improve glucose tolerance (i.e. the antagonists impair tolerance) and their effects are additive. GIP seems to be quantitatively the most important, particularly regarding insulin secretion, whereas the action of GLP-1 is mainly displayed via inhibition of glucagon secretion.
Collapse
Affiliation(s)
- Jens Juul Holst
- The NNF center for Basic Metabolic Research and Department of Biomedical Sciences, the Panum Institute, University of Copenhagen, DK-2200, Denmark.
| |
Collapse
|
24
|
Wauters L, Vanuytsel T. Applications of peptide hormone ligands for the treatment of dumping and short bowel syndrome. Curr Opin Pharmacol 2018; 43:118-123. [DOI: 10.1016/j.coph.2018.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/06/2018] [Accepted: 09/18/2018] [Indexed: 12/28/2022]
|
25
|
Vincent AD, Wang XY, Parsons SP, Khan WI, Huizinga JD. Abnormal absorptive colonic motor activity in germ-free mice is rectified by butyrate, an effect possibly mediated by mucosal serotonin. Am J Physiol Gastrointest Liver Physiol 2018; 315:G896-G907. [PMID: 30095295 DOI: 10.1152/ajpgi.00237.2017] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The role of short-chain fatty acids (SCFAs) in the control of colonic motility is controversial. Germ-free (GF) mice are unable to produce these metabolites and serve as a model to study how their absence affects colonic motility. GF transit is slower than controls, and colonization of these mice improves transit and serotonin [5-hydroxytryptamine (5-HT)] levels. Our aim was to determine the role SCFAs play in improving transit and whether this is dependent on mucosal 5-HT signaling. Motility was assessed in GF mice via spatiotemporal mapping. First, motor patterns in the whole colon were measured ex vivo with or without luminal SCFA, and outflow from the colon was recorded to quantify outflow caused by individual propulsive contractions. Second, artificial fecal pellet propulsion was measured. Motility was then assessed in tryptophan hydroxylase-1 (TPH1) knockout (KO) mice, devoid of mucosal 5-HT, with phosphate buffer, butyrate, or propionate intraluminal perfusion. GF mice exhibited a lower proportion of propulsive contractions, lower volume of outflow/contraction, slower velocity of contractions, and slower propulsion of fecal pellets compared with controls. SCFAs changed motility patterns to that of controls in all parameters. Butyrate administration increased the proportion of propulsive contractions in controls yet failed to in TPH1 KO mice. Propionate inhibited propulsive contractions in all mice. Our results reveal significant abnormalities in the propulsive nature of colonic motor patterns in GF mice, explaining the decreased transit time in in vivo studies. We show that butyrate but not propionate activates propulsive motility and that this may require mucosal 5-HT. NEW & NOTEWORTHY Understanding the role that the microbiota play in governing the physiology of colonic motility is lacking. Here, we offer for the first time, to our knowledge, a detailed analysis of colonic motor patterns and pellet propulsion using spatiotemporal mapping in the absence of microbiota. We show a striking difference in germ-free and control phenotypes and attribute this to a lack of fermentation-produced short-chain fatty acid. We then show that butyrate but not propionate can restore motility and that the butyrate effect likely requires mucosal 5-hydroxytryptamine.
Collapse
Affiliation(s)
- Alexander D Vincent
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Xuan-Yu Wang
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Sean P Parsons
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Waliul I Khan
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| | - Jan D Huizinga
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University , Hamilton, Ontario , Canada
| |
Collapse
|
26
|
Mussa BM, Sood S, Verberne AJM. Implication of neurohormonal-coupled mechanisms of gastric emptying and pancreatic secretory function in diabetic gastroparesis. World J Gastroenterol 2018; 24:3821-3833. [PMID: 30228777 PMCID: PMC6141338 DOI: 10.3748/wjg.v24.i34.3821] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/22/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
Recently, diabetic gastroparesis (DGP) has received much attention as its prevalence is increasing in a dramatic fashion and management of patients with DGP represents a challenge in the clinical practice due to the limited therapeutic options. DGP highlights an interrelationship between the gastric emptying and pancreatic secretory function that regulate a wide range of digestive and metabolic functions, respectively. It well documented that both gastric emptying and pancreatic secretion are under delicate control by multiple neurohormonal mechanisms including extrinsic parasympathetic pathways and gastrointestinal (GI) hormones. Interestingly, the latter released in response to various determinants that related to the rate and quality of gastric emptying. Others and we have provided strong evidence that the central autonomic nuclei send a dual output (excitatory and inhibitory) to the stomach and the pancreas in response to a variety of hormonal signals from the abdominal viscera. Most of these hormones released upon gastric emptying to provide feedback, and control this process and simultaneously regulate pancreatic secretion and postprandial glycemia. These findings emphasize an important link between gastric emptying and pancreatic secretion and its role in maintaining homeostatic processes within the GI tract. The present review deals with the neurohormonal-coupled mechanisms of gastric emptying and pancreatic secretory function that implicated in DGP and this provides new insights in our understanding of the pathophysiology of DGP. This also enhances the process of identifying potential therapeutic targets to treat DGP and limit the complications of current management practices.
Collapse
Affiliation(s)
- Bashair M Mussa
- Department of Basic Medical Science, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sanjay Sood
- Department of Basic Medical Science, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Anthony JM Verberne
- Department of Medicine, Austin Health, University of Melbourne, Melbourne 3084, Australia
| |
Collapse
|
27
|
Nauck MA, Meier JJ. Incretin hormones: Their role in health and disease. Diabetes Obes Metab 2018; 20 Suppl 1:5-21. [PMID: 29364588 DOI: 10.1111/dom.13129] [Citation(s) in RCA: 513] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
Abstract
Incretin hormones are gut peptides that are secreted after nutrient intake and stimulate insulin secretion together with hyperglycaemia. GIP (glucose-dependent insulinotropic polypeptide) und GLP-1 (glucagon-like peptide-1) are the known incretin hormones from the upper (GIP, K cells) and lower (GLP-1, L cells) gut. Together, they are responsible for the incretin effect: a two- to three-fold higher insulin secretory response to oral as compared to intravenous glucose administration. In subjects with type 2 diabetes, this incretin effect is diminished or no longer present. This is the consequence of a substantially reduced effectiveness of GIP on the diabetic endocrine pancreas, and of the negligible physiological role of GLP-1 in mediating the incretin effect even in healthy subjects. However, the insulinotropic and glucagonostatic effects of GLP-1 are preserved in subjects with type 2 diabetes to the degree that pharmacological stimulation of GLP-1 receptors significantly reduces plasma glucose and improves glycaemic control. Thus, it has become a parent compound of incretin-based glucose-lowering medications (GLP-1 receptor agonists and inhibitors of dipeptidyl peptidase-4 or DPP-4). GLP-1, in addition, has multiple effects on various organ systems. Most relevant are a reduction in appetite and food intake, leading to weight loss in the long term. Since GLP-1 secretion from the gut seems to be impaired in obese subjects, this may even indicate a role in the pathophysiology of obesity. Along these lines, an increased secretion of GLP-1 induced by delivering nutrients to lower parts of the small intestines (rich in L cells) may be one factor (among others like peptide YY) explaining weight loss and improvements in glycaemic control after bariatric surgery (e.g., Roux-en-Y gastric bypass). GIP and GLP-1, originally characterized as incretin hormones, have additional effects in adipose cells, bone, and the cardiovascular system. Especially, the latter have received attention based on recent findings that GLP-1 receptor agonists such as liraglutide reduce cardiovascular events and prolong life in high-risk patients with type 2 diabetes. Thus, incretin hormones have an important role physiologically, namely they are involved in the pathophysiology of obesity and type 2 diabetes, and they have therapeutic potential that can be traced to well-characterized physiological effects.
