1
|
Jalleh RJ, Rayner CK, Hausken T, Jones KL, Camilleri M, Horowitz M. Gastrointestinal effects of GLP-1 receptor agonists: mechanisms, management, and future directions. Lancet Gastroenterol Hepatol 2024; 9:957-964. [PMID: 39096914 DOI: 10.1016/s2468-1253(24)00188-2] [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: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 08/05/2024]
Abstract
The availability of glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) such as liraglutide and semaglutide, and a GLP-1 and glucose dependent insulinotropic polypeptide coagonist (tirzepatide) represents a paradigm shift in the management of both type 2 diabetes and obesity. There is now considerable attention, including in the public media, on the effect of both long-acting and short-acting GLP-1RAs to delay gastric emptying. Although slowed gastric emptying is integral to reducing post-prandial blood glucose responses in type 2 diabetes, marked slowing of gastric emptying might also increase the propensity for longer intragastric retention of food, with a consequent increased risk of aspiration at the time of surgery or upper gastrointestinal endoscopy. This Personal View summarises current knowledge of the effects of GLP-1 and GLP-1RAs on gastrointestinal physiology, particularly gastric emptying, and discusses the implications for the development of sound pre-operative or pre-procedural guidelines. The development of pre-procedural guidelines is currently compromised by the poor evidence base, particularly in relation to the effect of long-acting GLP-1RAs on gastric emptying. We suggest pre-procedural management pathways for individuals on GLP-1RA-based therapy and discuss priorities for future research.
Collapse
Affiliation(s)
- Ryan J Jalleh
- Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia
| | - Chris K Rayner
- Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia
| | - Trygve Hausken
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Karen L Jones
- Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Michael Horowitz
- Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
2
|
Borab ZM, Fisher SM, Gimenez A, Raskin P, Valek T, Rohrich RJ. Emerging Role of Semaglutide and GLP-1 Agonist Medications in Plastic Surgery: A Note of Caution. Plast Reconstr Surg 2024; 154:629e-631e. [PMID: 38507525 DOI: 10.1097/prs.0000000000011418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
|
3
|
Liu QK. Mechanisms of action and therapeutic applications of GLP-1 and dual GIP/GLP-1 receptor agonists. Front Endocrinol (Lausanne) 2024; 15:1431292. [PMID: 39114288 PMCID: PMC11304055 DOI: 10.3389/fendo.2024.1431292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are two incretins that bind to their respective receptors and activate the downstream signaling in various tissues and organs. Both GIP and GLP-1 play roles in regulating food intake by stimulating neurons in the brain's satiety center. They also stimulate insulin secretion in pancreatic β-cells, but their effects on glucagon production in pancreatic α-cells differ, with GIP having a glucagonotropic effect during hypoglycemia and GLP-1 exhibiting glucagonostatic effect during hyperglycemia. Additionally, GIP directly stimulates lipogenesis, while GLP-1 indirectly promotes lipolysis, collectively maintaining healthy adipocytes, reducing ectopic fat distribution, and increasing the production and secretion of adiponectin from adipocytes. Together, these two incretins contribute to metabolic homeostasis, preventing both hyperglycemia and hypoglycemia, mitigating dyslipidemia, and reducing the risk of cardiovascular diseases in individuals with type 2 diabetes and obesity. Several GLP-1 and dual GIP/GLP-1 receptor agonists have been developed to harness these pharmacological effects in the treatment of type 2 diabetes, with some demonstrating robust effectiveness in weight management and prevention of cardiovascular diseases. Elucidating the underlying cellular and molecular mechanisms could potentially usher in the development of new generations of incretin mimetics with enhanced efficacy and fewer adverse effects. The treatment guidelines are evolving based on clinical trial outcomes, shaping the management of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Qiyuan Keith Liu
- MedStar Medical Group, MedStar Montgomery Medical Center, Olney, MD, United States
| |
Collapse
|
4
|
Dumiaty Y, Underwood BM, Phy-Lim J, Chee MJ. Neurocircuitry underlying the actions of glucagon-like peptide 1 and peptide YY 3-36 in the suppression of food, drug-seeking, and anxiogenesis. Neuropeptides 2024; 105:102427. [PMID: 38579490 DOI: 10.1016/j.npep.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.
Collapse
Affiliation(s)
- Yasmina Dumiaty
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Brett M Underwood
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Jenny Phy-Lim
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
5
|
Huang RL, Huang WK, Xiao XY, Ma LF, Gu HZR, Yang GP. Diagnosis and treatment of post-cholecystectomy diarrhoea. World J Gastrointest Surg 2023; 15:2398-2405. [PMID: 38111762 PMCID: PMC10725554 DOI: 10.4240/wjgs.v15.i11.2398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/22/2023] [Accepted: 09/22/2023] [Indexed: 11/26/2023] Open
Abstract
The incidence of cholecystitis is relatively high in developed countries and may usually be attributed to gallstones, the treatment for which involves complete surgical removal of the gallbladder (cholecystectomy). Bile acids produced following cholecystectomy continue to flow into the duodenum but are poorly absorbed by the colon. Excessive bile acids in the colon stimulate mucosal secretion of water and electrolytes leading, in severe cases, to diarrhoea. Bile acid diarrhoea (BAD) is difficult to diagnose, requiring a comprehensive medical history and physical examination in combination with laboratory evaluation. The current work reviews the diagnosis and treatment of BAD following cholecystectomy.
Collapse
Affiliation(s)
- Rang-Lang Huang
- Department of Hepatobiliary and Pancreatic Surgery, The Third Xiangya Hospital of The Central South University, Changsha 410013, Hunan Province, China
| | - Wen-Kai Huang
- Department of General Medicine, The Third Xiangya Hospital of The Central South University, Changsha 410013, Hunan Province, China
| | - Xiang-Yi Xiao
- The Xiangya School of Medicine, The Central South University, Changsha 410013, Hunan Province, China
| | - Lin-Feng Ma
- The Xiangya School of Medicine, The Central South University, Changsha 410013, Hunan Province, China
| | - He-Zi-Rui Gu
- The Xiangya School of Medicine, The Central South University, Changsha 410013, Hunan Province, China
| | - Guo-Ping Yang
- Department of Clinical Pharmacy, The Third Hospital of The Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
6
|
Wang S, Wang Y, Lin L, Li Z, Liu F, Zhu L, Chen J, Zhang N, Cao X, Ran S, Liu G, Gao P, Sun W, Peng L, Zhuang J, Meng H. Layer-Specific BTX-A Delivery to the Gastric Muscularis Achieves Effective Weight Control and Metabolic Improvement. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300822. [PMID: 37552813 PMCID: PMC10558648 DOI: 10.1002/advs.202300822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/03/2023] [Indexed: 08/10/2023]
Abstract
The rising incidence of health-endangering obesity constantly calls for more effective treatments. Gastric intramural injection of botulinum neurotoxin A (BTX-A) as a new modality carries great promise yet inconsistent therapeutic efficacy. A layer-specific delivery strategy enabled by dissolving microneedles is hence pioneered to investigate the working site of BTX-A and the resulting therapeutic effects. The drug-loaded tips of the layer-specific gastric paralysis microneedles (LGP-MN) rapidly release and achieve uniform distribution of BTX-A within the designated gastric wall layers. In an obesity rat model, the LGP-MNs not only prove safer than conventional injection, but also demonstrate consistently better therapeutic effects with muscular layer delivery, including 16.23% weight loss (3.06-fold enhancement from conventional injection), 55.20% slower gastric emptying rate, improved liver steatosis, lowered blood lipids, and healthier gut microbiota. Further hormonal study reveals that the elevated production of stomach-derived glucagon-like peptide-1 due to the muscularis-targeting LGP-MN treatment is an important contributor to its unique glucose tolerance-improving effect. This study provides clear indication of the gastric muscularis as the most favorable working site of BTX-A for weight loss and metabolic improvement purposes, and meanwhile suggests that the LGP-MNs could serve as a novel clinical approach to treat obesity and metabolic syndromes.
Collapse
Affiliation(s)
- Siqi Wang
- Department of General Surgery and Obesity and Metabolic Disease CenterChina–Japan Friendship HospitalBeijing100029China
| | - Yuqiong Wang
- Department of Mechanical and Automation EngineeringThe Chinese University of HongkongHongkong999077China
- School of Biological Science and Medical EngineeringBeihang UniversityBeijing100191China
| | - Long Lin
- Engineering College of Peking UniversityPeking universityBeijing100029China
- School of Mechanical and Electrical EngineeringBeijing University of Chemical TechnologyBeijing100029China
| | - Zongjie Li
- Shanghai Veterinary Research InstituteChinese Academy of Agricultural ScienceShanghai200241China
| | - Fengyi Liu
- School of Mechanical and Electrical EngineeringBeijing University of Chemical TechnologyBeijing100029China
| | - Long Zhu
- School of Mechanical and Electrical EngineeringBeijing University of Chemical TechnologyBeijing100029China
| | - Jie Chen
- Department of UltrasoundChina–Japan Friendship HospitalBeijing100029China
| | - Nianrong Zhang
- Department of General Surgery and Obesity and Metabolic Disease CenterChina–Japan Friendship HospitalBeijing100029China
| | - Xinyu Cao
- Department of General Surgery and Obesity and Metabolic Disease CenterChina–Japan Friendship HospitalBeijing100029China
| | - Sunman Ran
- Department of General Surgery and Obesity and Metabolic Disease CenterChina–Japan Friendship HospitalBeijing100029China
| | - Genzheng Liu
- Department of General Surgery and Obesity and Metabolic Disease CenterChina–Japan Friendship HospitalBeijing100029China
| | - Peng Gao
- Department of Clinical LaboratoryChina–Japan Friendship HospitalBeijing100029China
| | - Weiliang Sun
- Institute of Clinical Medical SciencesChina–Japan Friendship HospitalBeijing100029China
| | - Liang Peng
- Institute of Clinical Medical SciencesChina–Japan Friendship HospitalBeijing100029China
| | - Jian Zhuang
- School of Mechanical and Electrical EngineeringBeijing University of Chemical TechnologyBeijing100029China
| | - Hua Meng
- Department of General Surgery and Obesity and Metabolic Disease CenterChina–Japan Friendship HospitalBeijing100029China
| |
Collapse
|
7
|
Gulak MA, Murphy P. Regurgitation under anesthesia in a fasted patient prescribed semaglutide for weight loss: a case report. Can J Anaesth 2023; 70:1397-1400. [PMID: 37280458 DOI: 10.1007/s12630-023-02521-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 06/08/2023] Open
Abstract
PURPOSE Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) such as semaglutide are a class of medications prescribed to treat type 2 diabetes mellitus, and more recently, as an adjunct for weight loss because of its effects of delaying gastric emptying and suppressing appetite. Semaglutide is a long-acting agent with a half-life of approximately one week, and there are currently no guidelines that address the perioperative management of such agents. CLINICAL FEATURES We describe an unexpected case of regurgitation of a large volume of gastric contents upon induction of general anesthesia in a nondiabetic, nonobese patient despite a long preoperative fasting period (20 hr for solids and eight hours for clear fluids). This patient had no traditional risk factors for regurgitation or aspiration but was taking the GLP-1 RA semaglutide for weight loss and had last taken the medication two days before their scheduled procedure. CONCLUSIONS Patients taking long-acting GLP-1 RAs such as semaglutide may be at risk of pulmonary aspiration under anesthesia. We propose strategies to mitigate this risk including holding the medication four weeks prior to a scheduled procedure when feasible and considering full stomach precautions.
Collapse
Affiliation(s)
- Michael A Gulak
- Department of Anesthesiology and Pain Medicine, University of Toronto, 12th Floor, 123 Edward Street, Toronto, ON, M5G 1E2, Canada.
| | - Patricia Murphy
- Department of Anesthesia and Pain Management, Toronto General Hospital, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| |
Collapse
|
8
|
Jones PM, Hobai IA, Murphy PM. Anesthesia and glucagon-like peptide-1 receptor agonists: proceed with caution! Can J Anaesth 2023; 70:1281-1286. [PMID: 37466910 DOI: 10.1007/s12630-023-02550-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/20/2023] Open
Affiliation(s)
- Philip M Jones
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Ion A Hobai
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Patricia M Murphy
- Department of Anesthesiology and Pain Medicine, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
9
|
Brodersen K, Mose M, Ramer Mikkelsen U, Jørgensen JOL, Festersen Nielsen M, Møller N, Wegeberg A, Brock C, Hartmann B, Holst JJ, Rittig N. Prolonged lipopolysaccharide-induced illness elevates glucagon-like peptide-1 and suppresses peptide YY: A human-randomized cross-over trial. Physiol Rep 2022; 10:e15462. [PMID: 36117310 PMCID: PMC9483438 DOI: 10.14814/phy2.15462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023] Open
Abstract
Severe systemic inflammation is associated with nausea, loss of appetite, and delayed gastric emptying, which increases hospitalization admission length and mortality rate. There is a lack of human controlled studies exploring gastric emptying rates and underlying mechanisms during inflammatory conditions. We aimed to investigate if systemic inflammation in young men delays gastro-intestinal transit times, lowers motility, and affects gastrointestinal hormone secretion. This substudy of a randomized crossover trial investigated eight healthy young men on two separate occasions; (I) following an overnight fast (healthy conditions/HC) and (II) fasting and bedrest combined with two lipopolysaccharide (LPS) injections of 1 ng kg-1 following an overnight fast and 0.5 ng kg-1 following another 24 h (systemic inflammation/SI). A standardized protein beverage and a SmartPill capsule (a wireless gastrointestinal monitoring system) were swallowed during each occasion. Whole gut transit time was comparable between HC and SI. SI decreased gastric mean pressure peak amplitude (p = 0.04) and increased pH rise across the pylorus and small bowel pH (p = 0.02) compared with HC. Glucagon-like peptide-1 was elevated during SI compared with HC (p = 0.04). Peptide YY was lower during SI compared with HC (p = 0.007). Prolonged LPS exposure combined with fasting and bedrest elevated glucagon-like peptide 1 concentrations, which may play a role for the nausea and loss of appetite typically associated with SI.
