1
|
Müller TD, Adriaenssens A, Ahrén B, Blüher M, Birkenfeld AL, Campbell JE, Coghlan MP, D'Alessio D, Deacon CF, DelPrato S, Douros JD, Drucker DJ, Figueredo Burgos NS, Flatt PR, Finan B, Gimeno RE, Gribble FM, Hayes MR, Hölscher C, Holst JJ, Knerr PJ, Knop FK, Kusminski CM, Liskiewicz A, Mabilleau G, Mowery SA, Nauck MA, Novikoff A, Reimann F, Roberts AG, Rosenkilde MM, Samms RJ, Scherer PE, Seeley RJ, Sloop KW, Wolfrum C, Wootten D, DiMarchi RD, Tschöp MH. Glucose-dependent insulinotropic polypeptide (GIP). Mol Metab 2025; 95:102118. [PMID: 40024571 PMCID: PMC11931254 DOI: 10.1016/j.molmet.2025.102118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glucose-dependent insulinotropic polypeptide (GIP) was the first incretin identified and plays an essential role in the maintenance of glucose tolerance in healthy humans. Until recently GIP had not been developed as a therapeutic and thus has been overshadowed by the other incretin, glucagon-like peptide 1 (GLP-1), which is the basis for several successful drugs to treat diabetes and obesity. However, there has been a rekindling of interest in GIP biology in recent years, in great part due to pharmacology demonstrating that both GIPR agonism and antagonism may be beneficial in treating obesity and diabetes. This apparent paradox has reinvigorated the field, led to new lines of investigation, and deeper understanding of GIP. SCOPE OF REVIEW In this review, we provide a detailed overview on the multifaceted nature of GIP biology and discuss the therapeutic implications of GIPR signal modification on various diseases. MAJOR CONCLUSIONS Following its classification as an incretin hormone, GIP has emerged as a pleiotropic hormone with a variety of metabolic effects outside the endocrine pancreas. The numerous beneficial effects of GIPR signal modification render the peptide an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, drug-induced nausea and both bone and neurodegenerative disorders.
Collapse
Affiliation(s)
- Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Walther-Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University Munich (LMU), Germany.
| | - Alice Adriaenssens
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Bo Ahrén
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen 72076, Germany; Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich, Tübingen, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA; Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Matthew P Coghlan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - David D'Alessio
- Department of Medicine, Division of Endocrinology, Duke University, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Carolyn F Deacon
- School of Biomedical Sciences, Ulster University, Coleraine, UK; Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefano DelPrato
- Interdisciplinary Research Center "Health Science", Sant'Anna School of Advanced Studies, Pisa, Italy
| | | | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Natalie S Figueredo Burgos
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Brian Finan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Ruth E Gimeno
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Fiona M Gribble
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Matthew R Hayes
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christian Hölscher
- Neurodegeneration Research Group, Henan Academy of Innovations in Medical Science, Xinzheng, China
| | - Jens J Holst
- Department of Biomedical Sciences and the Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Patrick J Knerr
- Indianapolis Biosciences Research Institute, Indianapolis, IN, USA
| | - Filip K Knop
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christine M Kusminski
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany; Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Guillaume Mabilleau
- Univ Angers, Nantes Université, ONIRIS, Inserm, RMeS UMR 1229, Angers, France; CHU Angers, Departement de Pathologie Cellulaire et Tissulaire, Angers, France
| | | | - Michael A Nauck
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Munich, Germany; German Center for Diabetes Research, DZD, Germany
| | - Frank Reimann
- Institute of Metabolic Science-Metabolic Research Laboratories & MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Anna G Roberts
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen, Copenhagen, Denmark
| | - Ricardo J Samms
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Philip E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kyle W Sloop
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092, Schwerzenbach, Switzerland
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia; ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | | | - Matthias H Tschöp
- Helmholtz Munich, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Liu X, Liu X, Hu Y, Wang X, Yang X, Yan B, Zhou Y, Zhou L, Fan G, Yang J. Secretagogin Is Highly Expressed in Enteroendocrine K Cells and Plays a Critical Role in Nutrient-Induced GIP Secretion. J Endocr Soc 2025; 9:bvaf022. [PMID: 40012909 PMCID: PMC11859952 DOI: 10.1210/jendso/bvaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Indexed: 02/28/2025] Open
Abstract
Context Incretin hormones, primarily composed of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), are secreted by enteroendocrine cells (EECs) and play crucial roles in maintaining blood glucose homeostasis. Notably, GIP accounts for two-thirds of the entire incretin effect. However, the secretion and function of GIP are impaired in individuals with type 2 diabetes mellitus (T2DM), and the regulatory mechanisms governing GIP secretion remain unclear. Objective Our study aims to explore the role of an EEC-enriched protein, Secretagogin (SCGN), in the regulation of GIP secretion. Methods We collected duodenal tissues from both humans and mice to observe the colocalization of SCGN and GIP in EECs. Additionally, we utilized human cohorts and gene-edited mouse models to investigate the effect of SCGN on GIP secretion. Our study included 128 subjects, comprising 64 individuals diagnosed with newly onset diabetes and 64 age- and sex-matched nondiabetic healthy controls. At the animal level, we employed leptin receptor-deficient (db/db) mice and Scgn knockout mice for our investigations. Results Our findings indicate that SCGN is abundantly expressed in GIP-producing K cells within the intestinal epithelium of both humans and mice. We observed a positive correlation between SCGN and GIP levels in postprandial states among patients with T2DM, db/db mice, and their healthy controls. Notably, Scgn knockout mice exhibited decreased GIP and insulin secretion. However, SCGN deficiency did not affect K-cell number, GIP mRNA expression, or intestinal morphology. Conclusion Collectively, these findings demonstrate that SCGN is a key regulator of nutrient-induced GIP secretion.
Collapse
Affiliation(s)
- Xinyu Liu
- Department of Endocrinology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen Nanshan People's Hospital, Shenzhen 518052, Guangdong, China
| | - Xuan Liu
- Department of Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- Department of Endocrinology, The Central Hospital of Shaoyang City, Shaoyang 422000, Hunan, China
| | - Yuanyuan Hu
- Department of Endocrinology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen Nanshan People's Hospital, Shenzhen 518052, Guangdong, China
| | - Xin Wang
- Department of Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Xin Yang
- Department of Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Bin Yan
- Department of Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yiting Zhou
- Department of Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lingzhi Zhou
- Department of Pediatrics, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen Nanshan People's Hospital, Shenzhen 518052, Guangdong, China
| | - Gang Fan
- Department of Urology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen Nanshan People's Hospital, Shenzhen 518052, Guangdong, China
| | - Jing Yang
- Department of Endocrinology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen Nanshan People's Hospital, Shenzhen 518052, Guangdong, China
- Department of Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
3
|
Usui R, Hamamoto Y, Imura M, Omori Y, Yamazaki Y, Kuwata H, Tatsuoka H, Shimomura K, Murotani K, Yamada Y, Seino Y. Differential effects of imeglimin and metformin on insulin and incretin secretion-An exploratory randomized controlled trial. Diabetes Obes Metab 2025; 27:856-865. [PMID: 39592886 PMCID: PMC11701202 DOI: 10.1111/dom.16086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024]
Abstract
AIMS Imeglimin is a new oral anti-diabetic drug with a similar structure to that of metformin; however, unlike metformin, clinical trials indicate that imeglimin elicits its glucose-lowering effect mainly by enhancement of insulin secretion. The comparative effects of the two drugs on incretin secretion remains to be elucidated. MATERIALS AND METHODS A single-center, open-label, randomized controlled trial was conducted in patients with type 2 diabetes who were drug-naïve or were on a single oral hypoglycaemic agent (OHA). For patients taking a single OHA, an 8-week washout period was employed before randomization. Participants were randomized to the imeglimin group (IME, 2000 mg/day) or the metformin group (MET, 1000 mg/day), and OGTT was performed before treatment and after 12 and 24 weeks of treatment. RESULTS The reduction in HbA1c at 24 weeks was similar in IME and MET. OGTT revealed a comparable decrease in post-challenge blood glucose excursion in both groups, but insulin levels were increased only in IME. Total and active glucagon-like peptide-1 (GLP-1) levels were increased in both IME and MET; however, total and active glucose-dependent insulinotropic peptide (GIP) levels were increased only in IME. Interestingly, while an increase in insulin levels in IME was positively correlated with an increase in GLP-1 at 12 weeks, it was correlated only with an increase in GIP at 24 weeks. CONCLUSIONS Unlike metformin, imeglimin enhances GIP secretion as well as GLP-1 secretion, in addition to its direct insulinotropic mechanism of glucose control, emphasizing its potential as a therapeutic option in the treatment of patients with diabetes.
Collapse
Affiliation(s)
- Ryota Usui
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Yoshiyuki Hamamoto
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Masahiro Imura
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Yasuhiro Omori
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Yuji Yamazaki
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Hitoshi Kuwata
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Hisato Tatsuoka
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
- Research and Development departmentGeneral Incorporated Association Kansai Healthcare Science InformaticsKyotoJapan
| | | | | | - Yuichiro Yamada
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Yutaka Seino
- Center for Diabetes, Endocrinology and MetabolismKansai Electric Power HospitalOsakaJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| |
Collapse
|
4
|
Shen C, Xie H, Jiang X, Wang L. A physiologically-based quantitative systems pharmacology model for mechanistic understanding of the response to alogliptin and its application in patients with renal impairment. J Pharmacokinet Pharmacodyn 2025; 52:13. [PMID: 39821812 DOI: 10.1007/s10928-025-09961-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025]
Abstract
Alogliptin is a highly selective inhibitor of dipeptidyl peptidase-4 and primarily excreted as unchanged drug in the urine, and differences in clinical outcomes in renal impairment patients increase the risk of serious adverse reactions. In this study, we developed a comprehensive physiologically-based quantitative systematic pharmacology model of the alogliptin-glucose control system to predict plasma exposure and use glucose as a clinical endpoint to prospectively understand its therapeutic outcomes with varying renal function. Our model incorporates a PBPK model for alogliptin, DPP-4 activity described by receptor occupancy theory, and the crosstalk and feedback loops for GLP-1-GIP-glucagon, insulin, and glucose. Based on the optimization of renal function-dependent parameters, the model was extrapolated to different stages renal impairment patients. Ultimately our model adequately describes the pharmacokinetics of alogliptin, the progression of DPP-4 inhibition over time and the dynamics of the glucose control system components. The extrapolation results endorse the dose adjustment regimen of 12.5 mg once daily for moderate patients and 6.25 mg once daily for severe and ESRD patients, while providing additional reflections and insights. In clinical practice, our model could provide additional information on the in vivo fate of DPP4 inhibitors and key regulators of the glucose control system.