Collapse
Affiliation(s)
- Michael A Nauck
- Diabetes Center Bochum-Hattingen, Medical Department I, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Juris J Meier
- Diabetes Center Bochum-Hattingen, Medical Department I, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| |
Collapse
|
28
|
Zhang G, Hamaker BR. The nutritional property of endosperm starch and its contribution to the health benefits of whole grain foods. Crit Rev Food Sci Nutr 2018; 57:3807-3817. [PMID: 26852626 DOI: 10.1080/10408398.2015.1130685] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Purported health benefits of whole grain foods in lowering risk of obesity, type 2 diabetes, cardiovascular disease, and cancer are supported by epidemiological studies and scientific researches. Bioactive components including dietary fibers, phytochemicals, and various micronutrients present in the bran and germ are commonly considered as the basis for such benefits. Endosperm starch, as the major constituent of whole grains providing glucose to the body, has been less investigated regarding its nutritional property and contribution to the value of whole grain foods. Nutritional quality of starch is associated with its rate of digestion and glucose absorption. In whole grain foods, starch digestion and glucose delivery may vary depending on the form in which the food is delivered, some with starch being rapidly and others slowly digested. Furthermore, there are other inherent factors in whole grain products, such as phenolic compounds and dietary fibers, that may moderate glycemic profiles. A good understanding of the nutritional properties of whole grain starch is important to the development of food processing technologies to maximize their health benefits.
Collapse
Affiliation(s)
- Genyi Zhang
- a State Key Laboratory of Food Science and Technology, School of Food Science and Technology , Jiangnan University , Wuxi , China
| | - Bruce R Hamaker
- b Whistler Center for Carbohydrate Research, Department of Food Science , Purdue University , West Lafayette , Indiana , USA
| |
Collapse
|
29
|
Hvistendahl M, Brandt CF, Tribler S, Naimi RM, Hartmann B, Holst JJ, Rehfeld JF, Hornum M, Andersen JR, Henriksen BM, Brøbech Mortensen P, Jeppesen PB. Effect of Liraglutide Treatment on Jejunostomy Output in Patients With Short Bowel Syndrome: An Open-Label Pilot Study. JPEN J Parenter Enteral Nutr 2017; 42:112-121. [PMID: 27875281 DOI: 10.1177/0148607116672265] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/08/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND An impaired hormonal "ileo-colonic brake" may contribute to rapid gastric emptying, gastric hypersecretion, high ostomy losses, and the need for parenteral support in end-jejunostomy short bowel syndrome (SBS) patients with intestinal failure (IF). Liraglutide, a glucagon-like peptide 1 receptor agonist, may reduce gastric hypersecretion and dampen gastric emptying, thereby improving conditions for intestinal absorption. MATERIALS AND METHODS In an 8-week, open-label pilot study, liraglutide was given subcutaneously once daily to 8 end-jejunostomy patients, aged 63.4 ± 10.9 years (mean ± SD) and with small bowel lengths of 110 ± 66 cm. The 72-hour metabolic balance studies were performed before and at the end of treatment. Food intake was unrestricted. Oral fluid intake and parenteral support volume were kept constant. The primary end point was change in the ostomy wet weight output. RESULTS Liraglutide reduced ostomy wet weight output by 474 ± 563 g/d from 3249 ± 1352 to 2775 ± 1187 g/d (P = .049, Student t test). Intestinal wet weight absorption tended to increase by 464 ± 557 g/d (P = .05), as did urine production by 765 ± 759 g/d (P = .02). Intestinal energy absorption improved by 902 ± 882 kJ/d (P = .02). CONCLUSION Liraglutide reduced ostomy wet weight output in end-jejunostomy patients with SBS-IF and increased their intestinal wet weight and energy absorption. If larger, randomized, placebo-controlled studies confirm these effects, it adds to the hypothesis that many ileo-colonic brake hormones in conjunction may be involved in the process of intestinal adaptation. By identification of key hormones and addressing their potential synergistic effects, better treatments may be provided to patients with SBS-IF. This trial was registered at clinicaltrialsregister.eu as 2013-005499-16.
Collapse
Affiliation(s)
- Mark Hvistendahl
- Department of Medical Gastroenterology CA-2121, Rigshospitalet, Copenhagen, Denmark
| | | | - Siri Tribler
- Department of Medical Gastroenterology CA-2121, Rigshospitalet, Copenhagen, Denmark
| | - Rahim Mohammad Naimi
- Department of Medical Gastroenterology CA-2121, Rigshospitalet, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, NNF Center of Basic Metabolic Research, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, NNF Center of Basic Metabolic Research, Copenhagen, Denmark
| | | | - Mads Hornum
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
| | - Jens Rikardt Andersen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
30
|
Badillo-Suárez PA, Rodríguez-Cruz M, Nieves-Morales X. Impact of Metabolic Hormones Secreted in Human Breast Milk on Nutritional Programming in Childhood Obesity. J Mammary Gland Biol Neoplasia 2017; 22:171-191. [PMID: 28653126 DOI: 10.1007/s10911-017-9382-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 06/02/2017] [Indexed: 12/20/2022] Open
Abstract
Obesity is the most common metabolic disease whose prevalence is increasing worldwide. This condition is considered a serious public health problem due to associated comorbidities such as diabetes mellitus and hypertension. Perinatal morbidity related to obesity does not end with birth; this continues affecting the mother/infant binomial and could negatively impact on metabolism during early infant nutrition. Nutrition in early stages of growth may be essential in the development of obesity in adulthood, supporting the concept of "nutritional programming". For this reason, breastfeeding may play an important role in this programming. Breast milk is the most recommended feeding for the newborn due to the provided benefits such as protection against obesity and diabetes. Health benefits are based on milk components such as bioactive molecules, specifically hormones involved in the regulation of food intake. Identification of these molecules has increased in recent years but its action has not been fully clarified. Hormones such as leptin, insulin, ghrelin, adiponectin, resistin, obestatin and insulin-like growth factor-1 copeptin, apelin, and nesfatin, among others, have been identified in the milk of normal-weight women and may influence the energy balance because they can activate orexigenic or anorexigenic pathways depending on energy requirements and body stores. It is important to emphasize that, although the number of biomolecules identified in milk involved in regulating food intake has increased considerably, there is a lack of studies aimed at elucidating the effect these hormones may have on metabolism and development of the newborn. Therefore, we present a state-of-the-art review regarding bioactive compounds such as hormones secreted in breast milk and their possible impact on nutritional programming in the infant, analyzing their functions in appetite regulation.