Collapse
Affiliation(s)
- Katrine Brodersen
- Department of SurgeryViborg Regional HospitalViborgDenmark
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
- Steno Diabetes Center AarhusAarhus University HospitalAarhusDenmark
| | - Maike Mose
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
| | | | - Jens Otto Lunde Jørgensen
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
- Department of Endocrinology and Internal MedicineAarhus University HospitalAarhusDenmark
| | | | - Niels Møller
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
| | - Anne‐Marie Wegeberg
- Mech‐Sense, Department of Gastroenterology and HepatologyAalborg University HospitalAalborgDenmark
| | - Christina Brock
- Mech‐Sense, Department of Gastroenterology and HepatologyAalborg University HospitalAalborgDenmark
- Steno Diabetes Center North DenmarkAalborg University HospitalAalborgDenmark
| | - Bolette Hartmann
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenKøbenhavnDenmark
| | - Jens Juul Holst
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenKøbenhavnDenmark
| | - Nikolaj Rittig
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
- Steno Diabetes Center AarhusAarhus University HospitalAarhusDenmark
| |
Collapse
|
10
|
Saito D, Nakagawa Y, Sato T, Fukunaka A, Pereye OB, Maruyama N, Watada H, Fujitani Y. Establishment of an enzyme-linked immunosorbent assay for mouse pancreatic polypeptide clarifies the regulatory mechanism of its secretion from pancreatic γ cells. PLoS One 2022; 17:e0269958. [PMID: 35976945 PMCID: PMC9385059 DOI: 10.1371/journal.pone.0269958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic polypeptide (PP), secreted from γ cells of the islets of Langerhans, is a 36 amino-acid peptide encoded by the Ppy gene. Although previous studies have reported that PP causes a decrease in appetite, the molecular mechanism that regulates PP secretion has not been fully elucidated. Lack of understanding of the regulatory mechanism of PP secretion may be partially owing to the lack of assay systems that can specifically detect PP. We recently developed the mouse monoclonal antibody 23-2D3 that specifically recognizes PP. In the present study, we developed a sandwich enzyme-linked immunosorbent assay for the measurement of mouse PP, and directly monitored intracellular Ca2+ concentrations in Ppy-expressing cells from a newly developed reporter mouse. Using these systems, we identified agonists, such as carbachol and glucose-dependent insulinotropic polypeptide (GIP), which stimulate PP secretion. We further demonstrated that, unlike the case of GIP-induced insulin secretion from β cells, there is a unique mechanism by which PP secretion is triggered by an increase in intracellular Ca2+ concentrations via voltage-dependent calcium channels even in low-glucose conditions.
Collapse
Affiliation(s)
- Daisuke Saito
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuko Nakagawa
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Takashi Sato
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Ayako Fukunaka
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Ofejiro Blessing Pereye
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | | | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshio Fujitani
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
- * E-mail:
| |
Collapse
|
11
|
Shepard BD, Ecelbarger CM. Sodium Glucose Transporter, Type 2 (SGLT2) Inhibitors (SGLT2i) and Glucagon-Like Peptide 1-Receptor Agonists: Newer Therapies in Whole-Body Glucose Stabilization. Semin Nephrol 2021; 41:331-348. [PMID: 34715963 DOI: 10.1016/j.semnephrol.2021.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes is a worldwide epidemic that is increasing rapidly to become the seventh leading cause of death in the world. The increased incidence of this disease mirrors a similar uptick in obesity and metabolic syndrome, and, collectively, these conditions can cause deleterious effects on a number of organ systems including the renal and cardiovascular systems. Historically, treatment of type 2 diabetes has focused on decreasing hyperglycemia and glycated hemoglobin levels. However, it now is appreciated that there is more to the puzzle. Emerging evidence has indicated that newer classes of diabetes drugs, sodium-glucose co-transporter 2 inhibitors and glucagon-like peptide 1-receptor agonists, improve cardiovascular and renal function, while appropriately managing hyperglycemia. In this review, we highlight the recent clinical and preclinical studies that have shed light on sodium-glucose co-transporter 2 inhibitors and glucagon-like peptide 1-receptor agonists and their ability to stabilize blood glucose levels while offering whole-body protection in diabetic and nondiabetic patient populations.
Collapse
Affiliation(s)
- Blythe D Shepard
- Department of Human Science, Georgetown University Medical Center, Washington, DC
| | | |
Collapse
|
12
|
O'Brien R, Buckley MM, O'Malley D. Divergent effects of exendin-4 and interleukin-6 on rat colonic secretory and contractile activity are associated with changes in regional vagal afferent signaling. Neurogastroenterol Motil 2021; 33:e14160. [PMID: 33945195 DOI: 10.1111/nmo.14160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The pro-inflammatory cytokine, interleukin (IL)-6 is elevated in individuals with the functional bowel disorder, irritable bowel syndrome (IBS). IL-6 can independently modify intestinal secreto-motor function, thereby contributing to IBS pathophysiology. Additionally, hormonal changes may underlie symptom flares. Post-prandial exacerbation of IBS symptoms has been linked to secretion of the incretin hormone, glucagon-like peptide-1 (GLP-1), which can also influence colonic secreto-motor activity. This study aimed to ascertain if the effects of GLP-1 on colonic secretory and contractile activity was impacted by elevated IL-6 levels and if sensory signals regarding such changes were reflected in altered vagal afferent activity. METHODS Colonic secretory currents and circular muscle contractile activity was investigated in Sprague Dawley rats using Ussing chamber and organ bath electrophysiology. Regional afferent signaling was assessed using extracellular electrophysiological recordings from colonic vagal afferents. KEY RESULTS Application of the GLP-1 receptor agonist, exendin-4 (Ex-4) in the presence of IL-6 potentiated colonic secretory currents and transepithelial resistance. Vagal afferent fibers originating in the submucosal layer exhibited larger responses to Ex-4 when IL-6 was also present. In contrast, co-application of Ex-4 and IL-6 to gut-bath chambers suppressed circular muscle contractile activity. The activity in extrinsic afferents originating in the colonic myenteric layer was similarly suppressed. CONCLUSIONS & INFERENCES Application of Ex-4 in the presence of IL-6 had divergent modulatory effects on colonic secretion and contractile activity. Similar patterns were observed in vagal afferent signaling originating in the submucosal and myenteric neuronal layers, indicating regional afferent activity reflected immune- and endocrine-mediated changes in colonic function.
Collapse
Affiliation(s)
- Rebecca O'Brien
- Department of Physiology, University College Cork, Cork, Ireland
| | - Maria M Buckley
- Department of Physiology, University College Cork, Cork, Ireland
| | - Dervla O'Malley
- Department of Physiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Ribeiro-Parenti L, Jarry AC, Cavin JB, Willemetz A, Le Beyec J, Sannier A, Benadda S, Pelletier AL, Hourseau M, Léger T, Morlet B, Couvelard A, Anini Y, Msika S, Marmuse JP, Ledoux S, Le Gall M, Bado A. Bariatric surgery induces a new gastric mucosa phenotype with increased functional glucagon-like peptide-1 expressing cells. Nat Commun 2021; 12:110. [PMID: 33397977 PMCID: PMC7782689 DOI: 10.1038/s41467-020-20301-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Glucagon-Like Peptide-1 (GLP-1) undergoes rapid inactivation by dipeptidyl peptidase-4 (DPP4) suggesting that target receptors may be activated by locally produced GLP-1. Here we describe GLP-1 positive cells in the rat and human stomach and found these cells co-expressing ghrelin or somatostatin and able to secrete active GLP-1 in the rats. In lean rats, a gastric load of glucose induces a rapid and parallel rise in GLP-1 levels in both the gastric and the portal veins. This rise in portal GLP-1 levels was abrogated in HFD obese rats but restored after vertical sleeve gastrectomy (VSG) surgery. Finally, obese rats and individuals operated on Roux-en-Y gastric bypass and SG display a new gastric mucosa phenotype with hyperplasia of the mucus neck cells concomitant with increased density of GLP-1 positive cells. This report brings to light the contribution of gastric GLP-1 expressing cells that undergo plasticity changes after bariatric surgeries, to circulating GLP-1 levels.
Collapse
Affiliation(s)
- Lara Ribeiro-Parenti
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service de Chirurgie Générale Œsogastrique et Bariatrique, Hôpital Bichat - Claude-Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anne-Charlotte Jarry
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Jean-Baptiste Cavin
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Alexandra Willemetz
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Johanne Le Beyec
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Sorbonne Université, AP-HP, Hôpital Pitié-Salpêtrière-Charles Foix, Biochimie Endocrinienne et Oncologique, Paris, France
| | - Aurélie Sannier
- Department of Pathology Bichat Hospital, AP-HP, 75018, Paris, France
| | - Samira Benadda
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Cell and Tissue Imaging Platform, Inserm, U1149, CNRS, ERL8252, 75018, Paris, France
| | - Anne-Laure Pelletier
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Muriel Hourseau
- Department of Pathology Bichat Hospital, AP-HP, 75018, Paris, France
| | - Thibaut Léger
- Université de Paris, Mass Spectrometry Laboratory, Institut Jacques Monod, UMR 7592, CNRS, 75205, Paris, France
- Université Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Bastien Morlet
- Université de Paris, Mass Spectrometry Laboratory, Institut Jacques Monod, UMR 7592, CNRS, 75205, Paris, France
| | - Anne Couvelard
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Department of Pathology Bichat Hospital, AP-HP, 75018, Paris, France
| | - Younes Anini
- Department of Obstetrics and Gynecology, Dalhousie University, IWK Health Centre, Halifax, New Brunswick, Canada
| | - Simon Msika
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service de Chirurgie Générale Œsogastrique et Bariatrique, Hôpital Bichat - Claude-Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Pierre Marmuse
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service de Chirurgie Générale Œsogastrique et Bariatrique, Hôpital Bichat - Claude-Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sévérine Ledoux
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service des Explorations Fonctionnelles Hôpital Louis Mourier, AP-HP, Centre Intégré Nord Francilien de prise en charge de l'Obésité (CINFO), 92701, Colombes, France
| | - Maude Le Gall
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France.
| | - André Bado
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France.
| |
Collapse
|
14
|
Malbert CH, Chauvin A, Horowitz M, Jones KL. Pancreatic GLP-1r binding potential is reduced in insulin-resistant pigs. BMJ Open Diabetes Res Care 2020; 8:8/2/e001540. [PMID: 33132211 PMCID: PMC7607594 DOI: 10.1136/bmjdrc-2020-001540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The insulinotropic capacity of exogenous glucagon like peptide-1 (GLP-1) is reduced in type 2 diabetes and the insulin-resistant obese. We have tested the hypothesis that this response is the consequence of a reduced pancreatic GLP-1 receptor (GLP-1r) density in insulin-resistant obese animals. RESEARCH DESIGN AND METHODS GLP-1r density was measured in lean and insulin-resistant adult miniature pigs after the administration of a 68Ga-labeled GLP-1r agonist. The effect of hyperinsulinemia on GLP-1r was assessed using sequential positron emission tomography (PET), both in the fasted state and during a clamp. The impact of tissue perfusion, which could account for changes in GLP-1r agonist uptake, was also investigated using 68Ga-DOTA imaging. RESULTS GLP-1r binding potential in the obese pancreas was reduced by 75% compared with lean animals. Similar reductions were evident for fat tissue, but not for the duodenum. In the lean group, induced hyperinsulinemia reduced pancreatic GLP-1r density to a level comparable with that of the obese group. The reduction in blood to tissue transfer of the GLP-1r ligand paralleled that of tissue perfusion estimated using 68Ga-DOTA. CONCLUSIONS These observations establish that a reduction in abdominal tissue perfusion and a lower GLP-1r density account for the diminished insulinotropic effect of GLP-1 agonists in type 2 diabetes.