Collapse
Affiliation(s)
- Chaozhuang Shen
- Department of Clinical Pharmacy and Pharmacy Administration, West China school of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Haitang Xie
- Anhui Provincial Center for Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, 241001, China
| | - Xuehua Jiang
- Department of Clinical Pharmacy and Pharmacy Administration, West China school of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Ling Wang
- Department of Clinical Pharmacy and Pharmacy Administration, West China school of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
5
|
Ungvari Z, Kunutsor SK. Coffee consumption and cardiometabolic health: a comprehensive review of the evidence. GeroScience 2024; 46:6473-6510. [PMID: 38963648 PMCID: PMC11493900 DOI: 10.1007/s11357-024-01262-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
This review provides a comprehensive synthesis of longitudinal observational and interventional studies on the cardiometabolic effects of coffee consumption. It explores biological mechanisms, and clinical and policy implications, and highlights gaps in the evidence while suggesting future research directions. It also reviews evidence on the causal relationships between coffee consumption and cardiometabolic outcomes from Mendelian randomization (MR) studies. Findings indicate that while coffee may cause short-term increases in blood pressure, it does not contribute to long-term hypertension risk. There is limited evidence indicating that coffee intake might reduce the risk of metabolic syndrome and non-alcoholic fatty liver disease. Furthermore, coffee consumption is consistently linked with reduced risks of type 2 diabetes (T2D) and chronic kidney disease (CKD), showing dose-response relationships. The relationship between coffee and cardiovascular disease is complex, showing potential stroke prevention benefits but ambiguous effects on coronary heart disease. Moderate coffee consumption, typically ranging from 1 to 5 cups per day, is linked to a reduced risk of heart failure, while its impact on atrial fibrillation remains inconclusive. Furthermore, coffee consumption is associated with a lower risk of all-cause mortality, following a U-shaped pattern, with the largest risk reduction observed at moderate consumption levels. Except for T2D and CKD, MR studies do not robustly support a causal link between coffee consumption and adverse cardiometabolic outcomes. The potential beneficial effects of coffee on cardiometabolic health are consistent across age, sex, geographical regions, and coffee subtypes and are multi-dimensional, involving antioxidative, anti-inflammatory, lipid-modulating, insulin-sensitizing, and thermogenic effects. Based on its beneficial effects on cardiometabolic health and fundamental biological processes involved in aging, moderate coffee consumption has the potential to contribute to extending the healthspan and increasing longevity. The findings underscore the need for future research to understand the underlying mechanisms and refine health recommendations regarding coffee consumption.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Setor K Kunutsor
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester General Hospital, Gwendolen Road, Leicester, LE5 4WP, UK.
- Section of Cardiology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Saint Boniface Hospital, Winnipeg, MB, R2H 2A6, Canada.
| |
Collapse
|
6
|
Lewandowski SL, El K, Campbell JE. Evaluating glucose-dependent insulinotropic polypeptide and glucagon as key regulators of insulin secretion in the pancreatic islet. Am J Physiol Endocrinol Metab 2024; 327:E103-E110. [PMID: 38775725 PMCID: PMC11390117 DOI: 10.1152/ajpendo.00360.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/27/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024]
Abstract
The incretin axis is an essential component of postprandial insulin secretion and glucose homeostasis. There are two incretin hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), which exert multiple actions throughout the body. A key cellular target for the incretins are pancreatic β-cells, where they potentiate nutrient-stimulated insulin secretion. This feature of incretins has made this system an attractive target for therapeutic interventions aimed at controlling glycemia. Here, we discuss the role of GIP in both β-cells and α-cells within the islet, to stimulate insulin and glucagon secretion, respectively. Moreover, we discuss how glucagon secretion from α-cells has important insulinotropic actions in β-cells through an axis termed α- to β-cell communication. These recent advances have elevated the potential of GIP and glucagon as a therapeutic targets, coinciding with emerging compounds that pharmacologically leverage the actions of these two peptides in the context of diabetes and obesity.
Collapse
Affiliation(s)
- Sophie L Lewandowski
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States
| | - Kimberley El
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, United States
- Division of Endocrinology, Department of Medicine, Duke University, Durham, North Carolina, United States
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, United States
| |
Collapse
|
7
|
Helsted MM, Schaltz NL, Gasbjerg LS, Christensen MB, Vilsbøll T, Knop FK. Safety of native glucose-dependent insulinotropic polypeptide in humans. Peptides 2024; 177:171214. [PMID: 38615716 DOI: 10.1016/j.peptides.2024.171214] [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: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
In this systematic review, we assessed the safety and possible safety events of native glucose-dependent insulinotropic polypeptide (GIP)(1-42) in human studies with administration of synthetic human GIP. We searched the PubMed database for all trials investigating synthetic human GIP(1-42) administration. A total of 67 studies were included. Study duration ranged from 30 min to 6 days. In addition to healthy individuals, the studies included individuals with impaired glucose tolerance, type 2 diabetes, type 1 diabetes, chronic pancreatitis and secondary diabetes, latent autoimmune diabetes in adults, diabetes caused by a mutation in the hepatocyte nuclear factor 1-alpha gene, end-stage renal disease, chronic renal insufficiency, critical illness, hypoparathyroidism, or cystic fibrosis-related diabetes. Of the included studies, 78% did not mention safety events, 10% of the studies reported that no safety events were observed in relation to GIP administration, and 15% of the studies reported safety events in relation to GIP administration with most frequently reported event being a moderate and transient increased heart rate. Gastrointestinal safety events, and changes in blood pressure were also reported. Plasma concentration of active GIP(1-42) increased linearly with dose independent of participant phenotype. There was no significant correlation between achieved maximal concentration of GIP(1-42) and reported safety events. Clearance rates of GIP(1-42) were similar between participant groups. In conclusion, the available data indicate that GIP(1-42) in short-term (up to 6 days) infusion studies is generally well-tolerated. The long-term safety of continuous GIP(1-42) administration is unknown.
Collapse
Affiliation(s)
- Mads M Helsted
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Nina L Schaltz
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel B Christensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Copenhagen Center for Translational Research, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark.
| |
Collapse
|
8
|
Liskiewicz A, Müller TD. Regulation of energy metabolism through central GIPR signaling. Peptides 2024; 176:171198. [PMID: 38527521 DOI: 10.1016/j.peptides.2024.171198] [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: 01/31/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024]
Abstract
In recent years, significant progress has been made to pharmacologically combat the obesity pandemic, particularly with regard to biochemically tailored drugs that simultaneously target the receptors for glucagon-like peptide-1 (GLP-1) and the glucose-dependent insulinotropic polypeptide (GIP). But while the pharmacological benefits of GLP-1 receptor (GLP-1R) agonism are widely acknowledged, the role of the GIP system in regulating systems metabolism remains controversial. When given in adjunct to GLP-1R agonism, both agonism and antagonism of the GIP receptor (GIPR) improves metabolic outcome in preclinical and clinical studies, and despite persistent concerns about its potential obesogenic nature, there is accumulating evidence indicating that GIP has beneficial metabolic effects via central GIPR agonism. Nonetheless, despite growing recognition of the GIP system as a valuable pharmacological target, there remains great uncertainty as to where and how GIP acts in the brain to regulate metabolism, and how GIPR agonism may differ from GIPR antagonism in control of energy metabolism. In this review we highlight current knowledge on the central action of GIP, and discuss open questions related to its multifaceted biology in the brain and the periphery.
Collapse
Affiliation(s)
- Arkadiusz Liskiewicz
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.
| |
Collapse
|
9
|
Novikoff A, Müller TD. Pharmacological Advances in Incretin-Based Polyagonism: What We Know and What We Don't. Physiology (Bethesda) 2024; 39:142-156. [PMID: 38353610 PMCID: PMC11368522 DOI: 10.1152/physiol.00032.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 02/21/2024] Open
Abstract
The prevalence of obesity continues to rise in both adolescents and adults, in parallel obesity is strongly associated with the increased incidence of type 2 diabetes, heart failure, certain types of cancer, and all-cause mortality. In relation to obesity, many pharmacological approaches of the past have tried and failed to combat the rising obesity epidemic, particularly due to insufficient efficacy or unacceptable side effects. However, while the history of antiobesity medication is plagued by failures and disappointments, we have witnessed over the last 10 years substantial progress, particularly in regard to biochemically optimized agonists at the receptor for glucagon-like peptide-1 (GLP-1R) and unimolecular coagonists at the receptors for GLP-1 and the glucose-dependent insulinotropic polypeptide (GIP). Although the GIP receptor:GLP-1R coagonists are being heralded as premier pharmacological tools for the treatment of obesity and diabetes, uncertainty remains as to why these drugs testify superiority over best-in-class GLP-1R monoagonists. Particularly with regard to GIP, there remains great uncertainty if and how GIP acts on systems metabolism and if the GIP system should be activated or inhibited to improve metabolic outcome in adjunct to GLP-1R agonism. In this review, we summarize recent advances in GLP-1- and GIP-based pharmacology and discuss recent findings and open questions related to how the GIP system affects systemic energy and glucose metabolism.
Collapse
Affiliation(s)
- Aaron Novikoff
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
10
|
Fernández-Cardero Á, Sierra-Cinos JL, López-Jiménez A, Beltrán B, Cuadrado C, García-Conesa MT, Bravo L, Sarriá B. Characterizing Factors Associated with Excess Body Weight: A Descriptive Study Using Principal Component Analysis in a Population with Overweight and Obesity. Nutrients 2024; 16:1143. [PMID: 38674834 PMCID: PMC11054611 DOI: 10.3390/nu16081143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity is a worldwide epidemic, making it crucial to understand how it can be effectively prevented/treated. Considering that obesity is a multifactorial condition, this article carried out a baseline cross-sectional study of the variables involved in the disorder. Eighty-four subjects with overweight/obesity were recruited. Dietary baseline information was obtained by analysing three 24 h recalls. Resting metabolic rate was measured using indirect calorimetry, physical activity was measured through accelerometry, cardiometabolic parameters were determined in blood samples and body composition via anthropometry and bioimpedance. A univariant and multivariate exploratory approach was carried out using principal component analysis (PCA). Large inter-individual variability was observed in dietetic, biochemical, and physical activity measurements (coefficient of variation ≥ 30%), but body composition was more uniform. Volunteers had an unbalanced diet and low levels of physical activity. PCA reduced the 26 analysed variables to 4 factors, accounting for 65.4% of the total data variance. The main factor was the "dietetic factor", responsible for 24.0% of the total variance and mainly related to energy intake, lipids, and saturated fatty acids. The second was the "cardiometabolic factor" (explaining 16.8% of the variability), the third was the "adiposity factor" (15.2%), and the last was the "serum cholesterol factor" (9.4%).