Collapse
Affiliation(s)
- Pilar Amellali Badillo-Suárez
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Av. Cuauhtémoc No. 330, Col. Doctores, Deleg. Cuauhtémoc, 06725, México, DF, México
| | - Maricela Rodríguez-Cruz
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Av. Cuauhtémoc No. 330, Col. Doctores, Deleg. Cuauhtémoc, 06725, México, DF, México.
| | - Xóchitl Nieves-Morales
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Av. Cuauhtémoc No. 330, Col. Doctores, Deleg. Cuauhtémoc, 06725, México, DF, México
| |
Collapse
|
31
|
Ji WW, Yu DA, Fan M, You M, Lu Y, Li EB, Xie N, Yan SS. Effects of GW002, a novel recombinant human glucagon-like peptide-1 (GLP-1) analog fusion protein, on CHO recombinant cells and BKS-db mice. Acta Diabetol 2017; 54:685-693. [PMID: 28424924 DOI: 10.1007/s00592-017-0992-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/03/2017] [Indexed: 12/17/2022]
Abstract
AIMS GLP-1-based strategies have many advantages in treatment of type 2 diabetes mellitus (T2DM), but native GLP-1 has a short half-life in the circulation, which limits its clinical application. The purpose of this study was to evaluate the effects of GW002, a novel recombinant GLP-1 analog fusion protein produced by linking the human GLP-1 analog C-terminus to the N-terminus of human serum albumin via a linker, in vitro and in BKS-db mice. METHODS To determine whether GW002 can activate the GLP-1 receptor in cells, the level of luciferase expression was evaluated in vitro. In vivo, body weight, food intake, non-fasting and fasting blood glucose, oral glucose tolerance test, blood glucose and insulin levels, liver histology, liver function parameters and antibody levels in BKS-db mice were investigated to evaluate the effects of GW002. Albiglutide was chosen as a positive comparator. RESULTS Cyclic adenosine monophosphate levels were increased in a dose-dependent manner in cells. In vivo studies demonstrated that GW002 lowers non-fasting and fasting blood glucose levels and improves glucose tolerance and insulin secretion in BKS-db mice. The degree of hepatic steatosis and hepatic biochemical indexes was also decreased. In this study, the mice body weight was not reduced significantly. CONCLUSIONS The above results showed that the efficacy of GW002 in BKS-db mice displayed a significant hypoglycemic effect, which indicated that GW002 might be a potential candidate for the treatment of T2DM.
Collapse
Affiliation(s)
- Wan-Wan Ji
- Jiangsu T-mab BioPharma Co., Ltd, G03, 1# Yao-Cheng Avenue, Taizhou, Jiangsu, People's Republic of China
| | - Dong-An Yu
- Jiangsu T-mab BioPharma Co., Ltd, G03, 1# Yao-Cheng Avenue, Taizhou, Jiangsu, People's Republic of China
| | - Min Fan
- Jiangsu T-mab BioPharma Co., Ltd, G03, 1# Yao-Cheng Avenue, Taizhou, Jiangsu, People's Republic of China
| | - Meng You
- Jiangsu T-mab BioPharma Co., Ltd, G03, 1# Yao-Cheng Avenue, Taizhou, Jiangsu, People's Republic of China
| | - You Lu
- Jiangsu T-mab BioPharma Co., Ltd, G03, 1# Yao-Cheng Avenue, Taizhou, Jiangsu, People's Republic of China
| | - Er-Bing Li
- Nanjing Biomedical Research Institute of Nanjing University, 12#, Xue-Fu Road, Nanjing, Jiangsu, People's Republic of China
| | - Ning Xie
- Jiangsu T-mab BioPharma Co., Ltd, G03, 1# Yao-Cheng Avenue, Taizhou, Jiangsu, People's Republic of China
| | - Shou-Sheng Yan
- Jiangsu T-mab BioPharma Co., Ltd, G03, 1# Yao-Cheng Avenue, Taizhou, Jiangsu, People's Republic of China.
| |
Collapse
|
32
|
Lee AA, Owyang C. Sugars, Sweet Taste Receptors, and Brain Responses. Nutrients 2017; 9:nu9070653. [PMID: 28672790 PMCID: PMC5537773 DOI: 10.3390/nu9070653] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/25/2022] Open
Abstract
Sweet taste receptors are composed of a heterodimer of taste 1 receptor member 2 (T1R2) and taste 1 receptor member 3 (T1R3). Accumulating evidence shows that sweet taste receptors are ubiquitous throughout the body, including in the gastrointestinal tract as well as the hypothalamus. These sweet taste receptors are heavily involved in nutrient sensing, monitoring changes in energy stores, and triggering metabolic and behavioral responses to maintain energy balance. Not surprisingly, these pathways are heavily regulated by external and internal factors. Dysfunction in one or more of these pathways may be important in the pathogenesis of common diseases, such as obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Allen A Lee
- 1500 East Medical Center Drive, Division of Gastroenterology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109-5362, USA.
| | - Chung Owyang
- 3912 Taubman Center, SPC 5362, Ann Arbor, MI 48109-5362, USA.
| |
Collapse
|
33
|
Nakatani Y, Maeda M, Matsumura M, Shimizu R, Banba N, Aso Y, Yasu T, Harasawa H. Effect of GLP-1 receptor agonist on gastrointestinal tract motility and residue rates as evaluated by capsule endoscopy. DIABETES & METABOLISM 2017. [PMID: 28648835 DOI: 10.1016/j.diabet.2017.05.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
AIM This study evaluated the effects of a glucagon-like peptide-1 receptor agonist on gastrointestinal (GI) tract motility and residue rates by examining GI transit time and lumen using capsule endoscopy. MATERIAL AND METHODS GI motility and lumen were assessed by capsule endoscopy before and after liraglutide administration in 14 patients with type 2 diabetes mellitus (T2DM). RESULTS Gastric transit time in the group with diabetic neuropathy (DN) was 1:12:36±1:04:30h before liraglutide administration and 0:48:40±0:32:52h after administration (nonsignificant difference, P=0.19). Gastric transit time in the non-DN group was 1:01:30±0:52:59h before administration and 2:33:29±1:37:24h after administration (significant increase, P=0.03). Duodenal and small intestine transit time in the DN group was 4:10:34±0:25:54h before and 6:38:42±3:52:42h after administration (not significant, P=0.09) and, in the non-DN group, 3:51:03±0:53:47h before and 6:45:31±2:41:36h after administration (significant increase, P=0.03). The GI residue rate in the DN group was 32.1±24% before administration and 90.0±9.1% after administration (significant increase, P<0.001), and increased in all patients; in the non-DN group, it was 32.1±35.3% before and 78.3±23.9% after administration (significant increase, P<0.001), and also increased in all patients. CONCLUSION Liraglutide causes delayed gastric emptying and inhibits duodenal and small intestine motility. However, these GI movement-inhibiting effects may be decreased or absent in patients with DN-associated dysautonomia.