Collapse
Affiliation(s)
| | - Alain Chauvin
- UEPR Unit, Department of Animal Physiology, INRAE, Saint-Gilles, France
| | - Michael Horowitz
- Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Karen L Jones
- Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
15
|
Higuchi K, Futagami S, Yamawaki H, Murakami M, Kirita K, Agawa S, Ikeda G, Noda H, Kodaka Y, Ueki N, Kaneko K, Gudis K, Ohashi R, Iwakiri K. Endosonographic features in patients with non-alcoholic early chronic pancreatitis improved with treatment at one year follow up. J Clin Biochem Nutr 2020; 68:86-94. [PMID: 33536717 PMCID: PMC7844654 DOI: 10.3164/jcbn.19-130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
Since the prevention of early chronic pancreatitis (ECP) into chronic pancreatitis might be critical for the reduction of pancreatic cancer, we tried to clarify the pathophysiology of ECP patients, focusing on ECP patients without alcoholic chronic pancreatitis. 27 ECP patients without alcoholic chronic pancreatitis and 33 patients with functional dyspepsia with pancreatic enzyme abnormalities (FD-P) were enrolled in this study. Diagnosis of ECP was made when imaging findings showed the presence of more than 2 out of 7 endoscopic ultrasound features. Duodenal degranulated eosinophils and glucagon-like peptide 1 producing cells were estimated by immunostaining. There were no significant differences in characteristics and psychogenic factors between ECP and FD-P patients. Interestingly, endoscopic ultrasound score in ECP patients significantly improved, albeit clinical symptoms in ECP patients showed no improvement at one year follow up. The extent of migration of duodenal degranulated eosinophils in FD-P patients was significantly higher compared to that in ECP patients. The levels of elastase-1 and trypsin in ECP patients with improved endoscopic ultrasound features were significantly reduced by the treatment. Further studies will be needed to clarify whether clinical symptoms and endoscopic ultrasound features in ECP patients without alcoholic chronic pancreatitis were improved in longer follow up study.
Collapse
Affiliation(s)
- Kazutoshi Higuchi
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Seiji Futagami
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Hiroshi Yamawaki
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Makoto Murakami
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Kumiko Kirita
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Shuhei Agawa
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Go Ikeda
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Hiroto Noda
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Yasuhiro Kodaka
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Nobue Ueki
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Keiko Kaneko
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Katya Gudis
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Ryuji Ohashi
- Department of Diagnostic Pathology, Nippon Medical School Musashi Kosugi Hospital, 1-396 Kosugi-cho, Nakahara-ku, Kawasaki, Kanagawa 211-8533, Japan
| | - Katsuhiko Iwakiri
- Department of Gastroenterology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| |
Collapse
|
16
|
Husnik R, Gaschen FP, Fletcher JM, Gaschen L. Ultrasonographic assessment of the effect of metoclopramide, erythromycin, and exenatide on solid-phase gastric emptying in healthy cats. J Vet Intern Med 2020; 34:1440-1446. [PMID: 32515089 PMCID: PMC7379023 DOI: 10.1111/jvim.15787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022] Open
Abstract
Background Available data on the effect of gastrointestinal motility‐modifying drugs in cats are limited. Most recommendations for drug usage and dosage are based on collective clinical experience. Objectives To assess the effects of metoclopramide, erythromycin, and exenatide on gastric emptying (GE) and gastric motility in comparison to placebo. We hypothesized that metoclopramide and erythromycin would have prokinetic gastric effects, whereas exenatide would prolong GE times and decrease the motility index (MI) of antral contractions. Animals Eight healthy domestic shorthair cats. Methods Each cat had 4 separate ultrasonographic assessments. In a prospective, randomized, double‐blind, 4‐way crossover design, cats received placebo, metoclopramide, erythromycin, or exenatide for 2 days followed by a minimum 5‐day washout period. Ultrasonographic GE times and MI were compared to placebo. Results When compared to placebo, the rate of GE was significantly faster after administration of metoclopramide and erythromycin. Significant differences were found at all fractions of GE after administration of erythromycin and all but 1 fraction after metoclopramide when compared to placebo. The rate of GE in the first half of the GE curve was significantly slower after exenatide administration. The total area under the Ml curve was significantly larger after administration of metoclopramide and erythromycin than after placebo. Conclusions and Clinical Importance Metoclopramide and erythromycin shorten GE times and increase the MI of antral contractions, thus having a prokinetic effect in the stomach of healthy cats, whereas exenatide causes an initial delay in GE.
Collapse
Affiliation(s)
- Roman Husnik
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Frederic P Gaschen
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | | | - Lorrie Gaschen
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| |
Collapse
|
17
|
O'Brien R, O'Malley D. The Glucagon-like peptide-1 receptor agonist, exendin-4, ameliorated gastrointestinal dysfunction in the Wistar Kyoto rat model of Irritable Bowel Syndrome. Neurogastroenterol Motil 2020; 32:e13738. [PMID: 31602785 DOI: 10.1111/nmo.13738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/15/2019] [Accepted: 09/18/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Glucagon-like peptide-1 (GLP-1) is beneficial in relieving pain-related symptoms of Irritable bowel syndrome (IBS), a prevalent, multi-factorial functional bowel disorder characterized by diarrhea and/or constipation, abdominal bloating, and pain. Activation of myenteric neurons has been implicated in the inhibitory effects of GLP-1 on gastrointestinal motility; however, the mechanisms of action underlying this are not clear. METHODS A rat model of IBS was used to examine physiological changes evoked by intraperitoneal administration of a GLP-1 receptor agonist, exendin-4. Behavioral and physiological analysis of stress-sensitive Wister Kyoto (WKY) rats was used to determine if administration of exendin-4, in the presence or absence of neutralizing interleukin-6 receptor monoclonal antibodies, modified IBS-like symptoms. Immunofluorescence, calcium imaging, and Western blotting techniques were used to investigate the potential role of enteric neural plexi and tight junction protein expression in this effect. KEY RESULTS Consistent with the expression of GLP-1 and interleukin-6 receptors in both submucosal and myenteric ganglia, exendin-4 and interleukin-6 stimulated calcium responses in these neurons. In vivo administration of exendin-4 normalized stress-induced defecation and visceral pain sensitivity in WKY rats. No additional changes were noted in rats co-treated with exendin-4 and anti-interleukin-6 receptor antibodies. Mucosal expression of occludin, a tight junction protein, was decreased by exendin-4. Centrally regulated anxiety-like behaviors were not modified. CONCLUSIONS AND INFERENCES These data suggest that intraperitoneal injection of exendin-4 improves bowel dysfunction in WKY rats without impacting on centrally regulated anxiety-like behaviors. Modulation of enteric neuronal function and tight junction expression appear to underlie the functional benefits of this intervention.
Collapse
Affiliation(s)
- Rebecca O'Brien
- Department of Physiology, University College Cork, Cork, Ireland
| | - Dervla O'Malley
- Department of Physiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
18
|
Buckley MM, O'Brien R, Buckley JM, O'Malley D. GHSR-1 agonist sensitizes rat colonic intrinsic and extrinsic neurons to exendin-4: A role in the manifestation of postprandial gastrointestinal symptoms in irritable bowel syndrome? Neurogastroenterol Motil 2019; 31:e13684. [PMID: 31311066 DOI: 10.1111/nmo.13684] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/19/2019] [Accepted: 07/08/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Patients with irritable bowel syndrome (IBS) may experience postprandial symptom exacerbation. Nutrients stimulate intestinal release of glucagon-like peptide 1 (GLP-1), an incretin hormone with known gastrointestinal effects. However, prior to the postprandial rise in GLP-1, levels of the hunger hormone, ghrelin, peak. The aims of this study were to determine if ghrelin sensitizes colonic intrinsic and extrinsic neurons to the stimulatory actions of a GLP-1 receptor agonist, and if this differs in a rat model of IBS. METHODS Calcium imaging of enteric neurons was compared between Sprague Dawley and Wistar Kyoto rats. Colonic contractile activity and vagal nerve recordings were also compared between strains. KEY RESULTS Circulating GLP-1 concentrations differ between IBS subtypes. Mechanistically, we have provided evidence that calcium responses evoked by exendin-4, a GLP-1 receptor agonist, are potentiated by a ghrelin receptor (GHSR-1) agonist, in both submucosal and myenteric neurons. Although basal patterns of colonic contractility varied between Sprague Dawley and Wister Kyoto rats, the capacity of exendin-4 to alter smooth muscle function was modified by a GHSR-1 agonist in both strains. Gut-brain signaling via GLP-1-mediated activation of vagal afferents was also potentiated by the GHSR-1 agonist. CONCLUSIONS & INFERENCES These findings support a temporal interaction between ghrelin and GLP-1, where the preprandial peak in ghrelin may temporarily sensitize colonic intrinsic and extrinsic neurons to the neurostimulatory actions of GLP-1. While the sensitizing effects of the GHSR-1 agonist were identified in both rat strains, in the rat model of IBS, underlying contractile activity was aberrant.
Collapse
Affiliation(s)
- Maria M Buckley
- Department of Physiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork, Ireland
| | - Rebecca O'Brien
- Department of Physiology, University College Cork, Cork, Ireland
| | - Julliette M Buckley
- Department of Surgery, University College Cork, Cork, and Mater Private Hospital, Cork, Ireland
| | - Dervla O'Malley
- Department of Physiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
O'Brien R, Buckley MM, Kelliher A, O'Malley D. PI 3-kinase- and ERK-MAPK-dependent mechanisms underlie Glucagon-Like Peptide-1-mediated activation of Sprague Dawley colonic myenteric neurons. Neurogastroenterol Motil 2019; 31:e13631. [PMID: 31121089 DOI: 10.1111/nmo.13631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/02/2019] [Accepted: 05/06/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Glucagon-like peptide (GLP-1) can modify colonic function, with beneficial effects reported in the functional bowel disorder, irritable bowel syndrome (IBS). IBS pathophysiology is characterized by hyper-activation of the hypothalamic-pituitary-adrenal stress axis and altered microbial profiles. This study aims to characterize the neuronal and functional effects of GLP-1 in healthy rat colons to aid understanding of its beneficial effects in moderating bowel dysfunction. METHODS Immunofluorescent and calcium imaging of myenteric neurons prepared from Sprague Dawley rat colons was carried out to elucidate the neuromodulatory actions of the GLP-1 receptor agonist, exendin-4 (Ex-4). Colonic contractile activity was assessed using organ bath physiological recordings. KEY RESULTS Ex-4 induced an elevation of intracellular calcium arising from store release and influx via voltage-gated calcium channels. Ex-4 activated both ERK-MAPK and PI 3-kinase signaling cascades. Neuronal activation was found to underlie suppression of contractile activity in colonic circular muscle. Although the stress hormone, corticotropin-releasing factor (CRF) potentiated the neuronal response to Ex-4, and the functional effects of Ex-4 on colonic circular muscle activity were not altered. CONCLUSIONS AND INFERENCES Ex-4 evoked neurally regulated suppression of rat colonic circular muscle activity. In myenteric neurons, the neurostimulatory effects of Ex-4 were dependent upon activation of PI 3-kinase and ERK-MAPK signaling cascades. No further change in circular muscle function was noted in the presence of CRF suggesting that stress does not impact on colonic function in health. Further studies in a model of IBS are needed to determine whether mechanisms are modified in the context of bowel dysfunction.
Collapse
Affiliation(s)
- Rebecca O'Brien
- Department of Physiology, University College Cork, Cork, Ireland
| | - Maria M Buckley
- Department of Physiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Amy Kelliher
- Department of Physiology, University College Cork, Cork, Ireland
| | - Dervla O'Malley
- Department of Physiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
20
|
Yan L, Tang Q, Quan X, Ren H, Chen W, Xia H, Luo H. Effects of exendin-4 on colonic motility in rats and its underlying mechanism. Neurogastroenterol Motil 2019; 31:e13482. [PMID: 30303298 DOI: 10.1111/nmo.13482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 09/01/2018] [Accepted: 09/04/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists modulate gastrointestinal motility; however, the effects of GLP-1R agonists on colonic motility are still controversial, and the molecular mechanism is unclear. Exendin-4 shares 53% homology with GLP-1 and is a full agonist of GLP-1R. In this study, our aims were to explore the role and mechanism of exendin-4 in isolated rat colonic tissues and cells. METHODS An organ bath system was used to examine the spontaneous contractions of smooth muscle strips. The whole-cell patch-clamp technique was used to investigate the currents of L-type voltage-dependent calcium channels and large conductance Ca2+ -activated K+ (BKCa ) channels in smooth muscle cells. KEY RESULTS Exendin-4 decreased both the amplitude and frequency of spontaneous contractions of smooth muscle strips in a concentration-dependent manner. The inhibitory effect was completely blocked by exendin-4(9-39), a GLP-1R antagonist. Moreover, this effect was partially abolished by tetrodotoxin (TTX), a blocker of neuronal voltage-dependent Na+ channels, Nω-Nitro-l-arginine (L-NNA), a nitric oxide synthase (NOS) inhibitor, apamin, an inhibitor of small-conductance Ca2+ -activated K+ (SK) channels. Whole-cell patch-clamp recordings revealed that exendin-4 inhibited the peak current of L-type calcium channels in colonic smooth muscle cells, but did not change the shape of the current-voltage (I-V) curves. The steady-state activation and steady-state inactivation of L-type calcium channels were not affected. Likewise, BKCa currents were significantly inhibited by exendin-4. CONCLUSIONS Exendin-4 indirectly inhibits colonic muscle activity via a nitrergic and a purinergic neural pathway through NO and ATP release and inhibits L-type voltage-dependent calcium channels and BKCa channels in smooth muscle cells.
