1
|
Wei L, Ji L, Miao Y, Han X, Li Y, Wang Z, Fu J, Guo L, Su Y, Zhang Y. Constipation in DM are associated with both poor glycemic control and diabetic complications: Current status and future directions. Biomed Pharmacother 2023; 165:115202. [PMID: 37506579 DOI: 10.1016/j.biopha.2023.115202] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Constipation is a major complications of diabetes mellitus. With the accelerating prevalence of diabetes worldwide and an aging population, there is considerable research interest regarding the altered function and structure of the gastrointestinal tract in diabetic patients. Despite current advances in hyperglycemic treatment strategies, the specific pathogenesis of diabetic constipation remains unknown. Patients with constipation, may be reluctant to eat regularly, which may worsen glycemic control and thus worsen symptoms associated with underlying diabetic bowel disease. This paper presents a review of the complex relationship between diabetes and constipation, exploring the morphological alterations and biomechanical remodeling associated with intestinal motility dysfunction, as well as alterations in intestinal neurons, cellular signaling pathways, and oxidative stress. Further studies focusing on new targets that may play a role in the pathogenesis of diabetic constipation may, provide new ideas for the development of novel therapies to treat or even prevent diabetic constipation.
Collapse
Affiliation(s)
- Luge Wei
- Tianjin University of Traditional Chinese Medicine, China.
| | - Lanqi Ji
- Tianjin University of Traditional Chinese Medicine, China
| | - Yulu Miao
- Tianjin University of Traditional Chinese Medicine, China
| | - Xu Han
- Tianjin University of Traditional Chinese Medicine, China
| | - Ying Li
- Tianjin University of Traditional Chinese Medicine, China
| | - Zhe Wang
- Tianjin University of Traditional Chinese Medicine, China
| | - Jiafeng Fu
- Tianjin University of Traditional Chinese Medicine, China
| | - Liuli Guo
- Tianjin University of Traditional Chinese Medicine, China
| | - Yuanyuan Su
- Tianjin University of Traditional Chinese Medicine, China
| | - Yanjun Zhang
- Tianjin University of Traditional Chinese Medicine, China; First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China
| |
Collapse
|
2
|
Abot A, Brochot A, Pomié N, Astre G, Druart C, de Vos WM, Knauf C, Cani PD. Pasteurized Akkermansia muciniphila improves glucose metabolism is linked with increased hypothalamic nitric oxide release. Heliyon 2023; 9:e18196. [PMID: 37501991 PMCID: PMC10368821 DOI: 10.1016/j.heliyon.2023.e18196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
Background and objective Pasteurized Akkermansia muciniphila cells have shown anti-diabetic effects in rodents and human. Although, its primary site of action consists in maintaining the gut barrier function, there are no study exploring if A. muciniphila controls glycemia via a gut to brain axis. Targeting the gut motility represents an alternative pathway to treat hyperglycemia. Here, we tested the impact of pasteurized A. muciniphila on gut motility, gut-brain axis and glucose metabolism. Methods We used mice fed a 45% high-fat (HFD) treated or not with pasteurized A. muciniphila MucT during 12 weeks. We measured the effects of the treatment on body weight gain, glucose metabolism (insulin, glycemia, glucose tolerance), gut contraction and enteric neurotransmitter release, and hypothalamic nitric oxide (NO) release. Results We show that pasteurized A. muciniphila exerts positive effects on different metabolic parameters such as body weight, fat mass, insulin, glycemia and glucose tolerance. This could be explained by the ability of pasteurized A. muciniphila supplementation to decrease duodenal contraction and to increase hypothalamic NO release in HFD mice. Conclusion We demonstrate a novel mode of action of pasteurized A. muciniphila explaining its beneficial impact on the control of glycemia in a preclinical model of type 2 diabetes via gut-brain axis signaling.