Collapse
Affiliation(s)
- Y Nakatani
- Department of Diabetes and Endocrinology, Dokkyo Medical University Nikko Medical Center, 632, Takatoku Nikkoshi, 321-2593 Tochigi, Japan.
| | - M Maeda
- Department of Gastroenterology, Dokkyo Medical University Nikko Medical Center, 632, Takatoku Nikkoshi, 321-2593 Tochigi, Japan
| | - M Matsumura
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880, Kitakobayashi Shimotsugagun Mibumachi, 321-0293 Tochigi, Japan
| | - R Shimizu
- Department of Cardiovascular Medicine, Dokkyo Medical University Nikko Medical Center, 632, Takatoku Nikkoshi, 321-2593 Tochigi, Japan
| | - N Banba
- Department of Diabetes and Endocrinology, Dokkyo Medical University Nikko Medical Center, 632, Takatoku Nikkoshi, 321-2593 Tochigi, Japan
| | - Y Aso
- Department of Endocrinology and Metabolism, Dokkyo Medical University, 880, Kitakobayashi Shimotsugagun Mibumachi, 321-0293 Tochigi, Japan
| | - T Yasu
- Department of Cardiovascular Medicine, Dokkyo Medical University Nikko Medical Center, 632, Takatoku Nikkoshi, 321-2593 Tochigi, Japan
| | - H Harasawa
- Department of Pulmonary Medicine, Dokkyo Medical University Nikko Medical Center, 632, Takatoku Nikkoshi, 321-2593 Tochigi, Japan
| |
Collapse
|
34
|
|
35
|
Smits MM, Tonneijck L, Muskiet MHA, Kramer MHH, Cahen DL, van Raalte DH. Gastrointestinal actions of glucagon-like peptide-1-based therapies: glycaemic control beyond the pancreas. Diabetes Obes Metab 2016; 18:224-35. [PMID: 26500045 DOI: 10.1111/dom.12593] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/22/2015] [Accepted: 10/17/2015] [Indexed: 12/23/2022]
Abstract
The gastrointestinal hormone glucagon-like peptide-1 (GLP-1) lowers postprandial glucose concentrations by regulating pancreatic islet-cell function, with stimulation of glucose-dependent insulin and suppression of glucagon secretion. In addition to endocrine pancreatic effects, mounting evidence suggests that several gastrointestinal actions of GLP-1 are at least as important for glucose-lowering. GLP-1 reduces gastric emptying rate and small bowel motility, thereby delaying glucose absorption and decreasing postprandial glucose excursions. Furthermore, it has been suggested that GLP-1 directly stimulates hepatic glucose uptake, and suppresses hepatic glucose production, thereby adding to reduction of fasting and postprandial glucose levels. GLP-1 receptor agonists, which mimic the effects of GLP-1, have been developed for the treatment of type 2 diabetes. Based on their pharmacokinetic profile, GLP-1 receptor agonists can be broadly categorized as short- or long-acting, with each having unique islet-cell and gastrointestinal effects that lower glucose levels. Short-acting agonists predominantly lower postprandial glucose excursions, by inhibiting gastric emptying and intestinal glucose uptake, with little effect on insulin secretion. By contrast, long-acting agonists mainly reduce fasting glucose levels, predominantly by increased insulin and reduced glucagon secretion, with potential additional direct inhibitory effects on hepatic glucose production. Understanding these pharmacokinetic and pharmacodynamic differences may allow personalized antihyperglycaemic therapy in type 2 diabetes. In addition, it may provide the rationale to explore treatment in patients with no or little residual β-cell function.
Collapse
Affiliation(s)
- M M Smits
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - L Tonneijck
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - M H A Muskiet
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - M H H Kramer
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - D L Cahen
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - D H van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Latorre R, Huynh J, Mazzoni M, Gupta A, Bonora E, Clavenzani P, Chang L, Mayer EA, De Giorgio R, Sternini C. Expression of the Bitter Taste Receptor, T2R38, in Enteroendocrine Cells of the Colonic Mucosa of Overweight/Obese vs. Lean Subjects. PLoS One 2016; 11:e0147468. [PMID: 26866366 PMCID: PMC4750998 DOI: 10.1371/journal.pone.0147468] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/03/2016] [Indexed: 12/12/2022] Open
Abstract
Bitter taste receptors (T2Rs) are expressed in the mammalian gastrointestinal mucosa. In the mouse colon, T2R138 is localized to enteroendocrine cells and is upregulated by long-term high fat diet that induces obesity. The aims of this study were to test whether T2R38 expression is altered in overweight/obese (OW/OB) compared to normal weight (NW) subjects and characterize the cell types expressing T2R38, the human counterpart of mouse T2R138, in human colon. Colonic mucosal biopsies were obtained during colonoscopy from 35 healthy subjects (20 OW/OB and 15 NW) and processed for quantitative RT-PCR and immunohistochemistry using antibodies to T2R38, chromogranin A (CgA), glucagon like peptide-1 (GLP-1), cholecystokinin (CCK), or peptide YY (PYY). T2R38 mRNA levels in the colonic mucosa of OW/OB were increased (> 2 fold) compared to NW subjects but did not reach statistical significance (P = 0.06). However, the number of T2R38 immunoreactive (IR) cells was significantly increased in OW/OB vs. NW subjects (P = 0.01) and was significantly correlated with BMI values (r = 0.7557; P = 0.001). In both OW/OB and NW individuals, all T2R38-IR cells contained CgA-IR supporting they are enteroendocrine. In both groups, T2R38-IR colocalized with CCK-, GLP1- or PYY-IR. The overall CgA-IR cell population was comparable in OW/OB and NW individuals. This study shows that T2R38 is expressed in distinct populations of enteroendocrine cells in the human colonic mucosa and supports T2R38 upregulation in OW/OB subjects. T2R38 might mediate host functional responses to increased energy balance and intraluminal changes occurring in obesity, which could involve peptide release from enteroendocrine cells.