Collapse
Affiliation(s)
- Lin Yan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qincai Tang
- Department of Pathology, China Three Gorges University College of Medicine, Yichang, China
| | - Xiaojing Quan
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haixia Ren
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hong Xia
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Gastroenterology, Renmin Hospital of Wuhan University, Key Laboratory of Hubei Province for Digestive System Diseases, Wuhan, China
| | - Hesheng Luo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
21
|
May AT, Crowe MS, Blakeney BA, Mahavadi S, Wang H, Grider JR, Murthy KS. Identification of expression and function of the glucagon-like peptide-1 receptor in colonic smooth muscle. Peptides 2019; 112:48-55. [PMID: 30508636 PMCID: PMC6342651 DOI: 10.1016/j.peptides.2018.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/07/2018] [Accepted: 11/26/2018] [Indexed: 01/01/2023]
Abstract
The insulinotropic effects of the incretin hormone, glucagon-like peptide-1 (GLP-1) are mediated via GLP-1 receptors (GLP-1R) present on pancreatic β cells. GLP-1 causes a decrease in the motility of stomach and intestine which involves both central and peripheral nervous systems. The expression and function of GLP-1R in gastrointestinal smooth muscle, however, are not clear. Muscle strips and isolated muscle cells were prepared from mouse colon and the effect of GLP-1(7-36) amide on acetylcholine (ACh)-induced contraction was measured. Muscle cells in culture were used to identify the expression of GLP-1R and the signaling pathways activated by GLP-1(7-36) amide. GLP-1R was expressed in the mucosal and non-mucosal tissue preparations derived from colon, and in smooth muscle cell cultures devoid of other cells such as enteric neurons. In colonic muscle strips, the addition of GLP-1(7-36) amide caused dose-dependent inhibition of acetylcholine-induced contractions. The effect of GLP-1(7-36) amide was partly inhibited by the neuronal blocker tetrodotoxin and nitric oxide (NO) synthase inhibitor l-NNA suggesting both NO-dependent neural and NO-independent direct effects on smooth muscle. In isolated colonic smooth muscle cells, GLP-1(7-36) amide caused an increase in Gαs activity, cAMP levels, and PKA activity, and inhibited ACh-induced contraction. The effect of GLP-1(7-36) amide on Gαs activity and cAMP levels was blocked by NF449, an inhibitor of Gαs, and the effect of GLP-1(7-36) amide on contraction was blocked by NF449 and myristoylated PKI, an inhibitor of PKA. We conclude that colonic smooth muscle cells express GLP-1R, and GLP-1(7-36) amide inhibits acetylcholine-induced contraction via GLP-1R coupled to the Gαs/cAMP/PKA pathway.
Collapse
Affiliation(s)
- Alexander T May
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Box 980551, Richmond, VA, 23298, United States
| | - Molly S Crowe
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Box 980551, Richmond, VA, 23298, United States
| | - Bryan A Blakeney
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Box 980551, Richmond, VA, 23298, United States
| | - Sunila Mahavadi
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Box 980551, Richmond, VA, 23298, United States
| | - Hongxia Wang
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Box 980551, Richmond, VA, 23298, United States
| | - John R Grider
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Box 980551, Richmond, VA, 23298, United States
| | - Karnam S Murthy
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Box 980551, Richmond, VA, 23298, United States.
| |
Collapse
|
22
|
Veedfald S, Wu T, Bound M, Grivell J, Hartmann B, Rehfeld JF, Deacon CF, Horowitz M, Holst JJ, Rayner CK. Hyperosmolar Duodenal Saline Infusion Lowers Circulating Ghrelin and Stimulates Intestinal Hormone Release in Young Men. J Clin Endocrinol Metab 2018; 103:4409-4418. [PMID: 30053031 DOI: 10.1210/jc.2018-00699] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
CONTEXT The mechanisms regulating the postprandial suppression of ghrelin secretion remain unclear, but recent observations in rats indicate that an increase in duodenal osmolarity is associated with a reduction in ghrelin levels. Several hormones have been implicated in the regulation of ghrelin. OBJECTIVE We hypothesized that intraduodenal infusion of a hyperosmolar solution would lower plasma ghrelin concentrations. DESIGN, SETTING, PARTICIPANTS, AND INTERVENTIONS Eighteen healthy young men were studied after an overnight fast on two occasions in a randomized double-blinded fashion. A nasoduodenal catheter was positioned and isoosmolar (300 mOsm/L) or hyperosmolar (1500 mOsm/L) saline was infused intraduodenally (4 mL/min, t = 0 to 45 minutes). Venous blood was sampled at t = -45, -30, -15, 0, 15, 30, 45, 60, 75, 90, 120, and 180 minutes. MAIN OUTCOME MEASURES Plasma concentrations of ghrelin, glucagonlike peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin (CCK), glucagon, pancreatic polypeptide (PP), neurotensin (NT), peptide YY (PYY), motilin, and glucose. RESULTS Ghrelin concentrations were suppressed with hyperosmolar when compared with isoosmolar saline, and remained lower until t = 180 minutes. CCK, NT, GLP-1, PYY, and glucagon all increased during hyperosmolar, but not isoosmolar, saline infusion (P < 0.01 for all), whereas GIP, PP, and motilin levels were not affected by either infusion. CONCLUSIONS Plasma ghrelin concentrations are lowered, whereas CCK, GLP-1, PYY, NT, and glucagon concentrations are augmented, by hyperosmolar duodenal content in healthy individuals. These observations have implications for the evaluation of studies comparing the effects of different types and loads of nutrients and chemicals on gut hormone secretion.
Collapse
Affiliation(s)
- Simon Veedfald
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tongzhi Wu
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Michelle Bound
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Jacqueline Grivell
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Carolyn F Deacon
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael Horowitz
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| | - Jens J Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christopher K Rayner
- Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
23
|
Wakabayashi M, Futagami S, Yamawaki H, Tatsuguchi A, Kaneko K, Agawa S, Higuchi K, Sakasegawa N, Murakami M, Akimoto T, Kodaka Y, Ueki N, Gudis K, Kawamoto C, Akamizu T, Sakamoto C, Iwakiri K. Comparison of clinical symptoms, gastric motility and fat intake in the early chronic pancreatitis patients with anti-acid therapy-resistant functional dyspepsia patients. PLoS One 2018; 13:e0205165. [PMID: 30403664 PMCID: PMC6221270 DOI: 10.1371/journal.pone.0205165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 08/30/2018] [Indexed: 12/16/2022] Open
Abstract
Background There was no available data concerning the clinical differentiation between the updated definition of early chronic pancreatitis (ECP) and anti-acid therapy-resistant functional dyspepsia (RFD). Aims We aimed to determine whether clinical symptoms, gastric motility, psychogenic factors and fat intake can help distinguish early chronic pancreatitis (ECP) from anti-acid therapy-resistant functional dyspepsia patients with pancreatic enzyme abnormalities (RFD-P) and anti-acid therapy-resistant functional dyspepsia (RFD) patients using endosonography. Methods We enrolled 102 consecutive patients presenting with typical symptoms of RFD patients (n = 52), ECP patients (n = 25) and RFD-P patients (n = 25). ECP patients were diagnosed based on the criteria recommended by the Japan Pancreatic Association. Gastric motility was evaluated by 13C-acetate breath tests. Severity of duodenal inflammation was examined. Results 24.5% of RFD patients were determined as ECP using endosonography. Abdominal pain score in Gastrointestinal Symptom Rating Scale (GSRS) in the patients with ECP was significantly lower compared to that in the patients with RFD-P. There were no significant differences in State-Trait Inventory (STAI)-state/-trait scores, Self-Rating Questionnaire for Depression (SRQ-D) scores and clinical symptoms for fat intake among three groups. The early phase of gastric emptying (AUC5; AUC15) in ECP and RFD-P patients were significantly disturbed compared to those in RFD patients. Conclusions Evaluation of severity of abdominal pain and measurement of the early phase of gastric emptying will be useful tools to distinguish ECP patients from RFD patients. Accurate diagnosis of ECP patients may contribute to the prevention from advancing of chronic pancreatitis.
Collapse
Affiliation(s)
- Mako Wakabayashi
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Seiji Futagami
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
- * E-mail:
| | - Hiroshi Yamawaki
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Atsushi Tatsuguchi
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Keiko Kaneko
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Shuhei Agawa
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Kazutoshi Higuchi
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Noriko Sakasegawa
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Makoto Murakami
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Teppei Akimoto
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Yasuhiro Kodaka
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Nobue Ueki
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Kaya Gudis
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Chiaki Kawamoto
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Takashi Akamizu
- The First Department of Medicine, Wakayama Medical Unversity, Wakayama, Japan
| | - Choitsu Sakamoto
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Katsuhiko Iwakiri
- Department of Internal Medicine, Division of Gastroenterology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
24
|
Halim MA, Degerblad M, Sundbom M, Karlbom U, Holst JJ, Webb DL, Hellström PM. Glucagon-Like Peptide-1 Inhibits Prandial Gastrointestinal Motility Through Myenteric Neuronal Mechanisms in Humans. J Clin Endocrinol Metab 2018; 103:575-585. [PMID: 29177486 DOI: 10.1210/jc.2017-02006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 11/16/2017] [Indexed: 02/07/2023]
Abstract
CONTEXT Glucagon-like peptide-1 (GLP-1) secretion from l-cells and postprandial inhibition of gastrointestinal motility. OBJECTIVE Investigate whether physiological plasma concentrations of GLP-1 inhibit human postprandial motility and determine mechanism of action of GLP-1 and analog ROSE-010 action. DESIGN Single-blind parallel study. SETTING University hospital laboratory. PARTICIPANTS Healthy volunteers investigated with antroduodenal manometry. Human gastric and intestinal muscle strips. INTERVENTIONS Motility indices (MIs) obtained before and during GLP-1 or saline infusion. Plasma GLP-1 and glucagon-like peptide-2 (GLP-2) measured by radioimmunoassay. Gastrointestinal muscle strips investigated for GLP-1- and ROSE-010-induced relaxation employing GLP-1 and GLP-2 and their receptor localization, and blockers exendin(9-39)amide, Lω-nitro-monomethylarginine (L-NMMA), 2',5'-dideoxyadenosine (DDA), and tetrodotoxin (TTX) to reveal target mechanism of GLP-1 action. MAIN OUTCOME MEASURES Postprandial gastrointestinal relaxation by GLP-1. RESULTS In humans, food intake increased MI to 6.4 ± 0.3 (antrum), 5.7 ± 0.4 (duodenum), and 5.9 ± 0.2 (jejunum). GLP-1 administered intravenously raised plasma GLP-1, but not GLP-2. GLP-1 0.7 pmol/kg/min suppressed corresponding MI to 4.6 ± 0.2, 4.7 ± 0.4, and 5.0 ± 0.2, whereas 1.2 pmol/kg/min suppressed MI to 5.4 ± 0.2, 4.4 ± 0.3, and 5.4 ± 0.3 (P < 0.0001 to 0.005). In vitro, GLP-1 and ROSE-010 prevented contractions by bethanechol and electric field stimulation (P < 0.005 to 0.05). These effects were disinhibited by exendin(9-39)amide, L-NMMA, DDA, or TTX. GLP-1 and GLP-2 were localized to epithelial cells, GLP-1 also at myenteric neurons. GLP-1R and GLP-2R were localized at myenteric neurons but not muscle. CONCLUSIONS GLP-1 and ROSE-010 inhibit postprandial gastrointestinal motility through GLP-1R at myenteric neurons, involving nitrergic and cyclic adenosine monophosphate-dependent mechanisms.