Collapse
Affiliation(s)
- Álvaro Fernández-Cardero
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/Jose Antonio Novais 6, 28040 Madrid, Spain; (Á.F.-C.); (A.L.-J.); (L.B.)
| | - José Luis Sierra-Cinos
- Department of Nutrition and Food Science I, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (J.L.S.-C.); (B.B.); (C.C.)
- Department of Health Science, School of Health Science, Universidad International Isabel I de Burgos (Ui1), C. de Fernán González, 76, 09003 Burgos, Spain
| | - Adrián López-Jiménez
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/Jose Antonio Novais 6, 28040 Madrid, Spain; (Á.F.-C.); (A.L.-J.); (L.B.)
| | - Beatriz Beltrán
- Department of Nutrition and Food Science I, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (J.L.S.-C.); (B.B.); (C.C.)
| | - Carmen Cuadrado
- Department of Nutrition and Food Science I, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (J.L.S.-C.); (B.B.); (C.C.)
| | - María Teresa García-Conesa
- Department of Food Science and Technology, Centro de Edafología y Biología Aplicada del Segura (CEBAS), Spanish National Research Council (CSIC), Campus de Espinardo, 30100 Murcia, Spain;
| | - Laura Bravo
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/Jose Antonio Novais 6, 28040 Madrid, Spain; (Á.F.-C.); (A.L.-J.); (L.B.)
| | - Beatriz Sarriá
- Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/Jose Antonio Novais 6, 28040 Madrid, Spain; (Á.F.-C.); (A.L.-J.); (L.B.)
- Department of Nutrition and Food Science I, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (J.L.S.-C.); (B.B.); (C.C.)
| |
Collapse
|
11
|
Wu N, An G. A Quantitative Systems Pharmacology Model of the Incretin Hormones GIP and GLP1, Glucagon, Glucose, Insulin, and the Small Molecule DPP-4 Inhibitor, Linagliptin. J Pharm Sci 2024; 113:278-289. [PMID: 37716531 DOI: 10.1016/j.xphs.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
In the current study, we established a comprehensive quantitative systems pharmacology (QSP) model using linagliptin as the model drug, where drug disposition, drug intervention on dipeptidyl peptidase-4 (DPP-4), glucose-dependent insulinotropic peptide (GIP), Glucagon-like peptide-1 (GLP-1), glucagon, glucose, and insulin are integrated together with the cross talk and feedback loops incorporated among the whole glycemic control system. In the final linagliptin QSP model, the complicated disposition of linagliptin was characterized by a 2-compartment pharmacokinetic (PK) model with an enterohepatic cycling (EHC) component as well as target-mediated drug disposition (TMDD) processes occurring in both tissues and plasma, and the inhibitory effect of linagliptin on DPP-4 was determined by the linagliptin-DPP-4 complex in the central compartment based on target occupancy principle. The integrated GIP-GLP1-glucagon-glucose-insulin system contains five indirect response models as the "skeleton" structure with 12 feedback loops incorporated within the glucose control system. Our model adequately characterized the substantial nonlinear PK of linagliptin, time course of DPP-4 inhibition, as well as the kinetics of GIP, GLP-1, glucagon, and glucose simultaneously in humans. Our model provided valuable insights on linagliptin pharmacokinetics/pharmacodynamics and complicated glucose homeostasis. Since the glucose regulation modeling framework within the QSP model is "drug-independent", our model can be easily adopted by others to evaluate the effect of other DPP-4 inhibitors on the glucose control system. In addition, our QSP model, which contains more components than other reported glucose regulation models, can potentially be used to evaluate the effect of combination antidiabetic therapy targeting different components of glucose control system.
Collapse
Affiliation(s)
- Nan Wu
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa city, IA, USA
| | - Guohua An
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa city, IA, USA.
| |
Collapse
|
12
|
Gasbjerg LS, Rosenkilde MM, Meier JJ, Holst JJ, Knop FK. The importance of glucose-dependent insulinotropic polypeptide receptor activation for the effects of tirzepatide. Diabetes Obes Metab 2023; 25:3079-3092. [PMID: 37551549 DOI: 10.1111/dom.15216] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/22/2023] [Accepted: 07/02/2023] [Indexed: 08/09/2023]
Abstract
Tirzepatide is a unimolecular co-agonist of the glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors recently approved for the treatment of type 2 diabetes by the US Food and Drug Administration and the European Medicine Agency. Tirzepatide treatment results in an unprecedented improvement of glycaemic control and lowering of body weight, but the contribution of the GIP receptor-activating component of tirzepatide to these effects is uncertain. In this review, we present the current knowledge about the physiological roles of the incretin hormones GLP-1 and GIP, their receptors, and previous results of co-targeting the two incretin hormone receptors in humans. We also analyse the molecular pharmacological, preclinical and clinical effects of tirzepatide to discuss the role of GIP receptor activation for the clinical effects of tirzepatide. Based on the available literature on the combination of GLP-1 and GIP receptor activation, tirzepatide does not seem to have a classical co-activating mode of action in humans. Rather, in vitro studies of the human GLP-1 and GIP receptors reveal a biased GLP-1 receptor activation profile and GIP receptor downregulation. Therefore, we propose three hypotheses for the mode of action of tirzepatide, which can be addressed in future, elaborate clinical trials.
Collapse
Affiliation(s)
- Laerke S Gasbjerg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juris J Meier
- Department of Internal Medicine, Gastroenterology and Diabetology, Augusta Clinic, Bochum, Germany
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
14
|
Regulation of feeding and therapeutic application of bioactive peptides. Pharmacol Ther 2022; 239:108187. [DOI: 10.1016/j.pharmthera.2022.108187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
15
|
Chueire VB, Muscelli E. Effect of free fatty acids on insulin secretion, insulin sensitivity and incretin effect - a narrative review. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:24-31. [PMID: 33320449 PMCID: PMC10528699 DOI: 10.20945/2359-3997000000313] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 09/26/2020] [Indexed: 11/23/2022]
Abstract
Deleterious effects of free fatty acids, FFAs, on insulin sensitivity are observed in vivo studies in humans. Mechanisms include impaired insulin signaling, oxidative stress, inflammation, and mitochondrial dysfunction, but the effects on insulin secretion are less well known. Our aim was to review the relationship of increased FFAs with insulin resistance, secretion and mainly with the incretin effect in humans. Narrative review. Increased endogenous or administered FFAs induce insulin resistance. FFAs effects on insulin secretion are debatable; inhibition and stimulation have been reported, depending on the type and duration of lipids exposition and the study subjects. Chronically elevated FFAs seem to decrease insulin biosynthesis, glucose-stimulated insulin secretion and β-cell glucose sensitivity. Lipids infusion decreases the response to incretins with unchanged incretin levels in volunteers with normal glucose tolerance. In contrast, FFAs reduction by acipimox did not restore the incretin effect in type-2 diabetes, probably due to the dysfunctional β-cell. Possible mechanisms of FFAs excess on incretin effect include reduction of the expression and levels of GLP-1 (glucagon like peptide-1) receptor, reduction of connexin-36 expression thus the coordinated secretory activity in response to GLP-1, and GIP (glucose-dependent insulinotropic polypeptide) receptors downregulation in islets cells. Increased circulating FFAs impair insulin sensitivity. Effects on insulin secretion are complex and controversial. Deleterious effects on the incretin-induced potentiation of insulin secretion were reported. More investigation is needed to better understand the extent and mechanisms of β-cell impairment and insulin resistance induced by increased FFAs and how to prevent them.
Collapse
Affiliation(s)
- Valeria Bahdur Chueire
- Departamento de Endocrinologia, Hospital da Pontifícia Universidade Católica de Campinas, Campinas, SP, Brasil,
| | - Elza Muscelli
- Departamento de Clínica Médica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brasil
| |
Collapse
|
16
|
Bowker N, Hansford R, Burgess S, Foley CN, Auyeung VPW, Erzurumluoglu AM, Stewart ID, Wheeler E, Pietzner M, Gribble F, Reimann F, Bhatnagar P, Coghlan MP, Wareham NJ, Langenberg C. Genetically Predicted Glucose-Dependent Insulinotropic Polypeptide (GIP) Levels and Cardiovascular Disease Risk Are Driven by Distinct Causal Variants in the GIPR Region. Diabetes 2021; 70:2706-2719. [PMID: 34426508 PMCID: PMC8564402 DOI: 10.2337/db21-0103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/16/2021] [Indexed: 12/30/2022]
Abstract
There is considerable interest in GIPR agonism to enhance the insulinotropic and extrapancreatic effects of GIP, thereby improving glycemic and weight control in type 2 diabetes (T2D) and obesity. Recent genetic epidemiological evidence has implicated higher GIPR-mediated GIP levels in raising coronary artery disease (CAD) risk, a potential safety concern for GIPR agonism. We therefore aimed to quantitatively assess whether the association between higher GIPR-mediated fasting GIP levels and CAD risk is mediated via GIPR or is instead the result of linkage disequilibrium (LD) confounding between variants at the GIPR locus. Using Bayesian multitrait colocalization, we identified a GIPR missense variant, rs1800437 (G allele; E354), as the putatively causal variant shared among fasting GIP levels, glycemic traits, and adiposity-related traits (posterior probability for colocalization [PPcoloc] > 0.97; PP explained by the candidate variant [PPexplained] = 1) that was independent from a cluster of CAD and lipid traits driven by a known missense variant in APOE (rs7412; distance to E354 ∼770 Kb; R 2 with E354 = 0.004; PPcoloc > 0.99; PPexplained = 1). Further, conditioning the association between E354 and CAD on the residual LD with rs7412, we observed slight attenuation in association, but it remained significant (odds ratio [OR] per copy of E354 after adjustment 1.03; 95% CI 1.02, 1.04; P = 0.003). Instead, E354's association with CAD was completely attenuated when conditioning on an additional established CAD signal, rs1964272 (R 2 with E354 = 0.27), an intronic variant in SNRPD2 (OR for E354 after adjustment for rs1964272: 1.01; 95% CI 0.99, 1.03; P = 0.06). We demonstrate that associations with GIP and anthropometric and glycemic traits are driven by genetic signals distinct from those driving CAD and lipid traits in the GIPR region and that higher E354-mediated fasting GIP levels are not associated with CAD risk. These findings provide evidence that the inclusion of GIPR agonism in dual GIPR/GLP1R agonists could potentiate the protective effect of GLP-1 agonists on diabetes without undue CAD risk, an aspect that has yet to be assessed in clinical trials.