Collapse
Affiliation(s)
- Rocco Latorre
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jennifer Huynh
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Maurizio Mazzoni
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy
| | - Arpana Gupta
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Elena Bonora
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Clavenzani
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy
| | - Lin Chang
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Emeran A. Mayer
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Oppenheimer Family Center for Neurobiology of Stress, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Roberto De Giorgio
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Catia Sternini
- CURE/DDRC, Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
37
|
Lin AHM, Lee BH, Chang WJ. Small intestine mucosal α-glucosidase: A missing feature of in vitro starch digestibility. Food Hydrocoll 2016. [DOI: 10.1016/j.foodhyd.2015.03.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
38
|
Dumping Syndrome: A Review of the Current Concepts of Pathophysiology, Diagnosis, and Treatment. Dig Dis Sci 2016; 61:11-8. [PMID: 26396002 DOI: 10.1007/s10620-015-3839-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/30/2015] [Indexed: 12/15/2022]
Abstract
Gastric surgery has long been known to be a cause of dumping syndrome (DS). However, the increasing incidence of gastric bypass surgery, as well as reports of DS unrelated to previous gastric surgeries, has increased the importance of understanding DS in recent years. DS is due to the gastrointestinal response to voluminous and hyperosmolar chyme that is rapidly expelled from the stomach into the small intestine. This response involves neural and hormonal mechanisms. This review encompasses the symptoms, diagnosis, and treatment approaches of DS and also focuses on the current research status of the pathophysiology of DS.
Collapse
|
39
|
Gibb RD, McRorie JW, Russell DA, Hasselblad V, D'Alessio DA. Psyllium fiber improves glycemic control proportional to loss of glycemic control: a meta-analysis of data in euglycemic subjects, patients at risk of type 2 diabetes mellitus, and patients being treated for type 2 diabetes mellitus. Am J Clin Nutr 2015; 102:1604-14. [PMID: 26561625 DOI: 10.3945/ajcn.115.106989] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND A number of health benefits are associated with intake of soluble, viscous, gel-forming fibers, including reduced serum cholesterol and the attenuation of postprandial glucose excursions. OBJECTIVE We assess the effects of psyllium, which is a soluble, gel-forming, nonfermented fiber supplement, on glycemic control in patients who were being treated for type 2 diabetes mellitus (T2DM) and in patients who were at risk of developing T2DM. DESIGN A comprehensive search was performed of available published literature (Scopus scientific database) and clinical records stored by Procter & Gamble with the use of key search terms to identify clinical studies that assessed the glycemic effects of psyllium in nondiabetic, pre-T2DM, and T2DM patients. RESULTS We identified 35 randomized, controlled, clinical studies that spanned 3 decades and 3 continents. These data were assessed in 8 meta-analyses. In patients with T2DM, multiweek studies (psyllium dosed before meals) showed significant improvement in both the fasting blood glucose (FBG) concentration (-37.0 mg/dL; P < 0.001) and glycated hemoglobin (HbA1c) [-0.97% (-10.6 mmol/mol); P = 0.048]. Glycemic effects were proportional to baseline FBG; no significant glucose lowering was observed in euglycemic subjects, a modest improvement was observed in subjects with pre-T2DM, and the greatest improvement was observed in subjects who were being treated for T2DM. CONCLUSIONS These data indicate that psyllium would be an effective addition to a lifestyle-intervention program. The degree of psyllium's glycemic benefit was commensurate with the loss of glycemic control. Because the greatest effect was seen in patients who were being treated for T2DM, additional studies are needed to determine how best to incorporate psyllium into existing prevention and treatment algorithms with concomitant hypoglycemic medications.
Collapse
Affiliation(s)
| | | | | | | | - David A D'Alessio
- Division of Endocrinology, Metabolism and Nutrition, Duke Department of Medicine, Duke University, Durham, NC
| |
Collapse
|
40
|
Manning S, Pucci A, Batterham RL. GLP-1: a mediator of the beneficial metabolic effects of bariatric surgery? Physiology (Bethesda) 2015; 30:50-62. [PMID: 25559155 DOI: 10.1152/physiol.00027.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
There has been increasing interest in the role that gut hormones may play in contributing to the physiological changes produced by certain bariatric procedures, such as Roux-en-Y gastric bypass and sleeve gastrectomy. Here, we review the evidence implicating one such gut hormone, glucagon-like peptide-1, as a mediator of the metabolic benefits of these two procedures.
Collapse
Affiliation(s)
- Sean Manning
- Department of Medicine, Centre for Obesity Research, Rayne Institute, University College London, London, United Kingdom; UCLH Centre for Weight Loss, Metabolic and Endocrine Surgery, University College London Hospitals, London, United Kingdom; National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Andrea Pucci
- Department of Medicine, Centre for Obesity Research, Rayne Institute, University College London, London, United Kingdom; UCLH Centre for Weight Loss, Metabolic and Endocrine Surgery, University College London Hospitals, London, United Kingdom
| | - Rachel L Batterham
- Department of Medicine, Centre for Obesity Research, Rayne Institute, University College London, London, United Kingdom; UCLH Centre for Weight Loss, Metabolic and Endocrine Surgery, University College London Hospitals, London, United Kingdom; National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
41
|
Mans E, Serra-Prat M, Palomera E, Suñol X, Clavé P. Sleeve gastrectomy effects on hunger, satiation, and gastrointestinal hormone and motility responses after a liquid meal test. Am J Clin Nutr 2015; 102:540-7. [PMID: 26201818 DOI: 10.3945/ajcn.114.104307] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 07/01/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The relation between hunger, satiation, and integrated gastrointestinal motility and hormonal responses in morbidly obese patients after sleeve gastrectomy has not been determined. OBJECTIVE The objective was to assess the effects of sleeve gastrectomy on hunger, satiation, gastric and gallbladder motility, and gastrointestinal hormone response after a liquid meal test. DESIGN Three groups were studied: morbidly obese patients (n = 16), morbidly obese patients who had had sleeve gastrectomy (n = 8), and nonobese patients (n = 16). The participants fasted for 10 h and then consumed a 200-mL liquid meal (400 kcal + 1.5 g paracetamol). Fasting and postprandial hunger, satiation, hormone concentrations, and gastric and gallbladder emptying were measured several times over 4 h. RESULTS No differences were observed in hunger and satiation curves between morbidly obese and nonobese groups; however, sleeve gastrectomy patients were less hungry and more satiated than the other groups. Antrum area during fasting in morbidly obese patients was statistically significant larger than in the nonobese and sleeve gastrectomy groups. Gastric emptying was accelerated in the sleeve gastrectomy group compared with the other 2 groups (which had very similar results). Gallbladder emptying was similar in the 3 groups. Sleeve gastrectomy patients showed the lowest ghrelin concentrations and higher early postprandial cholecystokinin and glucagon-like peptide 1 peaks than did the other participants. This group also showed an improved insulin resistance pattern compared with morbidly obese patients. CONCLUSIONS Sleeve gastrectomy seems to be associated with profound changes in gastrointestinal physiology that contribute to reducing hunger and increasing sensations of satiation. These changes include accelerated gastric emptying, enhanced postprandial cholecystokinin and glucagon-like peptide 1 concentrations, and reduced ghrelin release, which together may help patients lose weight and improve their glucose metabolism after surgery. This trial was registered at clinicaltrials.gov as NCT02414893.