Collapse
Affiliation(s)
- Md Abdul Halim
- Department of Medical Sciences, Gastroenterology Unit, Uppsala University, Uppsala, Sweden
| | - Marie Degerblad
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Urban Karlbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jens Juul Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dominic-Luc Webb
- Department of Medical Sciences, Gastroenterology Unit, Uppsala University, Uppsala, Sweden
| | - Per M Hellström
- Department of Medical Sciences, Gastroenterology Unit, Uppsala University, Uppsala, Sweden
| |
Collapse
|
25
|
Muscogiuri G, Balercia G, Barrea L, Cignarelli A, Giorgino F, Holst JJ, Laudisio D, Orio F, Tirabassi G, Colao A. Gut: A key player in the pathogenesis of type 2 diabetes? Crit Rev Food Sci Nutr 2017; 58:1294-1309. [PMID: 27892685 DOI: 10.1080/10408398.2016.1252712] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The gut regulates glucose and energy homeostasis; thus, the presence of ingested nutrients into the gut activates sensing mechanisms that affect both glucose homeostasis and regulate food intake. Increasing evidence suggest that gut may also play a key role in the pathogenesis of type 2 diabetes which may be related to both the intestinal microbiological profile and patterns of gut hormones secretion. Intestinal microbiota includes trillions of microorganisms but its composition and function may be adversely affected in type 2 diabetes. The intestinal microbiota may be responsible of the secretion of molecules that may impair insulin secretion/action. At the same time, intestinal milieu regulates the secretion of hormones such as GLP-1, GIP, ghrelin, gastrin, somatostatin, CCK, serotonin, peptide YY, GLP-2, all of which importantly influence metabolism in general and in particular glucose metabolism. Thus, the aim of this paper is to review the current evidence on the role of the gut in the pathogenesis of type 2 diabetes, taking into account both hormonal and microbiological aspects.
Collapse
Affiliation(s)
| | - Giancarlo Balercia
- b Division of Endocrinology, Department of Clinical and Molecular Sciences , Umberto I Hospital, Polytechnic University of Marche , Ancona , Italy
| | | | - Angelo Cignarelli
- c Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases , University of Bari Aldo Moro , Bari , Italy
| | - Francesco Giorgino
- c Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases , University of Bari Aldo Moro , Bari , Italy
| | - Jens J Holst
- d NNF Center for Basic Metabolic Research and Department of Biomedical Sciences , Panum Institute, University of Copenhagen, Copenhagen , Denmark
| | | | - Francesco Orio
- e Endocrinology, Department of Sports Science and Wellness , "Parthenope" University Naples , Naples , Italy
| | - Giacomo Tirabassi
- b Division of Endocrinology, Department of Clinical and Molecular Sciences , Umberto I Hospital, Polytechnic University of Marche , Ancona , Italy
| | - Annamaria Colao
- f Department of Clinical Medicine and Surgery , "Federico II" University of Naples , Naples , Italy
| |
Collapse
|
26
|
Jessen L, Smith EP, Ulrich-Lai Y, Herman JP, Seeley RJ, Sandoval D, D’Alessio D. Central Nervous System GLP-1 Receptors Regulate Islet Hormone Secretion and Glucose Homeostasis in Male Rats. Endocrinology 2017; 158:2124-2133. [PMID: 28430981 PMCID: PMC5505222 DOI: 10.1210/en.2016-1826] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/13/2017] [Indexed: 02/07/2023]
Abstract
The glucagon-like peptide 1 (GLP-1) system plays an important role in blood glucose regulation, in great part through coordinate control of insulin and glucagon secretion. These effects are generally attributed to GLP-1 produced in peripheral sites, principally the intestine. GLP-1 is also produced in hindbrain neurons that signal through GLP-1 receptors (GLP-1rs) expressed in brain regions involved in metabolic regulation. GLP-1 in the central nervous system (CNS) induces satiety, visceral illness, and stress responses. However, recent evidence suggests CNS GLP-1 is also involved in glucose regulation. To test the hypothesis that central GLP-1 regulates islet hormone secretion, conscious rats were given intracerebroventricular (ICV) GLP-1, GLP-1r antagonist exendin-[9-39] (Ex-9), or saline during fasting or hyperglycemia from intravenous glucose. Administration of CNS GLP-1 increased fasting glucose, glucagon, corticosterone, and epinephrine and blunted insulin secretion in response to hyperglycemia. Paradoxically, GLP-1r blockade with ICV Ex-9 also reduced glucose-stimulated insulin secretion, and administration of ICV Ex-9 to freely feeding rats caused mild glucose intolerance. Thus, direct administration of CNS GLP-1 affected islet hormone secretion counter to what is seen with peripherally administered GLP-1, an effect likely due to stimulation of sympathetic nervous system activity. In contrast, blockade of brain GLP-1r supports a role for CNS GLP-1 on glucose-stimulated insulin secretion and glucose control after a meal. These findings suggest a model in which activation of CNS GLP-1r by endogenous peptide promotes glucose tolerance, an effect that can be overridden by stress responses stimulated by exogenous GLP-1.
Collapse
Affiliation(s)
- Lene Jessen
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio 45219
| | - Eric P. Smith
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio 45219
| | - Yvonne Ulrich-Lai
- Department of Psychiatry and Behavioral Neursocience, University of Cincinnati, Cincinnati, Ohio 45219
| | - James P. Herman
- Department of Psychiatry and Behavioral Neursocience, University of Cincinnati, Cincinnati, Ohio 45219
| | - Randy J. Seeley
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio 45219
| | - Darleen Sandoval
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio 45219
| | - David D’Alessio
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio 45219
| |
Collapse
|
27
|
Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory Controls and Physiological Roles in Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 2017; 97:411-463. [PMID: 28003328 PMCID: PMC6151490 DOI: 10.1152/physrev.00031.2014] [Citation(s) in RCA: 367] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.
Collapse
Affiliation(s)
- Robert E Steinert
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christoph Beglinger
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Nori Geary
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
28
|
El Khoury D, Goff HD, Anderson GH. The role of alginates in regulation of food intake and glycemia: a gastroenterological perspective. Crit Rev Food Sci Nutr 2016; 55:1406-24. [PMID: 24915329 DOI: 10.1080/10408398.2012.700654] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulation of food intake through modulation of gastrointestinal responses to ingested foods is an ever-growing component of the therapeutic approaches targeting the obesity epidemic. Alginates, viscous and gel-forming soluble fibers isolated from the cell wall of brown seaweeds and some bacteria, are recently receiving considerable attention because of their potential role in satiation, satiety, and food intake regulation in the short term. Enhancement of gastric distension, delay of gastric emptying, and attenuation of postprandial glucose responses may constitute the basis of their physiological benefits. Offering physical, chemical, sensorial, and physiological advantages over other viscous and gel-forming fibers, alginates constitute promising functional food ingredients for the food industry. Therefore, the current review explores the role of alginates in food intake and glycemic regulation, their underlying modes of action and their potential in food applications.
Collapse
Affiliation(s)
- D El Khoury
- a Department of Nutritional Sciences, Faculty of Medicine, University of Toronto , Toronto , M5S 3E2 , ON , Canada
| | | | | |
Collapse
|
29
|
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: 48] [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
|
30
|
Abstract
After food is ingested, nutrients pass through the gastrointestinal tract, stimulating the release of a range of peptide hormones. Among their many local, central and peripheral actions, these hormones act to mediate glucose metabolism and satiety. Indeed, it is the modification of gut hormone secretion that is considered partly responsible for the normalization of glycaemic control and the reduction in appetite seen in many patients after certain forms of bariatric surgery. This review describes recent developments in our understanding of the secretion and action of anorexigenic gut hormones, primarily concentrating on glucagon-like peptide-1 (GLP-1).
Collapse
Affiliation(s)
- Helen E Parker
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Addenbrooke's Hospital, Cambridge CB2 0XY, UK
| | - Fiona M Gribble
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Addenbrooke's Hospital, Cambridge CB2 0XY, UK
| | - Frank Reimann
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Addenbrooke's Hospital, Cambridge CB2 0XY, UK
| |
Collapse
|
31
|
Plummer MP, Jones KL, Cousins CE, Trahair LG, Meier JJ, Chapman MJ, Horowitz M, Deane AM. Hyperglycemia potentiates the slowing of gastric emptying induced by exogenous GLP-1. Diabetes Care 2015; 38:1123-9. [PMID: 25784665 DOI: 10.2337/dc14-3091] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/23/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Acute hyperglycemia markedly slows gastric emptying. Exogenous GLP-1 also slows gastric emptying, leading to diminished glycemic excursions. The primary objective was to determine whether hyperglycemia potentiates the slowing of gastric emptying induced by GLP-1 administration. RESEARCH DESIGN AND METHODS Ten healthy participants were studied on 4 separate days. Blood glucose was clamped at hyperglycemia using an intravenous infusion of 25% dextrose (∼12 mmol/L; hyper) on 2 days, or maintained at euglycemia (∼6 mmol/L; eu) on 2 days, between t = -15 and 240 min. During hyperglycemic and euglycemic days, participants received intravenous GLP-1 (1.2 pmol/kg/min) and placebo in a randomized double-blind fashion. At t = 0 min, subjects ingested 100 g beef mince labeled with 20 MBq technetium-99m-sulfur colloid and 3 g 3-O-methyl-glucose (3-OMG), a marker of glucose absorption. Gastric emptying was measured scintigraphically from t = 0 to 240 min and serum 3-OMG taken at regular intervals from t = 15 to 240 min. The areas under the curve for gastric emptying and 3-OMG were analyzed using one-way repeated-measures ANOVA with Bonferroni-Holm adjusted post hoc tests. RESULTS Hyperglycemia slowed gastric emptying (eu/placebo vs. hyper/placebo; P < 0.001) as did GLP-1 (eu/placebo vs. eu/GLP-1; P < 0.001). There was an additive effect of GLP-1 and hyperglycemia, such that gastric emptying was markedly slower compared with GLP-1 administration during euglycemia (eu/GLP-1 vs. hyper/GLP-1; P < 0.01). CONCLUSIONS Acute administration of exogenous GLP-1 profoundly slows gastric emptying during hyperglycemia in excess of the slowing induced by GLP-1 during euglycemia. Studies are required to determine the effects of hyperglycemia on gastric emptying with the subcutaneously administered commercially available GLP-1 agonists in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Mark P Plummer
- Discipline of Acute Care Medicine, University of Adelaide, Adelaide, Australia Department of Critical Care Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Karen L Jones
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Caroline E Cousins
- Department of Critical Care Services, Royal Adelaide Hospital, Adelaide, Australia
| | | | - Juris J Meier
- Diabetes Division, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Marianne J Chapman
- Discipline of Acute Care Medicine, University of Adelaide, Adelaide, Australia Department of Critical Care Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Michael Horowitz
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Adam M Deane
- Discipline of Acute Care Medicine, University of Adelaide, Adelaide, Australia Department of Critical Care Services, Royal Adelaide Hospital, Adelaide, Australia
| |
Collapse
|
32
|
Ziegler D, Strom A, Strassburger K, Nowotny B, Zahiragic L, Nowotny PJ, Carstensen-Kirberg M, Herder C, Szendroedi J, Roden M. Differential Patterns and Determinants of Cardiac Autonomic Nerve Dysfunction during Endotoxemia and Oral Fat Load in Humans. PLoS One 2015; 10:e0124242. [PMID: 25893426 PMCID: PMC4403853 DOI: 10.1371/journal.pone.0124242] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/27/2015] [Indexed: 11/24/2022] Open
Abstract
The autonomic nervous system (ANS) plays an important role in regulating the metabolic homeostasis and controlling immune function. ANS alterations can be detected by reduced heart rate variability (HRV) in conditions like diabetes and sepsis. We determined the effects of experimental conditions mimicking inflammation and hyperlipidemia on HRV and heart rate (HR) in relation to the immune, metabolic, and hormonal responses resulting from these interventions. Sixteen lean healthy subjects received intravenous (i.v.) low-dose endotoxin (lipopolysaccharide [LPS]), i.v. fat, oral fat, and i.v. glycerol (control) for 6 hours, during which immune, metabolic, hormonal, and five HRV parameters (pNN50, RMSSD, low-frequency (LF) and high-frequency (HF) power, and LF/HF ratio) were monitored and energy metabolism and insulin sensitivity (M-value) were assessed. LPS infusion induced an increase (AUC) in HR and LF/HF ratio and decline in pNN50 and RMSSD, while oral fat resulted in elevated HR and a transient (hours 1-2) decrease in pNN50, RMSSD, and HF power. During LPS infusion, ΔIL-1ra levels and ΔIL-1ra and ΔIL-1ß gene expression correlated positively with ΔLF/HF ratio and inversely with ΔRMSSD. During oral fat intake, ΔGLP-1 tended to correlate positively with ΔHR and inversely with ΔpNN50 and ΔRMSSD. Following LPS infusion, lipid oxidation correlated positively with HR and inversely with pNN50 and RMSSD, whereas HRV was not related to M-value. In conclusion, suppression of vagal tone and sympathetic predominance during endotoxemia are linked to anti-inflammatory processes and lipid oxidation but not to insulin resistance, while weaker HRV changes in relation to the GLP-1 response are noted during oral fat load.