Collapse
Affiliation(s)
- Nicholas Bowker
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - Robert Hansford
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - Stephen Burgess
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, U.K
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Christopher N Foley
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, U.K
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Victoria P W Auyeung
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - A Mesut Erzurumluoglu
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - Isobel D Stewart
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - Fiona Gribble
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science, and MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Cambridge, U.K
| | - Frank Reimann
- University of Cambridge, Wellcome Trust/MRC Institute of Metabolic Science, and MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Cambridge, U.K
| | - Pallav Bhatnagar
- Diabetes and Complications Therapy Area, Eli Lilly & Company, Indianapolis, IN
| | - Matthew P Coghlan
- Diabetes and Complications Therapy Area, Eli Lilly & Company, Indianapolis, IN
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, U.K.
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
17
|
Smit FX, van der Velden WJC, Kizilkaya HS, Nørskov A, Lückmann M, Hansen TN, Sparre-Ulrich AH, Qvotrup K, Frimurer TM, Rosenkilde MM. Investigating GIPR (ant)agonism: A structural analysis of GIP and its receptor. Structure 2021; 29:679-693.e6. [PMID: 33891864 DOI: 10.1016/j.str.2021.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/01/2021] [Accepted: 04/02/2021] [Indexed: 12/21/2022]
Abstract
The glucose-dependent insulinotropic polypeptide (GIP) is a 42-residue metabolic hormone that is actively being targeted for its regulatory role of glycemia and energy balance. Limited structural data of its receptor has made ligand design tedious. This study investigates the structure and function of the GIP receptor (GIPR), using a homology model based on the GLP-1 receptor. Molecular dynamics combined with in vitro mutational data were used to pinpoint residues involved in ligand binding and/or receptor activation. Significant differences in binding mode were identified for the naturally occurring agonists GIP(1-30)NH2 and GIP(1-42) compared with high potency antagonists GIP(3-30)NH2 and GIP(5-30)NH2. Residues R1832.60, R1902.67, and R3005.40 are shown to be key for activation of the GIPR, and evidence suggests that a disruption of the K293ECL2-E362ECL3 salt bridge by GIPR antagonists strongly reduces GIPR activation. Combinatorial use of these findings can benefit rational design of ligands targeting the GIPR.
Collapse
Affiliation(s)
- Florent X Smit
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark; Section for Metabolic Receptology, Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Wijnand J C van der Velden
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Hüsün S Kizilkaya
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Amalie Nørskov
- Department of Chemistry, Technical University of Denmark, Lyngby 2800, Denmark
| | - Michael Lückmann
- Section for Metabolic Receptology, Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Tobias N Hansen
- Department of Chemistry, Technical University of Denmark, Lyngby 2800, Denmark
| | - Alexander H Sparre-Ulrich
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Katrine Qvotrup
- Department of Chemistry, Technical University of Denmark, Lyngby 2800, Denmark
| | - Thomas M Frimurer
- Section for Metabolic Receptology, Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| |
Collapse
|
18
|
Association of Gut Hormones and Microbiota with Vascular Dysfunction in Obesity. Nutrients 2021; 13:nu13020613. [PMID: 33668627 PMCID: PMC7918888 DOI: 10.3390/nu13020613] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
In the past few decades, obesity has reached pandemic proportions. Obesity is among the main risk factors for cardiovascular diseases, since chronic fat accumulation leads to dysfunction in vascular endothelium and to a precocious arterial stiffness. So far, not all the mechanisms linking adipose tissue and vascular reactivity have been explained. Recently, novel findings reported interesting pathological link between endothelial dysfunction with gut hormones and gut microbiota and energy homeostasis. These findings suggest an active role of gut secretome in regulating the mediators of vascular function, such as nitric oxide (NO) and endothelin-1 (ET-1) that need to be further investigated. Moreover, a central role of brain has been suggested as a main player in the regulation of the different factors and hormones beyond these complex mechanisms. The aim of the present review is to discuss the state of the art in this field, by focusing on the processes leading to endothelial dysfunction mediated by obesity and metabolic diseases, such as insulin resistance. The role of perivascular adipose tissue (PVAT), gut hormones, gut microbiota dysbiosis, and the CNS function in controlling satiety have been considered. Further understanding the crosstalk between these complex mechanisms will allow us to better design novel strategies for the prevention of obesity and its complications.
Collapse
|
19
|
Li W, Li P, Li R, Yu Z, Sun X, Ji G, Yang X, Zhu L, Zhu S. GLP1R Single-Nucleotide Polymorphisms rs3765467 and rs10305492 Affect β Cell Insulin Secretory Capacity and Apoptosis Through GLP-1. DNA Cell Biol 2020; 39:1700-1710. [PMID: 32721233 DOI: 10.1089/dna.2020.5424] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The increased secretion of glucagon-like peptide-1 (GLP-1) after Roux-en-Y gastric bypass (RYGB) is regarded as the main reason for the improvement of blood glucose. However, the single-nucleotide polymorphisms (SNPs) of GLP-1 Receptor (GLP1R) impair receptor function, subsequently affecting β cell insulin secretion function, ultimately affecting the efficacy of RYGB. In this study, we revealed that two SNPs in GLP1R gene, rs3765467 and rs10305492, could significantly reduce the insulin secreted by β cells and the cyclic AMP concentration, whereas promote β cell apoptosis. Under high glucose exposure, rs3765467 and rs10305492 impaired β cell secretion of insulin and β cell viability in the same way; in other words, GLP1R rs3765467 and rs10305492 exert an effect on pancreatic β cell glucose-stimulated insulin secretion. Moreover, GLP-1 antagonist Exendin (9-39) further enhanced, whereas GLP-1 agonist Exendin-4 partially attenuated the effects of SNPs on the functions and apoptosis of β cells. In conclusion, the rs3765467 and rs10305492 SNPs in GLP1R show to exert a critical effect on regulating insulin secretory capacity of β cells and β cell mass. Through leading to the dysfunction and apoptosis of β cells, GLP1R rs3765467 and rs10305492 might also impair GLP-1 interaction with GLP1R, therefore attenuating the therapeutic effect of RYGB.
Collapse
Affiliation(s)
- Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Rao Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhaomei Yu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guangnian Ji
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiangwu Yang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
Gabe MBN, van der Velden WJC, Gadgaard S, Smit FX, Hartmann B, Bräuner‐Osborne H, Rosenkilde MM. Enhanced agonist residence time, internalization rate and signalling of the GIP receptor variant [E354Q] facilitate receptor desensitization and long-term impairment of the GIP system. Basic Clin Pharmacol Toxicol 2020; 126 Suppl 6:122-132. [PMID: 31299132 PMCID: PMC7317972 DOI: 10.1111/bcpt.13289] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/03/2019] [Indexed: 12/21/2022]
Abstract
In patients with type 2 diabetes mellitus (T2DM), the insulinotropic action of the GIP system is desensitized, whereas this is not the case for the GLP-1 system. This has raised an interesting discussion of whether GIP agonists or antagonists are most suitable for future treatment of T2DM together with GLP-1-based therapies. Homozygous carriers of the GIP receptor (GIPR) variant, [E354Q], display lower bone mineral density, increased bone fracture risk and slightly increased blood glucose. Here, we present an in-depth molecular pharmacological phenotyping of GIPR-[E354Q]. In silico modelling suggested similar interaction of the endogenous agonist GIP(1-42) to [E354Q] as to GIPR wt. This was supported by homologous competition binding in COS-7 cells revealing GIPR wt-like affinities of GIP(1-42) with Kd values of ~2 nmol/L and wt-like agonist association rates (Kon ). In contrast, the dissociation rates (Koff ) were slower, resulting in 25% higher agonist residence time for GIPR-[E354Q]. Moreover, in Gαs signalling (cAMP production) GIP(1-42) was ~2-fold more potent and more efficacious on GIPR-[E354Q] compared to wt with 17.5% higher basal activity. No difference from GIPR wt was found in the recruitment of β-arrestin 2, whereas the agonist-induced internalization rate was 2.1- to 2.3-fold faster for [E354Q]. Together with the previously described impaired recycling of [E354Q], our findings with enhanced signalling and internalization rate possibly explained by an altered ligand-binding kinetics will lead to receptor desensitization and down-regulation. This could explain the long-term functional impairment of the GIP system in bone metabolism and blood sugar maintenance for [E354Q] carriers and may shed light on the desensitization of the insulinotropic action of GIP in patients with T2DM.
Collapse
Affiliation(s)
- Maria Buur Nordskov Gabe
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Wijnand J. C. van der Velden
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Sarina Gadgaard
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Florent Xavier Smit
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenCopenhagenDenmark
| | - Hans Bräuner‐Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
21
|
Nauck MA, Holle H, Kahle M, Tytko A, Deacon CF, Holst JJ, Meier JJ. No evidence of tachyphylaxis for insulinotropic actions of glucose-dependent insulinotropic polypeptide (GIP) in subjects with type 2 diabetes, their first-degree relatives, or in healthy subjects. Peptides 2020; 125:170176. [PMID: 31669136 DOI: 10.1016/j.peptides.2019.170176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND, AIMS In patients with type 2 diabetes, the lost insulinotropic effect of the incretin hormone glucose-dependent insulinotropic polypeptide (GIP) is more apparent after continuous versus bolus administration. To test whether the difference might be explained by rapid tachyphylaxis in response to elevated concentrations of GIP, and whether patients with type 2 diabetes and their relatives are more susceptible to tachyphylaxis than healthy subjects. PATIENTS AND METHODS In a two-way crossover design, insulinotropic responses to repeated bolus injection (50 pmol/kg body weight at 30 and 120 min) and continuous infusion of GIP (2 pmol.kg-1.min-1 from 30 to 180 min) under hyperglycaemic clamp conditions (8.5 mmol/l) was compared in age- gender- and weight-matched patients with type 2 diabetes, first degree relatives of such patients, and healthy subjects. RESULTS Insulin secretory responses to the first and second GIP bolus were not significantly different in any of the subject groups. Subjects with type 2 diabetes had a significant relative impairment versus healthy subjects with continuous (C-peptide, -13.2 %, p < 0.05), but not with repeated bolus administration of GIP (+11.1 %, n.s.). First-degree relatives tended to hyper-secrete insulin with bolus or continuous administrations of GIP. CONCLUSIONS Rapid tachyphylaxis in response to continuous exposure to slightly supraphysiological concentrations of GIP does not explain the reduced insulinotropic response to GIP infusions in patients with type 2 diabetes or their first-degree relatives.