Collapse
Affiliation(s)
| | - Mateu Serra-Prat
- Research Unit, and Center for Biomedical Research Network of Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain; and
| | | | | | - Pere Clavé
- Gastrointestinal Physiology Laboratory, Maresme Health Consortium, Barcelona, Spain; Center for Biomedical Research Network of Hepatic and Digestive Diseases (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain; and Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Overduin J, Guérin-Deremaux L, Wils D, Lambers TT. NUTRALYS(®) pea protein: characterization of in vitro gastric digestion and in vivo gastrointestinal peptide responses relevant to satiety. Food Nutr Res 2015; 59:25622. [PMID: 25882536 PMCID: PMC4400298 DOI: 10.3402/fnr.v59.25622] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 02/05/2015] [Accepted: 02/26/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Pea protein (from Pisum sativum) is under consideration as a sustainable, satiety-inducing food ingredient. OBJECTIVE In the current study, pea-protein-induced physiological signals relevant to satiety were characterized in vitro via gastric digestion kinetics and in vivo by monitoring post-meal gastrointestinal hormonal responses in rats. DESIGN Under in vitro simulated gastric conditions, the digestion of NUTRALYS(®) pea protein was compared to that of two dairy proteins, slow-digestible casein and fast-digestible whey. In vivo, blood glucose and gastrointestinal hormonal (insulin, ghrelin, cholecystokinin [CCK], glucagon-like peptide 1 [GLP-1], and peptide YY [PYY]) responses were monitored in nine male Wistar rats following isocaloric (11 kcal) meals containing 35 energy% of either NUTRALYS(®) pea protein, whey protein, or carbohydrate (non-protein). RESULTS In vitro, pea protein transiently aggregated into particles, whereas casein formed a more enduring protein network and whey protein remained dissolved. Pea-protein particle size ranged from 50 to 500 µm, well below the 2 mm threshold for gastric retention in humans. In vivo, pea-protein and whey-protein meals induced comparable responses for CCK, GLP-1, and PYY, that is, the anorexigenic hormones. Pea protein induced weaker initial, but equal 3-h integrated ghrelin and insulin responses than whey protein, possibly due to the slower gastric breakdown of pea protein observed in vitro. Two hours after meals, CCK levels were more elevated in the case of protein meals compared to that of non-protein meals. CONCLUSIONS These results indicate that 1) pea protein transiently aggregates in the stomach and has an intermediately fast intestinal bioavailability in between that of whey and casein; 2) pea-protein- and dairy-protein-containing meals were comparably efficacious in triggering gastrointestinal satiety signals.
Collapse
Affiliation(s)
- Joost Overduin
- Department of Health, NIZO Food Research, Ede, The Netherlands;
| | | | - Daniel Wils
- Nutrition Management, Roquette Frères, Lestrem, France
| | - Tim T Lambers
- Department of Health, NIZO Food Research, Ede, The Netherlands
| |
Collapse
|
43
|
Evidence-Based Approach to Fiber Supplements and Clinically Meaningful Health Benefits, Part 1: What to Look for and How to Recommend an Effective Fiber Therapy. ACTA ACUST UNITED AC 2015; 50:82-89. [PMID: 25972618 PMCID: PMC4415962 DOI: 10.1097/nt.0000000000000082] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dietary fiber that is intrinsic and intact in fiber-rich foods (eg, fruits, vegetables, legumes, whole grains) is widely recognized to have beneficial effects on health when consumed at recommended levels (25 g/d for adult women, 38 g/d for adult men). Most (90%) of the US population does not consume this level of dietary fiber, averaging only 15 g/d. In an attempt to bridge this “fiber gap,” many consumers are turning to fiber supplements, which are typically isolated from a single source. Fiber supplements cannot be presumed to provide the health benefits that are associated with dietary fiber from whole foods. Of the fiber supplements on the market today, only a minority possess the physical characteristics that underlie the mechanisms driving clinically meaningful health benefits. The first part (current issue) of this 2-part series will focus on the 4 main characteristics of fiber supplements that drive clinical efficacy (solubility, degree/rate of fermentation, viscosity, and gel formation), the 4 clinically meaningful designations that identify which health benefits are associated with specific fibers, and the gel-dependent mechanisms in the small bowel that drive specific health benefits (eg, cholesterol lowering, improved glycemic control). The second part (next issue) of this 2-part series will focus on the effects of fiber supplements in the large bowel, including the 2 mechanisms by which fiber prevents/relieves constipation (insoluble mechanical irritant and soluble gel-dependent water-holding capacity), the gel-dependent mechanism for attenuating diarrhea and normalizing stool form in irritable bowel syndrome, and the combined large bowel/small bowel fiber effects for weight loss/maintenance. The second part will also discuss how processing for marketed products can attenuate efficacy, why fiber supplements can cause gastrointestinal symptoms, and how to avoid symptoms for better long-term compliance.
Collapse
|
44
|
Belobrajdic DP, Jobling SA, Morell MK, Taketa S, Bird AR. Wholegrain barley β-glucan fermentation does not improve glucose tolerance in rats fed a high-fat diet. Nutr Res 2014; 35:162-8. [PMID: 25622537 DOI: 10.1016/j.nutres.2014.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/03/2014] [Accepted: 12/28/2014] [Indexed: 11/25/2022]
Abstract
Fermentation of oat and barley β-glucans is believed to mediate in part their metabolic health benefits, but the exact mechanisms remain unclear. In this study, we sought to test the hypothesis that barley β-glucan fermentation raises circulating incretin hormone levels and improves glucose control, independent of other grain components. Male Sprague-Dawley rats (n = 30) were fed a high-fat diet for 6 weeks and then randomly allocated to 1 of 3 dietary treatments for 2 weeks. The low- (LBG, 0% β-glucan) and high- (HBG, 3% β-glucan) β-glucan diets contained 25% wholegrain barley and similar levels of insoluble dietary fiber, available carbohydrate, and energy. A low-fiber diet (basal) was included for comparison. Immediately prior to the dietary intervention, gastric emptying rate (using the (13)C-octanoic breath test) and postprandial glycemic response of each diet were determined. At the end of the study, circulating gut hormone levels were determined; and a glucose tolerance test was performed. The rats were then killed, and indices of cecal fermentation were assessed. Diet did not affect live weight; however, the HBG diet, compared to basal and LBG, reduced food intake, tended to slow gastric emptying, increased cecal digesta mass and individual and total short-chain fatty acid pools, and lowered digesta pH. In contrast, circulating levels of glucose, insulin, gastric-inhibitory peptide, and glucagon-like peptide-1, and glucose tolerance were unaffected by diet. In conclusion, wholegrain barley β-glucan suppressed feed intake and increased cecal fermentation but did not improve postprandial glucose control or insulin sensitivity.