Collapse
Affiliation(s)
- Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
- * E-mail:
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Klaus Strassburger
- Institute of Biometrics and Epidemiology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Bettina Nowotny
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Lejla Zahiragic
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital, Düsseldorf, Germany
| | - Peter J. Nowotny
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Maren Carstensen-Kirberg
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
33
|
Plummer MP, Jones KL, Annink CE, Cousins CE, Meier JJ, Chapman MJ, Horowitz M, Deane AM. Glucagon-like peptide 1 attenuates the acceleration of gastric emptying induced by hypoglycemia in healthy subjects. Diabetes Care 2014; 37:1509-15. [PMID: 24598243 DOI: 10.2337/dc13-1813] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Exogenous GLP-1 slows gastric emptying in health and diabetes leading to diminished glycemic excursions. Gastric emptying is markedly accelerated by hypoglycemia. The primary objective was to determine whether GLP-1 attenuates the acceleration of gastric emptying induced by hypoglycemia. RESEARCH DESIGN AND METHODS Ten healthy volunteers were studied on four separate days in a randomized double-blind fashion. Blood glucose was stabilized using a glucose/insulin clamp at hypoglycemia (2.6 mmol/L on two occasions [hypo]) or euglycemia (6.0 mmol/L on two occasions [eu]) between T = -15 and 45 min before clamping at 6.0 mmol/L until 180 min. During hypoglycemia and euglycemia, subjects received intravenous GLP-1 (1.2 pmol/kg/min) or placebo. At T = 0 min, subjects ingested 100 g beef mince labeled with 20 MBq (99m)Tc-sulfur-colloid and 3 g of 3-O-methyl-glucose (3-OMG), a marker of glucose absorption. Gastric emptying was measured scintigraphically from T = 0 to 180 min and serum 3-OMG taken at 15-min intervals. The areas under the curve for gastric emptying and 3-OMG concentration were analyzed using one-way repeated-measures ANOVA with Bonferroni-Holm adjusted post hoc tests. RESULTS Gastric emptying was accelerated during hypoglycemia (hypo/placebo vs. eu/placebo; P < 0.001), as was glucose absorption (P < 0.03). GLP-1 slowed emptying during euglycemia (eu/placebo vs. eu/GLP-1; P < 0.001). However, hypoglycemia-induced acceleration of gastric emptying on placebo was markedly diminished by GLP-1 (hypo/placebo vs. hypo/GLP-1; P < 0.008), as was glucose absorption (P < 0.01). CONCLUSIONS Acute administration of exogenous GLP-1 attenuates, but does not abolish, the acceleration of gastric emptying by insulin-induced hypoglycemia in healthy subjects.
Collapse
Affiliation(s)
- Mark P Plummer
- Discipline of Acute Care Medicine, University of Adelaide, Adelaide, AustraliaDepartment of Critical Care Services, Royal Adelaide Hospital, Adelaide, AustraliaCentre for Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, Adelaide, Australia
| | - Karen L Jones
- Centre for Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, Adelaide, AustraliaDiscipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Chris E Annink
- Department of Critical Care Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Caroline E Cousins
- Department of Critical Care Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Juris J Meier
- Diabetes Division, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Marianne J Chapman
- Discipline of Acute Care Medicine, University of Adelaide, Adelaide, AustraliaDepartment of Critical Care Services, Royal Adelaide Hospital, Adelaide, AustraliaCentre for Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, Adelaide, Australia
| | - Michael Horowitz
- Centre for Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, Adelaide, AustraliaDiscipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Adam M Deane
- Discipline of Acute Care Medicine, University of Adelaide, Adelaide, AustraliaDepartment of Critical Care Services, Royal Adelaide Hospital, Adelaide, AustraliaCentre for Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, Adelaide, Australia
| |
Collapse
|
34
|
Werner U. Effects of the GLP-1 receptor agonist lixisenatide on postprandial glucose and gastric emptying--preclinical evidence. J Diabetes Complications 2014; 28:110-4. [PMID: 23992745 DOI: 10.1016/j.jdiacomp.2013.06.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/30/2013] [Accepted: 06/11/2013] [Indexed: 11/15/2022]
Abstract
In addition to promoting glucose homeostasis, glucagon-like peptide 1 (GLP-1) has a number of extra-pancreatic effects that regulate appetite and body weight. GLP-1 delays gastric emptying, which is vital for postprandial glucose (PPG) control. As GLP-1 is rapidly degraded by protease dipeptidyl peptidase-4, a number of degradation-resistant GLP-1 receptor agonists (GLP-1RAs) have been developed for the treatment of Type 2 diabetes mellitus. These agents can be broadly categorized as being short- or long-acting, based on their pharmacokinetic profile. Short-acting agonists predominantly affect PPG and delay gastric emptying in a sustained manner, whereas long-acting agents largely affect fasting plasma glucose and their delay in gastric emptying appears to be subjected to tachyphylaxis. Lixisenatide is a "short-acting" once-daily prandial GLP-1RA. This review provides an overview of the preclinical studies that are currently available and that evaluate the efficacy of lixisenatide on gastric emptying and PPG levels. The preclinical evidence outlined in this review supports the efficacy of lixisenatide in reducing PPG excursions and delaying gastric emptying. Furthermore, in contrast to long-acting agents, the actions of lixisenatide do not appear to be subjected to tachyphylaxis.
Collapse
Affiliation(s)
- Ulrich Werner
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany.
| |
Collapse
|
35
|
Marathe CS, Rayner CK, Jones KL, Horowitz M. Relationships between gastric emptying, postprandial glycemia, and incretin hormones. Diabetes Care 2013; 36:1396-405. [PMID: 23613599 PMCID: PMC3631884 DOI: 10.2337/dc12-1609] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/15/2012] [Indexed: 02/05/2023]
Affiliation(s)
- Chinmay S. Marathe
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, Australia
| | - Christopher K. Rayner
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, Australia
| | - Karen L. Jones
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, Australia
- Centre of Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide, Australia
| | | |
Collapse
|
36
|
Ogbu SO, Agwu KK, Asuzu IU. Effect of Gongronema latifolium on gastric emptying in healthy dogs. World J Gastroenterol 2013; 19:897-902. [PMID: 23431049 PMCID: PMC3574887 DOI: 10.3748/wjg.v19.i6.897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 10/31/2012] [Accepted: 11/24/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate sonographically the effect of Gonogronema latifolium (G. latifolium) on gastric emptying of semi-solid meals in healthy dogs.
METHODS: In a randomized, placebo-controlled experiment, twenty-five clinically healthy dogs were randomly allotted into five groups of five dogs in each group. The placebo group served as the control, and the low, moderate and high dose groups ingested the methanolic leaf extract of G. latifolium in capsules at 100 mg/kg, 250 mg/kg and 500 mg/kg, respectively, while the prokinetic group ingested 0.5 mg/kg capsules of metoclopramide. After a 12-h fast, each group ingested its treatment capsules 30 min before the administration of a test meal. Measurements of gastric emptying and blood glucose levels were obtained 30 min before and immediately after the ingestion of the test meal and thereafter every 15 min for 4 h. This was followed by further measurements every 30 min for another 2 h.
RESULTS: The gastric emptying times of the placebo, low dose, moderate dose, high dose and prokinetic dose groups were 127.0 ± 8.2 min, 135.5 ± 3.7 min, 155.5 ± 3.9 min, 198.0 ± 5.3 min and 59.0 ± 2.5 min, respectively. Gastric emptying times of the moderate and high dose groups were significantly slower than in the placebo control group (155.5 ± 3.9 min, 198.0 ± 5.3 min vs 127.0 ± 8.2 min, P = 0.000). No significant difference in gastric emptying between the low dose and placebo control groups was noted (135.5 ± 3.7 min vs 127.0 ± 8.2 min, P = 0.072). Gastric emptying of the prokinetic group was significantly faster than that of the control group (59.0 ± 2.5 min vs 127.0 ± 8.2 min, P = 0.000). The hypoglycaemic effect of G. latifolium and gastric emptying were inversely related (r = -0.95, P = 0.000).
CONCLUSION: G. latifolium delays gastric emptying and lowers postprandial blood glucose in healthy dogs. It reduces the postprandial blood glucose by delaying gastric emptying.
Collapse
|
37
|
Chen Y, Li Z, Yang Y, Lin L, Zhang H. Role of glucagon-like peptide-1 in the pathogenesis of experimental irritable bowel syndrome rat models. Int J Mol Med 2013; 31:607-13. [PMID: 23338623 DOI: 10.3892/ijmm.2013.1252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 11/01/2012] [Indexed: 12/17/2022] Open
Abstract
Alterations in gut motility and visceral hypersensitivity are two major features of irritable bowel syndrome (IBS). The aim of this study was to investigate the roles of glucagon-like peptide-1 (GLP-1) in the pathogenesis of experimental IBS. Rat models of constipation-predominant IBS (IBS-C) and diarrhea-predominant IBS (IBS-D) were established. Fecal water content and behavioral responses to colorectal distention (CRD), using electromyography (EMG), were measured. The expression of glucagon-like peptide-1 receptor (GLP-1R) in the colon was detected by immunohistochemistry, and the serum concentration of GLP-1 was measured by ELISA assay. The movement of circular and longitudinal colonic muscle was detected using an organ bath recording technique. Compared to controls, the fecal water contents were lower in the IBS-C group, while they were higher in the IBS-D group (P<0.05). EMG response to CRD in the experimental IBS groups was increased compared with their respective controls (P<0.05). GLP-1R was localized in the mucosa layer, circular muscle and myenteric nerve plexus of the colon. Notably, the expression of GLP-1R in the IBS-C group was higher, but in the IBS-D group, it was lower compared with controls. The serum levels of GLP-1 in the IBS-C group were higher compared to those in the IBS-D group (P<0.05). In addition, administration of exogenous GLP-1 and exendin-4 inhibited colonic circular muscle contraction, particularly in the IBS-C group, while there was no significant effect on longitudinal muscle contraction. In conclusion, these results indicated that GLP-1 and GLP-1R are implicated in the pathogenesis of IBS-C and IBS-D.
Collapse
Affiliation(s)
- Yan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | | | | | | | | |
Collapse
|
38
|
Ables GP, Yang KJZ, Vogel S, Hernandez-Ono A, Yu S, Yuen JJ, Birtles S, Buckett LK, Turnbull AV, Goldberg IJ, Blaner WS, Huang LS, Ginsberg HN. Intestinal DGAT1 deficiency reduces postprandial triglyceride and retinyl ester excursions by inhibiting chylomicron secretion and delaying gastric emptying. J Lipid Res 2012; 53:2364-79. [PMID: 22911105 PMCID: PMC3466005 DOI: 10.1194/jlr.m029041] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Acyl CoA:diacylglycerol acyltransferase (DGAT) 1 catalyzes the final step of triglyceride (TG) synthesis. We show that acute administration of a DGAT1 inhibitor (DGAT1i) by oral gavage or genetic deletion of intestinal Dgat1 (intestine-Dgat1(-/-)) markedly reduced postprandial plasma TG and retinyl ester excursions by inhibiting chylomicron secretion in mice. Loss of DGAT1 activity did not affect the efficiency of retinol esterification, but it did reduce TG and retinoid accumulation in the small intestine. In contrast, inhibition of microsomal triglyceride transfer protein (MTP) reduced chylomicron secretion after oral fat/retinol loads, but with accumulation of dietary TG and retinoids in the small intestine. Lack of intestinal accumulation of TG and retinoids in DGAT1i-treated or intestine-Dgat1(-/-) mice resulted, in part, from delayed gastric emptying associated with increased plasma levels of glucagon-like peptide (GLP)-1. However, neither bypassing the stomach through duodenal oil injection nor inhibiting the receptor for GLP-1 normalized postprandial TG or retinyl esters excursions in the absence of DGAT1 activity. In summary, intestinal DGAT1 inhibition or deficiency acutely delayed gastric emptying and inhibited chylomicron secretion; however, the latter occurred when gastric emptying was normal or when lipid was administered directly into the small intestine. Long-term hepatic retinoid metabolism was not impacted by DGAT1 inhibition.