Collapse
Affiliation(s)
- M A Nauck
- Diabeteszentrum Bad Lauterberg, Bad Lauterberg im Harz, Germany; Diabetes Center Bochum-Hattingen, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany.
| | - H Holle
- Diabeteszentrum Bad Lauterberg, Bad Lauterberg im Harz, Germany
| | - M Kahle
- Diabeteszentrum Bad Lauterberg, Bad Lauterberg im Harz, Germany; Diabetes Center Bochum-Hattingen, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - A Tytko
- Diabeteszentrum Bad Lauterberg, Bad Lauterberg im Harz, Germany
| | - C F Deacon
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - J J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - J J Meier
- Diabetes Center Bochum-Hattingen, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
22
|
Christensen MB, Gasbjerg LS, Heimbürger SM, Stensen S, Vilsbøll T, Knop FK. GIP's involvement in the pathophysiology of type 2 diabetes. Peptides 2020; 125:170178. [PMID: 31682875 DOI: 10.1016/j.peptides.2019.170178] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
During the past four decades derangements in glucose-dependent insulinotropic polypeptide (GIP) biology has been viewed upon as contributing factors to various parts of the pathophysiology type 2 diabetes. This overview outlines and discusses the impaired insulin responses to GIP as well as the effect of GIP on glucagon secretion and the potential involvement of GIP in the obesity and bone disease associated with type 2 diabetes. As outlined in this review, it is unlikely that the impaired insulinotropic effect of GIP occurs as a primary event in the development of type 2 diabetes, but rather develops once the diabetic state is present and beta cells are unable to maintain normoglycemia. In various models, GIP has effects on glucagon secretion, bone and lipid homeostasis, but whether these effects contribute substantially to the pathophysiology of type 2 diabetes is at present controversial. The review also discusses the substantial uncertainty surrounding the translation of preclinical data relating to the GIP system and outline future research directions.
Collapse
Affiliation(s)
- Mikkel B Christensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Biomedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sebastian M Heimbürger
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Stensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Biomedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Gentofte Hospital, Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Gentofte Hospital, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Gasbjerg LS, Bergmann NC, Stensen S, Christensen MB, Rosenkilde MM, Holst JJ, Nauck M, Knop FK. Evaluation of the incretin effect in humans using GIP and GLP-1 receptor antagonists. Peptides 2020; 125:170183. [PMID: 31693916 DOI: 10.1016/j.peptides.2019.170183] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) potentiate glucose-induced insulin secretion and are therefore thought to be responsible for the incretin effect. The magnitude of the incretin effect, defined as the fraction of postprandial insulin secretion stimulated by intestinal factors, has been reported to be up to ∼60% in healthy individuals. In several pathological conditions but especially in patients with type 2 diabetes, the incretin effect is severely reduced or even absent. In line with this, the insulinotropic effects of GIP and GLP-1 are impaired in patients with type 2 diabetes, even when administered in supraphysiological doses. In healthy individuals, GIP has been proposed to be the most important incretin hormone of the two, but the individual contribution of the two is difficult to determine. However, using incretin hormone receptor antagonists: the novel GIP receptor antagonist GIP(3-30)NH2 and the widely used GLP-1 receptor antagonist exendin(9-39)NH2, we can now distinguish between the effects of the two hormones. In this review, we present and discuss studies in which the individual contribution of GIP and GLP-1 to the incretin effect in healthy individuals have been estimated and discuss the limitations of using incretin hormone receptor antagonists.
Collapse
Affiliation(s)
- Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Natasha C Bergmann
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Signe Stensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Mikkel B Christensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Nauck
- Diabetes Division, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
24
|
Khan R, Tomas A, Rutter GA. Effects on pancreatic Beta and other Islet cells of the glucose-dependent insulinotropic polypeptide. Peptides 2020; 125:170201. [PMID: 31751656 DOI: 10.1016/j.peptides.2019.170201] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is a gut-derived incretin that, in common with glucagon-like peptide-1 (GLP-1), has both insulin releasing and extra-pancreatic glucoregulatory actions. GIP is released in response to glucose or fat absorption and acts on the GIP receptor (GIPR) to potentiate insulin release from pancreatic beta cells. GIP has also been shown to promote beta cell survival and stimulate the release of GLP-1 from islet alpha cells. There is now evidence to suggest that low levels of GIP are secreted from alpha cells and may act in a paracrine manner to prime neighboring beta cells for insulin release. In addition, GIP acts on adipocytes to stimulate fat storage and can exert anorexigenic effects via actions in the hypothalamus. Contrary to GLP-1, the development of effective GIP-based T2D treatments has been hindered by poor bioavailability and attenuation of beta cell responses to GIP in some patients with sub-optimally controlled T2D. Recently, longer-acting GIP agonists that exhibit enzymatic stability, as well as dual GLP-1/GIP agonists which provide simultaneous improvement in glucose and weight control have been generated and successfully tested in animal T2D models. This, together with reports on GIP antagonists that may protect against obesity, has revived the interest on the GIP/GIPR axis as a potential anti-diabetic pathway. In this review, we summarize the known aspects of the effects of GIP on beta and other islet cells and discuss the most recent developments on GIP-based therapeutic agents for the improvement of beta cell function in T2D patients.
Collapse
Affiliation(s)
- Rabeet Khan
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
25
|
Frost F, Jones GH, Dyce P, Jackson V, Nazareth D, Walshaw MJ. Loss of incretin effect contributes to postprandial hyperglycaemia in cystic fibrosis-related diabetes. Diabet Med 2019; 36:1367-1374. [PMID: 31466128 DOI: 10.1111/dme.14121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2019] [Indexed: 12/23/2022]
Abstract
AIM To investigate the incretin axis in people with cystic fibrosis. METHODS Adults with cystic fibrosis-related diabetes, cystic fibrosis without diabetes, and controls (adults without cystic fibrosis and without diabetes) underwent an oral glucose tolerance test and then a closely matched isoglycaemic i.v. glucose infusion. On each occasion, glucose, insulin, C-peptide, total and active glucagon-like peptide-1 and gastric inhibitory polypeptide responses were recorded and incremental areas under curves were calculated for 60 and 240 min. RESULTS Five adults with cystic fibrosis-related diabetes, six with cystic fibrosis without diabetes and six controls, matched for age and BMI, completed the study. Glucose during oral glucose tolerance test closely matched those during isoglycaemic i.v. glucose infusion. The calculated incretin effect was similar in the control group and the cystic fibrosis without diabetes group (28% and 29%, respectively), but was lost in the cystic fibrosis-related diabetes group (cystic fibrosis-related diabetes vs control group: -6% vs 28%; p=0.03). No hyposecretion of glucagon-like peptide-1 or gastric inhibitory polypeptide was observed; conversely, 60-min incremental area under the curve for total glucagon-like peptide-1 was significantly higher in the cystic fibrosis-related diabetes group than in the control group [1070.4 (254.7) vs 694.97 (308.1); p=0.03] CONCLUSIONS: The incretin effect was lost in cystic fibrosis-related diabetes despite adequate secretion of the incretin hormones. These data support the concept that reduced incretin hormone insulinotropic activity contributes significantly to postprandial hyperglycaemia in cystic fibrosis-related diabetes.
Collapse
Affiliation(s)
- F Frost
- Adult CF Centre, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - G H Jones
- Adult CF Centre, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
- Royal Liverpool and Broadgreen University NHS Foundation Trust, Liverpool, UK
| | - P Dyce
- Cystic Fibrosis Related Diabetes Service, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
| | - V Jackson
- Cystic Fibrosis Related Diabetes Service, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
| | - D Nazareth
- Adult CF Centre, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - M J Walshaw
- Adult CF Centre, Liverpool Heart and Chest NHS Foundation Trust, Liverpool, UK
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
26
|
Douros JD, Tong J, D’Alessio DA. The Effects of Bariatric Surgery on Islet Function, Insulin Secretion, and Glucose Control. Endocr Rev 2019; 40:1394-1423. [PMID: 31241742 PMCID: PMC6749890 DOI: 10.1210/er.2018-00183] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 04/23/2019] [Indexed: 01/19/2023]
Abstract
Although bariatric surgery was developed primarily to treat morbid obesity, evidence from the earliest clinical observations to the most recent clinical trials consistently demonstrates that these procedures have substantial effects on glucose metabolism. A large base of research indicates that bariatric surgeries such as Roux-en-Y gastric bypass (RYGB), vertical sleeve gastrectomy (VSG), and biliopancreatic diversion (BPD) improve diabetes in most patients, with effects frequently evident prior to substantial weight reduction. There is now unequivocal evidence from randomized controlled trials that the efficacy of surgery is superior to intensive life-style/medical management. Despite advances in the clinical understanding and application of bariatric surgery, there remains only limited knowledge of the mechanisms by which these procedures confer such large changes to metabolic physiology. The improvement of insulin sensitivity that occurs with weight loss (e.g., the result of diet, illness, physical training) also accompanies bariatric surgery. However, there is evidence to support specific effects of surgery on insulin clearance, hepatic glucose production, and islet function. Understanding the mechanisms by which surgery affects these parameters of glucose regulation has the potential to identify new targets for therapeutic discovery. Studies to distinguish among bariatric surgeries on key parameters of glucose metabolism are limited but would be of considerable value to assist clinicians in selecting specific procedures and investigators in delineating the resulting physiology. This review is based on literature related to factors governing glucose metabolism and insulin secretion after the commonly used RYGB and VSG, and the less frequently used BPD and adjustable gastric banding.
Collapse
Affiliation(s)
- Jonathan D Douros
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Jenny Tong
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - David A D’Alessio
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| |
Collapse
|
27
|
Chia CW, Egan JM. Incretins in obesity and diabetes. Ann N Y Acad Sci 2019; 1461:104-126. [PMID: 31392745 PMCID: PMC10131087 DOI: 10.1111/nyas.14211] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/13/2019] [Accepted: 07/18/2019] [Indexed: 12/11/2022]
Abstract
Incretins are hormones secreted from enteroendocrine cells after nutrient intake that stimulate insulin secretion from β cells in a glucose-dependent manner. Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are the only two known incretins. Dysregulation of incretin secretion and actions are noted in diseases such as obesity and diabetes. In this review, we first summarize our traditional understanding of the physiology of GIP and GLP-1, and our current knowledge of the relationships between GIP and GLP-1 and obesity and diabetes. Next, we present the results from major randomized controlled trials on the use of GLP-1 receptor agonists for managing type 2 diabetes, and emerging data on treating obesity and prediabetes. We conclude with a glimpse of the future with possible complex interactions between nutrients, gut microbiota, the endocannabinoid system, and enteroendocrine cells.