Collapse
Affiliation(s)
- Damien P Belobrajdic
- Commonwealth Scientific & Industrial Research Organisation (CSIRO) Food and Nutrition Flagship, Australia.
| | | | - Matthew K Morell
- Commonwealth Scientific & Industrial Research Organisation (CSIRO) Food and Nutrition Flagship, Australia.
| | - Shin Taketa
- Institute of Plant Science and Resources, Okayama University, Kurashiki 710-0046, Japan.
| | - Anthony R Bird
- Commonwealth Scientific & Industrial Research Organisation (CSIRO) Food and Nutrition Flagship, Australia.
| |
Collapse
|
45
|
Muscogiuri G, Cignarelli A, Giorgino F, Prodam F, Santi D, Tirabassi G, Balercia G, Modica R, Faggiano A, Colao A. GLP-1: benefits beyond pancreas. J Endocrinol Invest 2014; 37:1143-53. [PMID: 25107343 DOI: 10.1007/s40618-014-0137-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/10/2014] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Glucagon-like peptide 1 (GLP-1) is an intestinal hormone secreted after the ingestion of various nutrients. The main role of GLP-1 is to stimulate insulin secretion in a glucose-dependent manner. However, the expression of GLP-1 receptor was found to be expressed in a variety of tissues beyond pancreas such as lung, stomach, intestine, kidney, heart and brain. Beyond pancreas, a beneficial effect of GLP-1 on body weight reduction has been shown, suggesting its role for the treatment of obesity. In addition, GLP-1 has been demonstrated to reduce cardiovascular risk factors and to have a direct cardioprotective effect, fostering heart recovery after ischemic injury. Further, data from both experimental animal models and human studies have shown beneficial effect of GLP-1 on bone metabolism, either directly or indirectly on bone cells. MATERIALS AND METHODS We review here the recent findings of the extra-pancreatic effects of GLP-1 focusing on both basic and clinical studies, thus opening future perspectives to the use of GLP-1 analogs for the treatment of disease beyond type 2 diabetes. CONCLUSION Finally, the GLP-1 has been demonstrated to have a beneficial effect on both vascular, degenerative diseases of central nervous system and psoriasis.
Collapse
Affiliation(s)
- G Muscogiuri
- Section of Endocrinology, Department of Clinical Medicine and Surgery, University "Federico II", Via Sergio Pansini, 5, Naples, Italy,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Evaluation of intragastric vs intraperitoneal glucose tolerance tests in the evaluation of insulin resistance in a rodent model of burn injury and glucagon-like polypeptide-1 treatment. J Burn Care Res 2014; 35:e66-72. [PMID: 23511296 DOI: 10.1097/bcr.0b013e31828a8ede] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Evaluation of glucose tolerance in rodent models is usually performed after intraperitroneal administration of glucose (intraperitoneal glucose tolerance test [IPGTT]), whereas in humans the test is performed with oral glucose. Hyperglycemia is a major clinical manifestation of burn injury. Our previous studies using IPGTT have demonstrated burn injury-induced insulin resistance and the beneficial effects of glucagon-like polypeptide-1 (GLP-1) in improving insulin resistance. The goal of the present study is to compare the results of these two procedures under 1) burn injury-induced insulin resistance and 2) GLP-1 treatment after burn. Male CD rats were divided into three groups: sham burn, burn, and burn with GLP-1. Blood glucose and plasma insulin levels were measured during intragastric glucose tolerance test (IGGTT) on day 6 after 40% of full-thickness burn injury. The results were compared with our previous IPGTT. Blood glucose curves for IGGTT and IPGTT showed a similar pattern. However, IGGTT demonstrated a significant lower level of maximal blood glucose when compared with IPGTT. This was accompanied by higher peak insulin levels in sham burn and burn groups. In contrast, peak insulin levels of each burn with GLP-1 group were similar. 1) Both IPGTT and IGGTT demonstrated burn injury-induced insulin resistance and the efficacy of GLP-1 for reducing hyperglycemia after burn injury. 2) The observed differences in the plasma glucose and insulin levels between IGGTT and IPGTT suggest that endogenously produced GLP-1 during the IGGTT may play a role in ameliorating insulin resistance after burn injury.
Collapse
|
47
|
Wu T, Ma J, Bound MJ, Checklin H, Deacon CF, Jones KL, Horowitz M, Rayner CK. Effects of sitagliptin on glycemia, incretin hormones, and antropyloroduodenal motility in response to intraduodenal glucose infusion in healthy lean and obese humans and patients with type 2 diabetes treated with or without metformin. Diabetes 2014; 63:2776-2787. [PMID: 24647737 DOI: 10.2337/db13-1627] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The impact of variations in gastric emptying, which influence the magnitude of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) secretion, on glucose lowering by dipeptidyl peptidase-4 (DPP-4) inhibitors is unclear. We evaluated responses to intraduodenal glucose infusion (60 g over 120 min [i.e., 2 kcal/min], a rate that predominantly stimulates GIP but not GLP-1) after sitagliptin versus control in 12 healthy lean, 12 obese, and 12 type 2 diabetic subjects taking metformin 850 mg b.i.d. versus placebo. As expected, sitagliptin augmented plasma-intact GIP substantially and intact GLP-1 modestly. Sitagliptin attenuated glycemic excursions in healthy lean and obese but not type 2 diabetic subjects, without affecting glucagon or energy intake. In contrast, metformin reduced fasting and glucose-stimulated glycemia, suppressed energy intake, and augmented total and intact GLP-1, total GIP, and glucagon in type 2 diabetic subjects, with no additional glucose lowering when combined with sitagliptin. These observations indicate that in type 2 diabetes, 1) the capacity of endogenous GIP to lower blood glucose is impaired; 2) the effect of DPP-4 inhibition on glycemia is likely to depend on adequate endogenous GLP-1 release, requiring gastric emptying >2 kcal/min; and 3) the action of metformin to lower blood glucose is not predominantly by way of the incretin axis.