Collapse
Affiliation(s)
- Gene P Ables
- Department of Medicine, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Stevens JE, Horowitz M, Deacon CF, Nauck M, Rayner CK, Jones KL. The effects of sitagliptin on gastric emptying in healthy humans - a randomised, controlled study. Aliment Pharmacol Ther 2012; 36:379-90. [PMID: 22738299 DOI: 10.1111/j.1365-2036.2012.05198.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/03/2012] [Accepted: 06/06/2012] [Indexed: 02/05/2023]
Abstract
BACKGROUND The rate of gastric emptying (GE) and subsequent release of the incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) are critical determinants of postprandial glycaemia in health and type 2 diabetes. Slowing of GE may be the dominant mechanism by which exogenous GLP-1, and some GLP-1 analogues, improve postprandial glycaemia. AIM To determine the effect of sitagliptin on GE in healthy subjects, and the relationships between GE with glycaemia and incretin hormone secretion. METHODS Fifteen volunteers (22.8 ± 0.7 years) were studied on two occasions following 2 days dosing with sitagliptin (100 mg/day) or placebo. GE (scintigraphy), glycaemia and plasma GLP-1 and GIP (total and intact), insulin and glucagon were measured for 240 min following a mashed potato meal (1808 kJ). RESULTS There was no difference in GE between sitgaliptin and placebo [50% emptying time (T50): P = 0.4]. Mean blood glucose was slightly less (P = 0.02) on sitagliptin. Sitagliptin reduced plasma glucagon between 75 and 120 min (P < 0.05), and increased intact GLP-1 (P = 0.0002) and intact GIP (P = 0.0001) by approximately twofold, but reduced total GIP (P = 0.0003) and had no effect on total GLP-1 (P = 0.16) or insulin (P = 0.75). On sitagliptin the initial rise in blood glucose (r = -0.66, P = 0.008) and the intact GIP response (r = -0.66, P = 0.007) were inversely related, whereas the intact GLP-1 response was related directly (r = 0.52, P = 0.05) to the T50. CONCLUSIONS While the effects of sitagliptin on glycaemic control are unlikely to relate to slowing of GE in healthy humans, the rate of GE is a significant determinant of postprandial glycaemia on sitagliptin.
Collapse
Affiliation(s)
- J E Stevens
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | | | | | | | | | | |
Collapse
|
40
|
Effect of the GLP-1 analog liraglutide on satiation and gastric sensorimotor function during nutrient-drink ingestion. Int J Obes (Lond) 2012; 37:693-8. [PMID: 22846777 DOI: 10.1038/ijo.2012.101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND/AIM Liraglutide, a glucagon-like peptide-1 analog, induces weight loss. We investigated whether liraglutide affects gastric accommodation and satiation by measuring the intragastric pressure (IGP) during nutrient-drink consumption and using the barostat technique. METHODS Ten healthy volunteers (HVs) were tested after placebo, 0.3, 0.6 or 1.2 mg liraglutide administration. IGP was studied during intragastric nutrient-drink (1.5 kcal ml(-1)) infusion (60 ml min(-1)), while the HVs scored their satiation on a graded scale until maximal satiation. In a separate session, isobaric distentions were performed using the barostat with stepwise increments of 2 mm Hg starting from minimal distending pressure, although HVs scored their perception; gastric volume was monitored 30 min before and until 60 min after ingestion of 200 ml of nutrient drink. Data are presented as mean±s.e.m. comparisons were performed with ANOVA (P<0.05 was significant). RESULTS During nutrient-drink infusion, IGP decreased with 4.1±0.7, 3.0±0.4, 2.1±0.3 and 2.6±0.4 mm Hg (placebo, 0.3, 0.6 and 1.2 mg liraglutide, respectively; P<0.05). The maximum-tolerated volume was not different, except after treatment with 1.2 mg liraglutide (695±135 ml) compared with placebo (1008±197 ml; P<0.05); however, 1.2 mg liraglutide induced nausea in all volunteers. In the barostat study, liraglutide did not affect the perception or compliance, but significantly decreased gastric accommodation to the meal (168±27 vs 78.8±36.4 ml after treatment with placebo and 0.6 mg liraglutide, respectively; P<0.05). CONCLUSION Although no effect on perception, compliance or satiation was observed, liraglutide inhibited gastric accommodation. Whether this effect is involved in the anorectic effect of liraglutide remains to be determined.
Collapse
|
41
|
Witte AB, Grybäck P, Jacobsson H, Näslund E, Hellström PM, Holst JJ, Hilsted L, Schmidt PT. Involvement of endogenous glucagon-like peptide-1 in regulation of gastric motility and pancreatic endocrine secretion. Scand J Gastroenterol 2011; 46:428-35. [PMID: 21114428 DOI: 10.3109/00365521.2010.537680] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To study the role of endogenous glucagon-like peptide-1 (GLP-1) on gastric emptying rates of a solid meal as well as postprandial hormone secretion and glucose disposal. MATERIAL AND METHODS In nine healthy subjects, gastric emptying of a 310-kcal radio-labelled solid meal and plasma concentrations of insulin, glucagon and glucose were measured during infusion of saline or the GLP-1 receptor antagonist exendin(9-39)amide (Ex(9-39)) at 300 pmol·kg(-1)·min(-1). RESULTS Ex(9-39) infusion had no effect on the total gastric emptying curve, but changed the intra-gastric distribution of the meal. During infusion of Ex(9-39), more content stayed in the upper stomach (79.1 ± 2.5% of total during Ex(9-39) compared to 66.6 ± 5.7% during saline at 5 min). During Ex(9-39) infusion, higher concentrations of plasma glucagon were measured both before (after 40 min of Ex(9-39) infusion the glucagon level was 15.1 ± 0.7 pmol·L(-1) compared to 5.4 ± 1.4 during saline) and after the meal, and postprandial GLP-1 levels increased. Basal insulin and glucose levels were not affected by Ex(9-39), but the postprandial rise of insulin and glucose enhanced during Ex(9-39). CONCLUSIONS Endogenous GLP-1 is involved in the regulation of gastric motility in relation to meal intake and also in the regulation of postprandial insulin and glucose levels. Furthermore, endogenous GLP-1 seems to tonically restrain glucagon secretion.
Collapse
Affiliation(s)
- Anne-Barbara Witte
- Department of Gastroenterology and Hepatology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
42
|
The once-daily human glucagon-like peptide-1 (GLP-1) analog liraglutide improves postprandial glucose levels in type 2 diabetes patients. Adv Ther 2011; 28:213-26. [PMID: 21340616 DOI: 10.1007/s12325-010-0110-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Fasting and postprandial plasma glucose (FPG, PPG) control are both necessary to achieve glycosylated hemoglobin (HbA(1c)) regulation goals. Liraglutide, based on its glucagon-like peptide 1 (GLP-1)-mediated pharmacology and pharmacokinetics may reduce HbA(1c) through both FPG and PPG levels. The objective of the present study was to investigate the effect of once-daily liraglutide (0.6, 1.2, and 1.8 mg) at steady state on FPG, PPG, postprandial insulin, and gastric emptying. METHODS Eighteen subjects with type 2 diabetes, aged 18-70 years, with a body mass index of 18.5-40 kg/m(2) and HbA(1c) of 7.0%-9.5% were included in this single-centre, randomized, placebo-controlled, double-blind, two-period, cross over trial. Patients were randomized into two groups (A or B). Group A received oncedaily liraglutide for 3 weeks, followed by a 3-4-week washout period and 3 weeks of oncedaily placebo. Group B was treated as for Group A, but treatment periods were reversed (ie, placebo followed by liraglutide). A meal test was performed at steady-state liraglutide/placebo doses of 0.6, 1.2, and 1.8 mg/day. Plasma glucose, insulin, and paracetamol (acetaminophen) concentrations (to assess gastric emptying) were measured pre- and postmeal. RESULTS PPG levels significantly decreased (P<0.001) after all three liraglutide doses when compared with placebo. This decrease was also apparent when corrected for baseline (incremental excursions), with the exception of average incremental increase calculated as area under the concentration curve (AUC) over the fasting value from time zero to 5 hours (iAUC (0-5 h)/5 hours) after liraglutide 0.6 mg, where there was a trend to decrease (P=0.082). In addition, FPG levels significantly decreased at all three liraglutide dose levels when compared to placebo (P<0.001). Fasting and postprandial insulin levels significantly increased with liraglutide versus placebo at all doses studied (P<0.001). A significant delay in gastric emptying during the first hour postmeal was observed at the two highest liraglutide doses versus placebo. CONCLUSION In addition to lowering FPG levels, liraglutide improves PPG levels (absolute and incremental) possibly by both stimulating postprandial insulin secretion and delaying gastric emptying.
Collapse
|
43
|
Rotondo A, Amato A, Lentini L, Baldassano S, Mulè F. Glucagon-like peptide-1 relaxes gastric antrum through nitric oxide in mice. Peptides 2011; 32:60-4. [PMID: 20933560 DOI: 10.1016/j.peptides.2010.09.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/30/2010] [Accepted: 09/30/2010] [Indexed: 02/07/2023]
Abstract
Glucagon-like-peptide-1 (GLP-1) is a proglucagon-derived peptide expressed in the intestinal enteroendocrine-L cells and released after meal ingestion. GLP-1 reduces postprandial glycemia not only by its hormonal effects, but also by its inhibitory effects on gastrointestinal motility. Recently, we showed that GLP-1 acts in the enteric nervous system of mouse intestine. Therefore our working hypothesis was that GLP-1 may have also a direct influence on the gastric mechanical activity since the major part of experimental studies about its involvement in the regulation of gastric motility have been conducted in in vivo conditions. The purposes of this study were (i) to examine exogenous GLP-1 effects on mouse gastric mechanical activity using isolated whole stomach; (ii) to clarify the regional activity of GLP-1 using circular muscular strips from gastric fundus or antrum; (iii) to analyze the mechanism of action underlying the observed effects; (iv) to verify regional differences of GLP-1 receptors (GLP-1R) expression by RT-PCR. In the whole stomach GLP-1 caused concentration-dependent relaxation significantly anatagonized by exendin (9-39), an antagonist of GLP-1R and abolished by tetrodotoxin (TTX) or N(ω)-nitro-l-arginine methyl ester (l-NAME), inhibitor of nitric oxide (NO) synthase. GLP-1 was without any effect in fundic strips, but it induced concentration-dependent relaxation in carbachol-precontracted antral strips. The effect was abolished by TTX or l-NAME. RT-PCR analysis revealed a higher expression of GLP-1R mRNA in antrum than in fundus. These results suggest that exogenous GLP-1 is able to reduce mouse gastric motility by acting peripherally in the antral region, through neural NO release.
Collapse
Affiliation(s)
- Alessandra Rotondo
- Dipartimento di Biologia cellulare e dello Sviluppo, Università di Palermo, 90128 Palermo, Italy
| | | | | | | | | |
Collapse
|
44
|
Deane AM, Summers MJ, Zaknic AV, Chapman MJ, Fraser RJL, Di Bartolomeo AE, Wishart JM, Horowitz M. Exogenous glucagon-like peptide-1 attenuates the glycaemic response to postpyloric nutrient infusion in critically ill patients with type-2 diabetes. Crit Care 2011; 15:R35. [PMID: 21255422 PMCID: PMC3222072 DOI: 10.1186/cc9983] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 12/14/2010] [Accepted: 01/21/2011] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Glucagon-like peptide-1 (GLP-1) attenuates the glycaemic response to small intestinal nutrient infusion in stress-induced hyperglycaemia and reduces fasting glucose concentrations in critically ill patients with type-2 diabetes. The objective of this study was to evaluate the effects of acute administration of GLP-1 on the glycaemic response to small intestinal nutrient infusion in critically ill patients with pre-existing type-2 diabetes. METHODS Eleven critically ill mechanically-ventilated patients with known type-2 diabetes received intravenous infusions of GLP-1 (1.2 pmol/kg/minute) and placebo from t = 0 to 270 minutes on separate days in randomised double-blind fashion. Between t = 30 to 270 minutes a liquid nutrient was infused intraduodenally at a rate of 1 kcal/min via a naso-enteric catheter. Blood glucose, serum insulin and C-peptide, and plasma glucagon were measured. Data are mean ± SEM. RESULTS GLP-1 attenuated the overall glycaemic response to nutrient (blood glucose AUC30-270 min: GLP-1 2,244 ± 184 vs. placebo 2,679 ± 233 mmol/l/minute; P = 0.02). Blood glucose was maintained at < 10 mmol/l in 6/11 patients when receiving GLP-1 and 4/11 with placebo. GLP-1 increased serum insulin at 270 minutes (GLP-1: 23.4 ± 6.7 vs. placebo: 16.4 ± 5.5 mU/l; P < 0.05), but had no effect on the change in plasma glucagon. CONCLUSIONS Exogenous GLP-1 in a dose of 1.2 pmol/kg/minute attenuates the glycaemic response to small intestinal nutrient in critically ill patients with type-2 diabetes. Given the modest magnitude of the reduction in glycaemia the effects of GLP-1 at higher doses and/or when administered in combination with insulin, warrant evaluation in this group. TRIAL REGISTRATION ANZCTR:ACTRN12610000185066.