Collapse
Affiliation(s)
- Chee W Chia
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Josephine M Egan
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
28
|
Shah R, Subhan F, Sultan SM, Haq M, Ahmad H, Khan QR, Ali G, Ullah S, Ullah I. Metabolic dysregulation in early onset psychiatric disorder before and after exposure to antipsychotic drugs. BRAZ J PHARM SCI 2019. [DOI: 10.1590/s2175-97902019000117825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Rehmat Shah
- University of Peshawar, Pakistan; Health Department, Pakistan
| | | | | | - Matiul Haq
- , Institute of Radiotherapy and Nuclear Medicine, Pakistan
| | | | | | | | | | | |
Collapse
|
29
|
Bliss ES, Whiteside E. The Gut-Brain Axis, the Human Gut Microbiota and Their Integration in the Development of Obesity. Front Physiol 2018; 9:900. [PMID: 30050464 PMCID: PMC6052131 DOI: 10.3389/fphys.2018.00900] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022] Open
Abstract
Obesity is a global epidemic, placing socioeconomic strain on public healthcare systems, especially within the so-called Western countries, such as Australia, United States, United Kingdom, and Canada. Obesity results from an imbalance between energy intake and energy expenditure, where energy intake exceeds expenditure. Current non-invasive treatments lack efficacy in combating obesity, suggesting that obesity is a multi-faceted and more complex disease than previously thought. This has led to an increase in research exploring energy homeostasis and the discovery of a complex bidirectional communication axis referred to as the gut-brain axis. The gut-brain axis is comprised of various neurohumoral components that allow the gut and brain to communicate with each other. Communication occurs within the axis via local, paracrine and/or endocrine mechanisms involving a variety of gut-derived peptides produced from enteroendocrine cells (EECs), including glucagon-like peptide 1 (GLP1), cholecystokinin (CCK), peptide YY3-36 (PYY), pancreatic polypeptide (PP), and oxyntomodulin. Neural networks, such as the enteric nervous system (ENS) and vagus nerve also convey information within the gut-brain axis. Emerging evidence suggests the human gut microbiota, a complex ecosystem residing in the gastrointestinal tract (GIT), may influence weight-gain through several inter-dependent pathways including energy harvesting, short-chain fatty-acids (SCFA) signalling, behaviour modifications, controlling satiety and modulating inflammatory responses within the host. Hence, the gut-brain axis, the microbiota and the link between these elements and the role each plays in either promoting or regulating energy and thereby contributing to obesity will be explored in this review.
Collapse
Affiliation(s)
- Edward S. Bliss
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | | |
Collapse
|
30
|
Nauck MA, Meier JJ. Incretin hormones: Their role in health and disease. Diabetes Obes Metab 2018; 20 Suppl 1:5-21. [PMID: 29364588 DOI: 10.1111/dom.13129] [Citation(s) in RCA: 500] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
Abstract
Incretin hormones are gut peptides that are secreted after nutrient intake and stimulate insulin secretion together with hyperglycaemia. GIP (glucose-dependent insulinotropic polypeptide) und GLP-1 (glucagon-like peptide-1) are the known incretin hormones from the upper (GIP, K cells) and lower (GLP-1, L cells) gut. Together, they are responsible for the incretin effect: a two- to three-fold higher insulin secretory response to oral as compared to intravenous glucose administration. In subjects with type 2 diabetes, this incretin effect is diminished or no longer present. This is the consequence of a substantially reduced effectiveness of GIP on the diabetic endocrine pancreas, and of the negligible physiological role of GLP-1 in mediating the incretin effect even in healthy subjects. However, the insulinotropic and glucagonostatic effects of GLP-1 are preserved in subjects with type 2 diabetes to the degree that pharmacological stimulation of GLP-1 receptors significantly reduces plasma glucose and improves glycaemic control. Thus, it has become a parent compound of incretin-based glucose-lowering medications (GLP-1 receptor agonists and inhibitors of dipeptidyl peptidase-4 or DPP-4). GLP-1, in addition, has multiple effects on various organ systems. Most relevant are a reduction in appetite and food intake, leading to weight loss in the long term. Since GLP-1 secretion from the gut seems to be impaired in obese subjects, this may even indicate a role in the pathophysiology of obesity. Along these lines, an increased secretion of GLP-1 induced by delivering nutrients to lower parts of the small intestines (rich in L cells) may be one factor (among others like peptide YY) explaining weight loss and improvements in glycaemic control after bariatric surgery (e.g., Roux-en-Y gastric bypass). GIP and GLP-1, originally characterized as incretin hormones, have additional effects in adipose cells, bone, and the cardiovascular system. Especially, the latter have received attention based on recent findings that GLP-1 receptor agonists such as liraglutide reduce cardiovascular events and prolong life in high-risk patients with type 2 diabetes. Thus, incretin hormones have an important role physiologically, namely they are involved in the pathophysiology of obesity and type 2 diabetes, and they have therapeutic potential that can be traced to well-characterized physiological effects.
Collapse
Affiliation(s)
- Michael A Nauck
- Diabetes Center Bochum-Hattingen, Medical Department I, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - Juris J Meier
- Diabetes Center Bochum-Hattingen, Medical Department I, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| |
Collapse
|
31
|
Gasbjerg LS, Gabe MBN, Hartmann B, Christensen MB, Knop FK, Holst JJ, Rosenkilde MM. Glucose-dependent insulinotropic polypeptide (GIP) receptor antagonists as anti-diabetic agents. Peptides 2018; 100:173-181. [PMID: 29412817 DOI: 10.1016/j.peptides.2017.11.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 01/07/2023]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is an intestinal hormone with a broad range of physiological actions. In the postprandial state, the hormone stimulates insulin secretion and during eu- and hypoglycemia, it stimulates glucagon secretion. In addition, GIP increases triacylglycerol (TAG) uptake in adipose tissue and decreases bone resorption. However, the importance of these actions in humans are not clearly understood as a specific GIP receptor (GIPR) antagonist - an essential tool to study GIP physiology - has been missing. Several different GIPR antagonists have been identified comprising both peptides, vaccines against GIP, GIP antibodies or antibodies against the GIPR. However, most of these have only been tested in rodents. In vitro, N- and C-terminally truncated GIP variants are potent and efficacious GIPR antagonists. Recently, GIP(3-30)NH2, a naturally occurring peptide, was shown to block the GIPR in humans and decrease GIP-induced insulin secretion as well as adipose tissue blood flow and TAG uptake. So far, there are no studies with a GIPR antagonist in patients with type 2 diabetes (T2D), but because the elevations in fasting plasma glucagon and paradoxical postprandial glucagon excursions, seen in patients with T2D, are aggravated by GIP, a GIPR antagonist could partly alleviate this and possibly improve the fasting and postprandial glycemia. Since the majority of patients with T2D are overweight, inhibition of GIP-induced fat deposition may be beneficial as well. Here we summarize the studies of GIPR antagonists and discuss the therapeutic potential of the GIP system in humans.
Collapse
Affiliation(s)
- Lærke Smidt Gasbjerg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
| | - Maria Buur Nordskov Gabe
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Bring Christensen
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Filip Krag Knop
- NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; NNF Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Sun EWL, Martin AM, Young RL, Keating DJ. The Regulation of Peripheral Metabolism by Gut-Derived Hormones. Front Endocrinol (Lausanne) 2018; 9:754. [PMID: 30662430 PMCID: PMC6328484 DOI: 10.3389/fendo.2018.00754] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Enteroendocrine cells lining the gut epithelium constitute the largest endocrine organ in the body and secrete over 20 different hormones in response to cues from ingested foods and changes in nutritional status. Not only do these hormones convey signals from the gut to the brain via the gut-brain axis, they also act directly on metabolically important peripheral targets in a highly concerted fashion to maintain energy balance and glucose homeostasis. Gut-derived hormones released during fasting tend to be orexigenic and have hyperglycaemic potential. Conversely, gut hormones secreted postprandially generally promote satiety and facilitate glucose clearance. Although some of the metabolic benefits conferred by bariatric surgeries have been ascribed to changes in the secretory profiles of various gut hormones, the therapeutic potential of the enteroendocrine system as a viable target against metabolic diseases remain largely underexploited, except for incretin-mimetics. This review provides a brief overview of the physiological importance and highlights the therapeutic potential of the following gut hormones: serotonin, glucose-dependent insulinotropic peptide, glucagon-like peptide 1, oxyntomodulin, peptide YY, insulin-like peptide 5, and ghrelin.
Collapse
Affiliation(s)
- Emily W. L. Sun
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Alyce M. Martin
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Richard L. Young
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Damien J. Keating
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Damien J. Keating
| |
Collapse
|
33
|
Paternoster S, Falasca M. Dissecting the Physiology and Pathophysiology of Glucagon-Like Peptide-1. Front Endocrinol (Lausanne) 2018; 9:584. [PMID: 30364192 PMCID: PMC6193070 DOI: 10.3389/fendo.2018.00584] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/14/2018] [Indexed: 12/11/2022] Open
Abstract
An aging world population exposed to a sedentary life style is currently plagued by chronic metabolic diseases, such as type-2 diabetes, that are spreading worldwide at an unprecedented rate. One of the most promising pharmacological approaches for the management of type 2 diabetes takes advantage of the peptide hormone glucagon-like peptide-1 (GLP-1) under the form of protease resistant mimetics, and DPP-IV inhibitors. Despite the improved quality of life, long-term treatments with these new classes of drugs are riddled with serious and life-threatening side-effects, with no overall cure of the disease. New evidence is shedding more light over the complex physiology of GLP-1 in health and metabolic diseases. Herein, we discuss the most recent advancements in the biology of gut receptors known to induce the secretion of GLP-1, to bridge the multiple gaps into our understanding of its physiology and pathology.
Collapse
|
34
|
Microbial Regulation of Glucose Metabolism and Insulin Resistance. Genes (Basel) 2017; 9:genes9010010. [PMID: 29286343 PMCID: PMC5793163 DOI: 10.3390/genes9010010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes is a combined disease, resulting from a hyperglycemia and peripheral and hepatic insulin resistance. Recent data suggest that the gut microbiota is involved in diabetes development, altering metabolic processes including glucose and fatty acid metabolism. Thus, type 2 diabetes patients show a microbial dysbiosis, with reduced butyrate-producing bacteria and elevated potential pathogens compared to metabolically healthy individuals. Furthermore, probiotics are a known tool to modulate the microbiota, having a therapeutic potential. Current literature will be discussed to elucidate the complex interaction of gut microbiota, intestinal permeability and inflammation leading to peripheral and hepatic insulin resistance. Therefore, this review aims to generate a deeper understanding of the underlying mechanism of potential microbial strains, which can be used as probiotics.