Collapse
Affiliation(s)
- Tongzhi Wu
- Discipline of Medicine, University of Adelaide, Adelaide, SA, AustraliaCentre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Jing Ma
- Discipline of Medicine, University of Adelaide, Adelaide, SA, AustraliaCentre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Michelle J Bound
- Discipline of Medicine, University of Adelaide, Adelaide, SA, AustraliaCentre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Helen Checklin
- Discipline of Medicine, University of Adelaide, Adelaide, SA, AustraliaCentre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Carolyn F Deacon
- Department of Biomedical Science, University of Copenhagen, Copenhagen, Denmark
| | - Karen L Jones
- Discipline of Medicine, University of Adelaide, Adelaide, SA, AustraliaCentre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Michael Horowitz
- Discipline of Medicine, University of Adelaide, Adelaide, SA, AustraliaCentre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Christopher K Rayner
- Discipline of Medicine, University of Adelaide, Adelaide, SA, AustraliaCentre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
48
|
Tokuda M, Katsuno T, Ochi F, Miyakoshi K, Kusunoki Y, Murai K, Miuchi M, Hamaguchi T, Miyagawa JI, Namba M. Effects of exenatide on metabolic parameters/control in obese Japanese patients with type 2 diabetes. Endocr J 2014; 61:365-72. [PMID: 24452017 DOI: 10.1507/endocrj.ej14-0009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The effects of exenatide on glycemic control, lipid metabolism, blood pressure, and gastrointestinal symptoms were investigated in obese Japanese patients with type 2 diabetes mellitus. Twenty-six outpatients were enrolled and administered 5 μg of exenatide twice daily. If there was insufficient weight loss and/or insufficient improvement in glycemic control, the dose was increased to 10 μg twice daily. Follow-up was continued until the 12th week of administration. Hemoglobin A1c, glycoalbumin, fasting plasma glucose, body weight, fasting serum C-peptide, serum lipids, blood pressure, and pulse rate were measured before and after the observation period. In the initial phase of exenatide therapy, each patient received a diary to record gastrointestinal symptoms. During treatment with exenatide, hemoglobin A1c decreased significantly and serum C-peptide increased significantly. Body weight, low-density lipoprotein cholesterol, and systolic blood pressure decreased significantly. Nausea was the most frequent gastrointestinal symptom and occurred in 16 patients. Its onset was noted at a mean of 1.7 h after injection, the mean duration was 1.1 h, and it continued for a mean of 9.3 days after the initiation of administration. Patients with nausea showed a significant decrease in hemoglobin Alc, glycoalbumin, or body weight compared with those without nausea. These findings suggest that a more marked improvement in metabolic parameters by exenatide can be partly dependent on the manifestation of gastrointestinal symptoms.
Collapse
Affiliation(s)
- Masaru Tokuda
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya 663-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Shin HS, Ingram JR, McGill AT, Poppitt SD. Lipids, CHOs, proteins: can all macronutrients put a 'brake' on eating? Physiol Behav 2013; 120:114-23. [PMID: 23911804 DOI: 10.1016/j.physbeh.2013.07.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 01/09/2013] [Accepted: 07/23/2013] [Indexed: 01/18/2023]
Abstract
The gastrointestinal (GI) tract and specifically the most distal part of the small intestine, the ileum, has become a renewed focus of interest for mechanisms targeting appetite suppression. The 'ileal brake' is stimulated when energy-containing nutrients are delivered beyond the duodenum and jejunum and into the ileum, and is named for the feedback loop which slows or 'brakes' gastric emptying and duodeno-jejunal motility. More recently it has been hypothesized that the ileal brake also promotes secretion of satiety-enhancing GI peptides and suppresses hunger, placing a 'brake' on food intake. Postprandial delivery of macronutrients to the ileum, other than unavailable carbohydrates (CHO) which bypass absorption in the small intestine en route to fermentation in the large bowel, is an uncommon event and hence this brake mechanism is rarely activated following a meal. However the ability to place a 'brake' on food intake through delivery of protected nutrients to the ileum is both intriguing and challenging. This review summarizes the current clinical and experimental evidence for activation of the ileal brake by the three food macronutrients, with emphasis on eating behavior and satiety as well as GI function. While clinical studies have shown that exposure of the ileum to lipids, CHOs and proteins may activate GI components of the ileal brake, such as decreased gut motility, gastric emptying and secretion of GI peptides, there is less evidence as yet to support a causal relationship between activation of the GI brake by these macronutrients and the suppression of food intake. The predominance of evidence for an ileal brake on eating comes from lipid studies, where direct lipid infusion into the ileum suppresses both hunger and food intake. Outcomes from oral feeding studies are less conclusive with no evidence that 'protected' lipids have been successfully delivered into the ileum in order to trigger the brake. Whether CHO or protein may induce the ileal brake and suppress food intake has to date been little investigated, although both clearly have GI mediated effects. This review provides an overview of the mechanisms and mediators of activation of the ileal brake and assesses whether it may play an important role in appetite suppression.
Collapse
Affiliation(s)
- H S Shin
- Human Nutrition Unit, University of Auckland, Auckland, New Zealand; School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
50
|
Wu T, Bound MJ, Zhao BR, Standfield SD, Bellon M, Jones KL, Horowitz M, Rayner CK. Effects of a D-xylose preload with or without sitagliptin on gastric emptying, glucagon-like peptide-1, and postprandial glycemia in type 2 diabetes. Diabetes Care 2013; 36:1913-1918. [PMID: 23359361 PMCID: PMC3687261 DOI: 10.2337/dc12-2294] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 12/24/2012] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Macronutrient "preloads" can reduce postprandial glycemia by slowing gastric emptying and stimulating glucagon-like peptide-1 (GLP-1) secretion. An ideal preload would entail minimal additional energy intake and might be optimized by concurrent inhibition of dipeptidyl peptidase-4 (DPP-4). We evaluated the effects of a low-energy D-xylose preload, with or without sitagliptin, on gastric emptying, plasma intact GLP-1 concentrations, and postprandial glycemia in type 2 diabetes. RESEARCH DESIGN AND METHODS Twelve type 2 diabetic patients were studied on four occasions each. After 100 mg sitagliptin (S) or placebo (P) and an overnight fast, patients consumed a preload drink containing either 50 g D-xylose (X) or 80 mg sucralose (control [C]), followed after 40 min by a mashed potato meal labeled with (13)C-octanoate. Blood was sampled at intervals. Gastric emptying was determined. RESULTS Both peak blood glucose and the amplitude of glycemic excursion were lower after PX and SC than PC (P < 0.01 for each) and were lowest after SX (P < 0.05 for each), while overall blood glucose was lower after SX than PC (P < 0.05). The postprandial insulin-to-glucose ratio was attenuated (P < 0.05) and gastric emptying was slower (P < 0.01) after D-xylose, without any effect of sitagliptin. Plasma GLP-1 concentrations were higher after D-xylose than control only before the meal (P < 0.05) but were sustained postprandially when combined with sitagliptin (P < 0.05). CONCLUSIONS In type 2 diabetes, acute administration of a D-xylose preload reduces postprandial glycemia and enhances the effect of a DPP-4 inhibitor.
Collapse
Affiliation(s)
- Tongzhi Wu
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Michelle J. Bound
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Beiyi R. Zhao
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Scott D. Standfield
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Max Bellon
- Department of Nuclear Medicine, Royal Adelaide Hospital, Adelaide, Australia
| | - Karen L. Jones
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Michael Horowitz
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| | - Christopher K. Rayner
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, Australia
| |
Collapse
|