Collapse
Affiliation(s)
- Adam M Deane
- Discipline of Acute Care Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, 5000, Australia
- Intensive Care Unit, Level 4, Emergency Services Building, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, 5000, Australia
- National Health and Medical Research Council of Australia Centre for Clinical Research Excellence in Nutritional Physiology and Outcomes, Level 6, Eleanor Harrald Building, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Matthew J Summers
- Intensive Care Unit, Level 4, Emergency Services Building, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Antony V Zaknic
- Intensive Care Unit, Level 4, Emergency Services Building, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Marianne J Chapman
- Discipline of Acute Care Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, 5000, Australia
- Intensive Care Unit, Level 4, Emergency Services Building, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, 5000, Australia
- National Health and Medical Research Council of Australia Centre for Clinical Research Excellence in Nutritional Physiology and Outcomes, Level 6, Eleanor Harrald Building, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Robert JL Fraser
- National Health and Medical Research Council of Australia Centre for Clinical Research Excellence in Nutritional Physiology and Outcomes, Level 6, Eleanor Harrald Building, North Terrace, Adelaide, South Australia, 5000, Australia
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Level 6 Eleanor Harrald Building, North Terrace, Adelaide, South Australia, 5000, Australia
- Investigation and Procedures Unit, Repatriation General Hospital, Daws Road, Daw Park, South Australia, 5041, Australia
| | - Anna E Di Bartolomeo
- Discipline of Acute Care Medicine, University of Adelaide, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Judith M Wishart
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Level 6 Eleanor Harrald Building, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Michael Horowitz
- Discipline of Medicine, University of Adelaide, Royal Adelaide Hospital, Level 6 Eleanor Harrald Building, North Terrace, Adelaide, South Australia, 5000, Australia
| |
Collapse
|
45
|
Marathe CS, Rayner CK, Jones KL, Horowitz M. Effects of GLP-1 and incretin-based therapies on gastrointestinal motor function. EXPERIMENTAL DIABETES RESEARCH 2011; 2011:279530. [PMID: 21747825 PMCID: PMC3124003 DOI: 10.1155/2011/279530] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 04/04/2011] [Accepted: 04/19/2011] [Indexed: 02/07/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) is a hormone secreted predominantly by the distal small intestine and colon and released in response to enteral nutrient exposure. GLP-1-based therapies are now used widely in the management of type 2 diabetes and have the potential to be effective antiobesity agents. Although widely known as an incretin hormone, there is a growing body of evidence that GLP-1 also acts as an enterogastrone, with profound effects on the gastrointestinal motor system. Moreover, the effects of GLP-1 on gastrointestinal motility appear to be pivotal to its effect of reducing postprandial glycaemic excursions and may, potentially, represent the dominant mechanism. This review summarizes current knowledge of the enterogastrone properties of GLP-1, focusing on its effects on gut motility at physiological and pharmacological concentrations, and the motor actions of incretin-based therapies. While of potential importance, the inhibitory action of GLP-1 on gastric acid secretion is beyond the scope of this paper.
Collapse
Affiliation(s)
- Chinmay S. Marathe
- 1Discipline of Medicine, Royal Adelaide Hospital, University of Adelaide, Adelaide SA 5000, Australia
- 2Centre of Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide SA 5000, Australia
| | - Christopher K. Rayner
- 1Discipline of Medicine, Royal Adelaide Hospital, University of Adelaide, Adelaide SA 5000, Australia
- 2Centre of Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide SA 5000, Australia
- *Christopher K. Rayner:
| | - Karen L. Jones
- 1Discipline of Medicine, Royal Adelaide Hospital, University of Adelaide, Adelaide SA 5000, Australia
- 2Centre of Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide SA 5000, Australia
| | - Michael Horowitz
- 1Discipline of Medicine, Royal Adelaide Hospital, University of Adelaide, Adelaide SA 5000, Australia
- 2Centre of Clinical Research Excellence in Nutritional Physiology, Interventions and Outcomes, University of Adelaide, Adelaide SA 5000, Australia
| |
Collapse
|
46
|
Abstract
Symptoms suggestive of gastroparesis occur in 5% to 12% of patients with diabetes. Such a complication can affect both prognosis and management of the diabetes; therefore, practicing clinicians are challenged by the complex management of such cases. Gastroparesis is a disorder characterized by a delay in gastric emptying after a meal in the absence of a mechanical gastric outlet obstruction. This article is an evidence-based overview of current management strategies for diabetic gastroparesis. The cardinal symptoms of diabetic gastroparesis are nausea and vomiting. Gastroesophageal scintiscanning at 15-minute intervals for 4 hours after food intake is considered the gold standard for measuring gastric emptying. Retention of more than 10% of the meal after 4 hours is considered an abnormal result, for which a multidisciplinary management approach is required. Treatment should be tailored according to the severity of gastroparesis, and 25% to 68% of symptoms are controlled by prokinetic agents. Commonly prescribed prokinetics include metoclopramide, domperidone, and erythromycin. In addition, gastric electrical stimulation has been shown to improve symptoms, reduce hospitalizations, reduce the need for nutritional support, and improve quality of life in several open-label studies.
Collapse
Affiliation(s)
- Badr M. Aljarallah
- Department of Medicine, Gastroenterology Division, King Fahad Specialist Hospital, Faculty of Medicine, Qassim University, Maledia, Saudi Arabia,Address for correspondence: Dr. Badr M. Aljarallah, Department of Medicine, Gastroenterology Division, King Fahad Specialist Hospital, Faculty of Medicine, Qassim University, Maledia - 51452, Saudi Arabia. E-mail:
| |
Collapse
|
47
|
You CZ, Dong R, Sun JJ, Xiao JQ, Qu HC, Du MH, Huang HQ, Tang WH. Pyloric resection and delayed gastric liquid emptying in rats. Dig Dis Sci 2011; 56:49-58. [PMID: 20431948 DOI: 10.1007/s10620-010-1238-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 04/06/2010] [Indexed: 01/19/2023]
Abstract
BACKGROUND Surgical resection of the distal stomach impairs gastric emptying. Generally, pylorus and the antrum are removed in the distal gastrectomy, however, the pylorus is removed individually under specific circumstances. We focus on the relation between the pyloric resection and the gastric liquid emptying. AIMS The present investigation aimed to explore the pylorectomy how to influence gastric liquid emptying in rats. METHODS Pylorectomy and end-to-end gastroduodenal anastomosis were conducted in rats. Electrodes were implanted in the gastrointestinal serosal surface near the stoma. Total stomach, proximal stomach, distal stomach and duodenal liquid emptying, myoelectricities in the gastrointestinal tract near the stoma, and structures were examined with scintigraphy, electrode recording in vivo, and electron microscopy, respectively. RESULTS Delayed total stomach and distal stomach emptying were found in pylorectomy rats (p<0.001). However, there was no difference in the proximal stomach and the duodenal liquid emptying compared to the controls (p>0.05). The myoelectricity of 3-5 cpm (cycles/min) in antrum and 10-12 cpm in duodenum were found in the controls and no retrograde or antegrade myoelectricities were recorded in the duodenum and antrum. High-frequency myoelectricities (tachygastria) were recorded in the antrum near the stoma (p<0.01), the retrograde and antegrade myoelectricities propagating through the stoma were recorded, and the regenerated interstitial cells of Cajal were found in stoma under electron microscope observation in pylorectomy rat. CONCLUSIONS The gastroduodenal incoordination and abnormal myoelectricity related to impaired contraction in the antrum caused the delayed liquid gastric emptying in pylorectomy rats.
Collapse
Affiliation(s)
- Cheng-Zhong You
- Department of General Surgery, Zhong-Da Hospital, and Department of Physiology, Southeast University School of Medicine, 210009, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kuo P, Bellon M, Wishart J, Smout AJ, Holloway RH, Fraser RJL, Horowitz M, Jones KL, Rayner CK. Effects of metoclopramide on duodenal motility and flow events, glucose absorption, and incretin hormone release in response to intraduodenal glucose infusion. Am J Physiol Gastrointest Liver Physiol 2010; 299:G1326-33. [PMID: 20829521 DOI: 10.1152/ajpgi.00476.2009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The contribution of small intestinal motor activity to nutrient absorption is poorly defined. A reduction in duodenal flow events after hyoscine butylbromide, despite no change in pressure waves, was associated with reduced secretion of the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) and a delay in glucose absorption. The aim of this study was to investigate the effect of metoclopramide on duodenal motility and flow events, incretin hormone secretion, and glucose absorption. Eight healthy volunteers (7 males and 1 female; age 29.8 ± 4.6 yr; body mass index 24.5 ± 0.9 kg/m²) were studied two times in randomized order. A combined manometry and impedance catheter was used to measure pressure waves and flow events in the same region of the duodenum simultaneously. Metoclopramide (10 mg) or control was administered intravenously as a bolus, followed by an intraduodenal glucose infusion for 60 min (3 kcal/min) incorporating the ¹⁴C-labeled glucose analog 3-O-methylglucose (3-OMG). We found that metoclopramide was associated with more duodenal pressure waves and propagated pressure sequences than control (P < 0.05 for both) during intraduodenal glucose infusion. However, the number of duodenal flow events, blood glucose concentration, and plasma 3-[¹⁴C]OMG activity did not differ between the two study days. Metoclopramide was associated with increased plasma concentrations of GLP-1 (P < 0.05) and GIP (P = 0.07) but lower plasma insulin concentrations (P < 0.05). We concluded that metoclopramide was associated with increased frequency of duodenal pressure waves but no change in duodenal flow events and glucose absorption. Furthermore, GLP-1 and GIP release increased with metoclopramide, but insulin release paradoxically decreased.
Collapse
Affiliation(s)
- Paul Kuo
- Royal Adelaide Hospital, University of Adelaide, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Quantification of the effects of the volume and viscosity of gastric contents on antral and fundic activity in the rat stomach maintained ex vivo. Dig Dis Sci 2010; 55:3349-60. [PMID: 20198425 DOI: 10.1007/s10620-010-1164-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 02/11/2010] [Indexed: 12/13/2022]
Abstract
AIMS The aim of this study was to examine the effect of varying the rheological properties of perfusate on the volume and muscular activity of the various compartments of the rat stomach. METHODS Image analysis was used to quantify the activity of the ex vivo stomach preparations when perfused according to a ramp profile. RESULTS The area of the fundus increased to a greater extent than that of the body when watery or viscous material was perfused. However, initial distension of the corpus was greater and occurred more rapidly when viscous material was perfused. Only the fundus expanded when perfusion followed the administration of verapamil. The frequency of antrocorporal contractions decreased significantly and the amplitude of antrocorporal contractions increased significantly with increase in gastric volume. The velocity of antrocorporal contractions did not vary with gastric volume but varied regionally in some preparations being faster distally than proximally. Neither the frequency, amplitude or velocity of antrocorporal contractions differed when pseudoplastic rather than watery fluid was perfused. However, the characteristics of antrocorporal contractions changed significantly when the stomach was perfused with material with rheological characteristics that induce different patterns of wall tension to those normally encountered. Hence, the mean frequency and speed of propagation of antrocorporal contractions increased and their direction of propagation became inconstant.
Collapse
|
50
|
Tatarkiewicz K, Smith PA, Sablan EJ, Polizzi CJ, Aumann DE, Villescaz C, Hargrove DM, Gedulin BR, Lu MGW, Adams L, Whisenant T, Roy D, Parkes DG. Exenatide does not evoke pancreatitis and attenuates chemically induced pancreatitis in normal and diabetic rodents. Am J Physiol Endocrinol Metab 2010; 299:E1076-86. [PMID: 20923958 PMCID: PMC3006257 DOI: 10.1152/ajpendo.00479.2010] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The risk of developing pancreatitis is elevated in type 2 diabetes and obesity. Cases of pancreatitis have been reported in type 2 diabetes patients treated with GLP-1 (GLP-1R) receptor agonists. To examine whether the GLP-1R agonist exenatide potentially induces or modulates pancreatitis, the effect of exenatide was evaluated in normal or diabetic rodents. Normal and diabetic rats received a single exenatide dose (0.072, 0.24, and 0.72 nmol/kg) or vehicle. Diabetic ob/ob or HF-STZ mice were infused with exenatide (1.2 and 7.2 nmol·kg(-1)·day(-1)) or vehicle for 4 wk. Post-exenatide treatment, pancreatitis was induced with caerulein (CRN) or sodium taurocholate (ST), and changes in plasma amylase and lipase were measured. In ob/ob mice, plasma cytokines (IL-1β, IL-2, IL-6, MCP-1, IFNγ, and TNFα) and pancreatitis-associated genes were assessed. Pancreata were weighed and examined histologically. Exenatide treatment alone did not modify plasma amylase or lipase in any models tested. Exenatide attenuated CRN-induced release of amylase and lipase in normal rats and ob/ob mice but did not modify the response to ST infusion. Plasma cytokines and pancreatic weight were unaffected by exenatide. Exenatide upregulated Reg3b but not Il6, Ccl2, Nfkb1, or Vamp8 expression. Histological analysis revealed that the highest doses of exenatide decreased CRN- or ST-induced acute inflammation, vacuolation, and acinar single cell necrosis in mice and rats, respectively. Ductal cell proliferation rates were low and similar across all groups of ob/ob mice. In conclusion, exenatide did not modify plasma amylase and lipase concentrations in rodents without pancreatitis and improved chemically induced pancreatitis in normal and diabetic rodents.
Collapse
|