Collapse
|
35
|
Shalaby SM, Zidan HE, Shokry A, Saeed J, El-Sokkary RH. Association of incretin receptors genetic polymorphisms with type 2 diabetes mellitus in Egyptian patients. J Gene Med 2017; 19:e2973. [DOI: 10.1002/jgm.2973] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Sally M. Shalaby
- Medical Biochemistry Department, Faculty of Medicine; Zagazig University; Zagazig Egypt
| | - Haidy E. Zidan
- Medical Biochemistry Department, Faculty of Medicine; Zagazig University; Zagazig Egypt
| | - Amira Shokry
- Internal Medicine Department, Faculty of Medicine; Zagazig University; Zagazig Egypt
| | - Jehan Saeed
- Internal Medicine Department, Faculty of Medicine; Zagazig University; Zagazig Egypt
| | - Rehab H. El-Sokkary
- Microbiology & Immunolgy Department, Faculty of Medicine; Zagazig University; Zagazig Egypt
| |
Collapse
|
36
|
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: 2.8] [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
|
37
|
Neonatal milk supplementation in lambs has persistent effects on growth and metabolic function that differ by sex and gestational age. Br J Nutr 2016; 116:1912-1925. [DOI: 10.1017/s0007114516004013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
AbstractThe perinatal environment has a major influence on long-term health and disease risk. Preterm birth alters early-life environment and is associated with altered metabolic function in adulthood. Whether preterm birthper seor the early nutritional interventions used to support growth in preterm infants underpins this association is unknown. Lambs born preterm, following dexamethasone induction of labour, or spontaneously at term were randomised to receive nutrient supplementation, analogous to the milk fortifier used clinically or water as a control for the first 2 weeks after birth. Thereafter, nutrition was not different between groups. Growth was monitored, and the glucose–insulin axis function was assessed in juvenile (4 months) and adult life (14 months). Early nutrition influenced adult metabolic function and body composition to a greater extent than preterm birth. In supplemented females, arginine-stimulated insulin secretion was increased in preterm but reduced in term-born juveniles compared with controls (repeated-measures ANOVAP<0·01). In supplemented preterm males, adult weight, ponderal index (PI) and fasting insulin concentrations were elevated compared with preterm controls (weight, 75 (sem3)v. 69 (sem2) kg; PI, 48·0 (sem2·1)v. 43·7 (sem1·7) kg/m3; fasting insulin, 0·19 (sem0·02)v. 0·10 (sem0·02) ng/ml). Conversely, supplemented term-born males had reduced adult weight, PI and fasting insulin concentrations compared with term-born controls (weight, 64 (sem2)v. 70 (sem2) kg; PI, 44·4 (sem1·8)v. 48·2 (sem1·7) kg/m3; fasting insulin, 0·09 (sem0·02)v. 0·14 (sem0·02) ng/ml; all group×supplement interactionsP<0·05). Adult metabolic health may reflect both gestational age at birth and early nutrition. Human studies are urgently needed to investigate the adult sex-specific health implications of neonatal nutritional strategies.
Collapse
|
38
|
Campbell RK. Diabetes: Rationale for Dipeptidyl Peptidase 4 Inhibitors: A New Class of Oral Agents for the Treatment of Type 2 Diabetes Mellitus. Ann Pharmacother 2016; 41:51-60. [PMID: 17190843 DOI: 10.1345/aph.1h459] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Objective: To review advances in understanding the pathophysiologic basis of type 2 diabetes mellitus and the pharmacology and mechanism of action of dipeptidyl peptidase 4 (DPP-4) inhibition in correcting the underlying defects in glycemic control. Data Sources: Articles were identified through MEDLINE for the period 1966 through November 2006. Abstracts and presentations from the American Diabetes Association Scientific Sessions and the European Association for the Study of Diabetes (2002–2006) were also searched for scientific reports on DPP-4 inhibitors. Study Selection And Data Extraction: Abstracts, original clinical and preclinical research reports, and review articles published in the English language were identified for review. Literature discussing glucose regulation, incretin hormones, type 2 diabetes pathophysiology, and DPP-4 inhibition were evaluated and selected based on consideration of their support for the proof of concept, mechanistic and in vivo findings, and timeliness. Data Synthesis: The search for new and effective therapies for type 2 diabetes has led to the identification of a novel therapeutic target, the incretin hormones, which play a role in mediating glucose homeostasis via effects on glucagon and insulin secretion from pancreatic islet α- and β-cells, respectively. The incretins' glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide are rapidly inactivated by the enzyme DPP-4. DPP-4 inhibitor agents act by blocking the active site of DPP-4, thereby preventing inactivation of and prolonging the duration of action of incretins, which in turn helps to correct the defective insulin and glucagon secretion that marks type 2 diabetes. Clinical studies to date indicate that DPP-4 inhibitors effectively stimulate insulin secretion, suppress glucagon release, and improve glucose control in patients with type 2 diabetes. These agents are well tolerated and have a low incidence of adverse effects. Conclusions: The DPP-4 inhibitors are novel agents for the treatment of type 2 diabetes. Compounds under development in this new class of oral antidiabetic drugs may be free of the limitations of current therapies.
Collapse
Affiliation(s)
- R Keith Campbell
- College of Pharmacy, Wegner Hall #147, PO Box 646510, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
39
|
Long-term consumption of a green/roasted coffee blend positively affects glucose metabolism and insulin resistance in humans. Food Res Int 2016. [DOI: 10.1016/j.foodres.2015.12.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
40
|
|
41
|
Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides 2016; 77:28-37. [PMID: 26344355 DOI: 10.1016/j.peptides.2015.08.013] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/17/2022]
Abstract
Bariatric surgery for obesity has proved to be an extremely effective method of promoting long-term weight reduction with additional beneficial metabolic effects, such as improved glucose tolerance and remission of type 2 diabetes. A range of bariatric procedures are in common use, including gastric banding, sleeve gastrectomy and the Roux-en-Y gastric bypass. Although the mechanisms underlying the efficacy of bariatric surgery are unclear, gastrointestinal and pancreatic peptides are thought to play an important role. The aim of this review is to summarise the effects of different bariatric surgery procedures upon gastrointestinal and pancreatic peptides, including ghrelin, gastrin, cholecystokinin (CCK), glucose-dependent insulinotropic hormone (GIP), glucagon-like peptide 1 (GLP-1), peptide YY (PYY), oxyntomodulin, insulin, glucagon and somatostatin.
Collapse
Affiliation(s)
- Claire L Meek
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Hannah B Lewis
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Frank Reimann
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Fiona M Gribble
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Adrian J Park
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
42
|
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are the known incretin hormones in humans, released predominantly from the enteroendocrine K and L cells within the gut. Their secretion is regulated by a complex of integrated mechanisms involving direct contact for the activation of different chemo-sensors on the brush boarder of K and L cells and several indirect neuro-immuno-hormonal loops. The biological actions of GIP and GLP-1 are fundamental determinants of islet function and blood glucose homeostasis in health and type 2 diabetes. Moreover, there is increasing recognition that GIP and GLP-1 also exert pleiotropic extra-glycaemic actions, which may represent therapeutic targets for human diseases. In this review, we summarise current knowledge of the biology of incretin hormones in health and metabolic disorders and highlight the therapeutic potential of incretin hormones in metabolic regulation.
Collapse
Affiliation(s)
- Tongzhi Wu
- Discipline of Medicine, The University of Adelaide, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Christopher K Rayner
- Discipline of Medicine, The University of Adelaide, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, 5000, Australia.
- Centre of Research Excellence in Translating Nutritional Science into Good Health, The University of Adelaide, Adelaide, Australia.
| | - Michael Horowitz
- Discipline of Medicine, The University of Adelaide, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, 5000, Australia
- Centre of Research Excellence in Translating Nutritional Science into Good Health, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
43
|
Cheng J, Wang Y, Zhang Z, Jin Y, Li Q, Want R, Wang Y, Li X, Xiong Q, Zhang M, Li R, Dai Y. Dominant-negative inhibition of glucose-dependent insulinotropic polypeptide impairs function of β cells in transgenic pigs. J Biomed Res 2015; 29:512-4. [PMID: 26664355 PMCID: PMC4662214 DOI: 10.7555/jbr.29.20150046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/11/2015] [Indexed: 12/04/2022] Open
Affiliation(s)
- JunLin Cheng
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China . ; Department of Clinical Pharmacology, Nanjing First Hospital, Nanjing Medical University , Nanjing, Jiangsu 210012 , China
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Zhengwei Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Yong Jin
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - QianKun Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - RongGen Want
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Yan Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - XiaoKang Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - Qiang Xiong
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - ManLing Zhang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - RongFeng Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| | - YiFan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University , Nanjing, Jiangsu 210029 , China
| |
Collapse
|
44
|
Puddu A, Sanguineti R, Montecucco F, Viviani GL. Effects of High Glucose Levels and Glycated Serum on GIP Responsiveness in the Pancreatic Beta Cell Line HIT-T15. J Diabetes Res 2015; 2015:326359. [PMID: 26221611 PMCID: PMC4499629 DOI: 10.1155/2015/326359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 06/17/2015] [Indexed: 11/17/2022] Open
Abstract
Glucose-dependent insulinotropic peptide (GIP) is an incretin hormone produced in the gastrointestinal tract that stimulates glucose dependent insulin secretion. Impaired incretin response has been documented in diabetic patients and was mainly related to the inability of the pancreatic beta cells to secrete insulin in response to GIP. Advanced Glycation End Products (AGEs) have been shown to play an important role in pancreatic beta cell dysfunction. The aim of this study is to investigate whether the exposure to AGEs can induce GIP resistance in the pancreatic beta cell line HIT-T15. Cells were cultured for 5 days in low (CTR) or high glucose (HG) concentration in the presence of AGEs (GS) to evaluate the expression of GIP receptor (GIPR), the intracellular signaling activated by GIP, and secretion of insulin in response to GIP. The results showed that incubation with GS alone altered intracellular GIP signaling and decreased insulin secretion as compared to CTR. GS in combination with HG reduced the expression of GIPR and PI3K and abrogated GIP-induced AKT phosphorylation and GIP-stimulated insulin secretion. In conclusion, we showed that treatment with GS is associated with the loss of the insulinotropic effect of GIP in hyperglycemic conditions.
Collapse
Affiliation(s)
- Alessandra Puddu
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 16143 Genoa, Italy
| | - Roberta Sanguineti
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 16143 Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 16143 Genoa, Italy
- Division of Cardiology, Department of Medicine, Geneva University Hospitals, Faculty of Medicine, Foundation for Medical Researches, 64 Avenue de la Roseraie, 1211 Geneva, Switzerland
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 Rue Gabrielle-Perret-Gentil, 1205 Geneva, Switzerland
| | - Giorgio Luciano Viviani
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 16143 Genoa, Italy
| |
Collapse
|
45
|
Eriksen M, Jensen DH, Tribler S, Holst JJ, Madsbad S, Krarup T. Reduction of insulinotropic properties of GLP-1 and GIP after glucocorticoid-induced insulin resistance. Diabetologia 2015; 58:920-8. [PMID: 25748606 DOI: 10.1007/s00125-015-3522-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/26/2015] [Indexed: 01/15/2023]
Abstract
AIMS/HYPOTHESIS We evaluated the insulinotropic properties of glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) in healthy individuals at risk of developing type 2 diabetes before and after glucocorticoid-induced insulin resistance. METHODS Nineteen healthy, glucose tolerant, first-degree relatives of type 2 diabetic patients underwent OGTT and 7 mmol/l and 15 mmol/l glucose clamps with concomitant infusions of GLP-1, GIP or NaCl and a final infusion of arginine for determination of maximum beta cell capacity before and after treatment with dexamethasone. In addition, first-phase insulin responses were determined at 7 mmol/l and 15 mmol/l and second-phase insulin responses at 7 mmol/l. RESULTS After dexamethasone treatment, all 19 participants had increased insulin resistance (HOMA-IR and insulin sensitivity index [M/I] values) and 2 h plasma glucose concentrations, while beta cell function indices generally increased according to the increased resistance. First-phase insulin responses induced by GLP-1 and GIP at 7 mmol/l and maximal beta cell secretory capacity did not differ before and after dexamethasone, while second-phase responses to 7 mmol/l and first-phase responses to 15 mmol/l glucose were reduced equally for both hormones. CONCLUSIONS/INTERPRETATION Glucocorticoid-induced insulin resistance in individuals at risk of type 2 diabetes leads to a reduced insulinotropic effect of the incretin hormones. This reduction was not associated with a decrease in the maximal beta cell secretory capacity, indicating that the reduced incretin effect in the developing dysglycaemia of the present experimental model is due to a specific early reduction of the insulinotropic effects of the incretin hormones. TRIAL REGISTRATION Clinicaltrials.gov NCT02235584.
Collapse
Affiliation(s)
- Marie Eriksen
- Department of Endocrinology, Bispebjerg University Hospital, Bispebjerg Bakke 23, DK-2400, Copenhagen, Denmark,
| | | | | | | | | | | |
Collapse
|
46
|
Sharma A, Paliwal G, Upadhyay N, Tiwari A. Therapeutic stimulation of GLP-1 and GIP protein with DPP-4 inhibitors for type-2 diabetes treatment. J Diabetes Metab Disord 2015; 14:15. [PMID: 26473146 PMCID: PMC4607261 DOI: 10.1186/s40200-015-0143-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 02/28/2015] [Indexed: 12/25/2022]
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibition is a new treatment for type-2 diabetes. DPP-4 inhibition increases levels of active GLP-1. GLP-1 enhances insulin secretion and diminishes glucagon secretion, in this manner reducing glucose concentrations in blood. A number of DPP-4 inhibitors are under clinical development. However, the durability and long-term safety of DPP-4 inhibition remain to be established. These synthetic DPP-4 inhibitors are showing some side effects. Herbal medicines are alternative medicine over synthetic drugs that can relieve the patients. Various research studies have been carried all over the world to evaluate the efficacy of herbs in the treatment of Type II diabetes mellitus. For a long time type II diabetes mellitus has been treated orally with herbal medicines, because plant products are frequently prescribed due to their less toxicity than conventional medicines.
Collapse
Affiliation(s)
- Alok Sharma
- School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya (State
Technological University of Madhya Pradesh), Bhopal, India
| | - Geetanjali Paliwal
- School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya (State
Technological University of Madhya Pradesh), Bhopal, India
| | - Nisha Upadhyay
- School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya (State
Technological University of Madhya Pradesh), Bhopal, India
| | - Archana Tiwari
- School of Biotechnology, Rajiv Gandhi Proudyogiki Vishwavidyalaya (State
Technological University of Madhya Pradesh), Bhopal, India
| |
Collapse
|
47
|
Chen S, Okahara F, Osaki N, Shimotoyodome A. Increased GIP signaling induces adipose inflammation via a HIF-1α-dependent pathway and impairs insulin sensitivity in mice. Am J Physiol Endocrinol Metab 2015; 308:E414-25. [PMID: 25537494 DOI: 10.1152/ajpendo.00418.2014] [Citation(s) in RCA: 60] [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] [Indexed: 12/17/2022]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is a gut hormone secreted in response to dietary fat and glucose. The blood GIP level is elevated in obesity and diabetes. GIP stimulates proinflammatory gene expression and impairs insulin sensitivity in cultured adipocytes. In obesity, hypoxia within adipose tissue can induce inflammation. The aims of this study were 1) to examine the proinflammatory effect of increased GIP signaling in adipose tissues in vivo and 2) to clarify the association between GIP and hypoxic signaling in adipose tissue inflammation. We administered GIP intraperitoneally to misty (lean) and db/db (obese) mice and examined adipose tissue inflammation and insulin sensitivity. We also examined the effects of GIP and hypoxia on expression of the GIP receptor (GIPR) gene and proinflammatory genes in 3T3-L1 adipocytes. GIP administration increased monocyte chemoattractant protein-1 (MCP-1) expression and macrophage infiltration into adipose tissue and increased blood glucose in db/db mice. GIPR and hypoxia-inducible factor-1α (HIF-1α) expressions were positively correlated in the adipose tissue in mice. GIPR expression increased dramatically in differentiated adipocytes. GIP treatment of adipocytes increased MCP-1 and interleukin-6 (IL-6) production. Adipocytes cultured either with RAW 264 macrophages or under hypoxia expressed more GIPR and HIF-1α, and GIP treatment increased gene expression of plasminogen activator inhibitor 1 and IL-6. HIF-1α gene silencing diminished both macrophage- and hypoxia-induced GIPR expression and GIP-induced IL-6 expression in adipocytes. Thus, increased GIP signaling plays a significant role in adipose tissue inflammation and thereby insulin resistance in obese mice, and HIF-1α may contribute to this process.
Collapse
Affiliation(s)
- Shu Chen
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Fumiaki Okahara
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | - Noriko Osaki
- Biological Science Laboratories, Kao Corporation, Tochigi, Japan
| | | |
Collapse
|
48
|
Abstract
In normal physiology, glucagon from pancreatic alpha cells plays an important role in maintaining glucose homeostasis via its regulatory effect on hepatic glucose production. Patients with type 2 diabetes suffer from fasting and postprandial hyperglucagonemia, which stimulate hepatic glucose production and, thus, contribute to the hyperglycemia characterizing these patients. Although this has been known for years, research focusing on alpha cell (patho)physiology has historically been dwarfed by research on beta cells and insulin. Today the mechanisms behind type 2 diabetic hyperglucagonemia are still poorly understood. Preclinical and clinical studies have shown that the gastrointestinal hormone glucose-dependent insulinotropic polypeptide (GIP) might play an important role in this pathophysiological phenomenon. Furthermore, it has become apparent that suppression of glucagon secretion or antagonization of the glucagon receptor constitutes potentially effective treatment strategies for patients with type 2 diabetes. In this review, we focus on the regulation of glucagon secretion by the incretin hormones glucagon-like peptide-1 (GLP-1) and GIP. Furthermore, potential advantages and limitations of suppressing glucagon secretion or antagonizing the glucagon receptor, respectively, in the treatment of patients with type 2 diabetes will be discussed.
Collapse
Affiliation(s)
- Asger Lund
- Center for Diabetes Research, Department of Medicine, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | | | | | | |
Collapse
|
49
|
García-Martínez JM, Chocarro-Calvo A, De la Vieja A, García-Jiménez C. Insulin drives glucose-dependent insulinotropic peptide expression via glucose-dependent regulation of FoxO1 and LEF1/β-catenin. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:1141-50. [PMID: 25091498 DOI: 10.1016/j.bbagrm.2014.07.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 07/19/2014] [Accepted: 07/25/2014] [Indexed: 11/15/2022]
Abstract
Minutes after ingestion of fat or carbohydrates, vesicles stored in enteroendocrine cells release their content of incretin peptide hormones that, together with absorbed glucose, enhance insulin secretion by beta-pancreatic cells. Freshly-made incretins must therefore be packed into new vesicles in anticipation of the next meal with cells adjusting new incretin production to be proportional to the level of previous insulin release and absorbed blood glucose. Here we show that insulin stimulates the expression of the major human incretin, glucose-dependent insulinotropic peptide (GIP) in enteroendocrine cells but requires glucose to do it. Akt-dependent release of FoxO1 and glucose-dependent binding of LEF1/β-catenin mediate induction of Gip expression while insulin-induced phosphorylation of β-catenin does not alter its localization or transcriptional activity in enteroendocrine cells. Our results reveal a glucose-regulated feedback loop at the entero-insular axis, where glucose levels determine basal and insulin-induced Gip expression; GIP stimulation of insulin release, physiologically ensures a fine control of glucose homeostasis. How enteroendocrine cells adjust incretin production to replace incretin stores for future use is a key issue because GIP malfunction is linked to all forms of diabetes.
Collapse
Affiliation(s)
- Jose Manuel García-Martínez
- Department of Physiology, Biochemistry and Human Genetics, Faculty of Health Science, Rey Juan Carlos University, 28922 Alcorcon, Madrid, Spain
| | - Ana Chocarro-Calvo
- Department of Physiology, Biochemistry and Human Genetics, Faculty of Health Science, Rey Juan Carlos University, 28922 Alcorcon, Madrid, Spain
| | - Antonio De la Vieja
- Endocrine Tumor Unit (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Custodia García-Jiménez
- Department of Physiology, Biochemistry and Human Genetics, Faculty of Health Science, Rey Juan Carlos University, 28922 Alcorcon, Madrid, Spain.
| |
Collapse
|
50
|
Akash MSH, Rehman K, Chen S. Effects of coffee on type 2 diabetes mellitus. Nutrition 2014; 30:755-63. [DOI: 10.1016/j.nut.2013.11.020] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 11/05/2013] [Accepted: 11/08/2013] [Indexed: 12/13/2022]
|