Collapse
Affiliation(s)
- Anne Abot
- Enterosys SAS, 31670, Labège, France
| | | | | | | | - Céline Druart
- The Akkermansia Company, 1435, Mont-Saint-Guibert, Belgium
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, 6700, EH Wageningen, the Netherlands
- Human Microbiome Research Program, University of Helsinki, 00014 Helsinki, Finland
| | - Claude Knauf
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Place du Docteur Baylac, CS, 60039, CEDEX 3, 31024, Toulouse, France
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France
| | - Patrice D. Cani
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, France
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO department, WEL Research Institute, Avenue Pasteur, 6, 1300, Wavre, Belgium
| |
Collapse
|
3
|
Wang S, Zeng F, Ma Y, Yu J, Xiang C, Feng X, Wang S, Wang J, Zhao S, Zhu X. Strontium Attenuates Hippocampal Damage via Suppressing Neuroinflammation in High-Fat Diet-Induced NAFLD Mice. Int J Mol Sci 2023; 24:10248. [PMID: 37373395 DOI: 10.3390/ijms241210248] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) leads to hippocampal damage and causes a variety of physiopathological responses, including the induction of endoplasmic reticulum stress (ERS), neuroinflammation, and alterations in synaptic plasticity. As an important trace element, strontium (Sr) has been reported to have antioxidant effects, to have anti-inflammatory effects, and to cause the inhibition of adipogenesis. The present study was undertaken to investigate the protective effects of Sr on hippocampal damage in NAFLD mice in order to elucidate the underlying mechanism of Sr in NAFLD. The mouse model of NAFLD was established by feeding mice a high-fat diet (HFD), and the mice were treated with Sr. In the NAFLD mice, we found that treatment with Sr significantly increased the density of c-Fos+ cells in the hippocampus and inhibited the expression of caspase-3 by suppressing ERS. Surprisingly, the induction of neuroinflammation and the increased expression of inflammatory cytokines in the hippocampus following an HFD were attenuated by Sr treatment. Sr significantly attenuated the activation of microglia and astrocytes induced by an HFD. The expression of phospho-p38, ERK, and NF-κB was consistently significantly increased in the HFD group, and treatment with Sr decreased their expression. Moreover, Sr prevented HFD-induced damage to the ultra-structural synaptic architecture. This study implies that Sr has beneficial effects on repairing the damage to the hippocampus induced by an HFD, revealing that Sr could be a potential candidate for protection from neural damage caused by NAFLD.
Collapse
Affiliation(s)
- Shuai Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Fangyuan Zeng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Jiaojiao Yu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Chenyao Xiang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Xiao Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Songlin Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| |
Collapse
|
4
|
Pandey S, Mangmool S, Parichatikanond W. Multifaceted Roles of GLP-1 and Its Analogs: A Review on Molecular Mechanisms with a Cardiotherapeutic Perspective. Pharmaceuticals (Basel) 2023; 16:836. [PMID: 37375783 DOI: 10.3390/ph16060836] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetes is one of the chronic metabolic disorders which poses a multitude of life-debilitating challenges, including cardiac muscle impairment, which eventually results in heart failure. The incretin hormone glucagon-like peptide-1 (GLP-1) has gained distinct recognition in reinstating glucose homeostasis in diabetes, while it is now largely accepted that it has an array of biological effects in the body. Several lines of evidence have revealed that GLP-1 and its analogs possess cardioprotective effects by various mechanisms related to cardiac contractility, myocardial glucose uptake, cardiac oxidative stress and ischemia/reperfusion injury, and mitochondrial homeostasis. Upon binding to GLP-1 receptor (GLP-1R), GLP-1 and its analogs exert their effects via adenylyl cyclase-mediated cAMP elevation and subsequent activation of cAMP-dependent protein kinase(s) which stimulates the insulin release in conjunction with enhanced Ca2+ and ATP levels. Recent findings have suggested additional downstream molecular pathways stirred by long-term exposure of GLP-1 analogs, which pave the way for the development of potential therapeutic molecules with longer lasting beneficial effects against diabetic cardiomyopathies. This review provides a comprehensive overview of the recent advances in the understanding of the GLP-1R-dependent and -independent actions of GLP-1 and its analogs in the protection against cardiomyopathies.
Collapse
Affiliation(s)
- Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | |
Collapse
|
5
|
The Anti-Seizure Effect of Liraglutide on Ptz-Induced Convulsions Through its Anti-Oxidant and Anti-Inflammatory Properties. Neurochem Res 2023; 48:188-195. [PMID: 36040609 DOI: 10.1007/s11064-022-03736-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 01/11/2023]
Abstract
Epilepsy is a prevalent and frequently devastating neurological disorder defined by recurring spontaneous seizures caused by aberrant electrical activity in the brain. Over ten million people worldwide suffer from drug-resistant epilepsy. This severe condition requires novel treatment approaches. Both oxidative and nitrosative stress are thought to have a role in the etiology of epilepsy. Liraglutide is a glucagon-like peptide-1 (GLP-1) analogue that is used to treat type-2 diabetes mellitus. According to recent studies, Liraglutide also shows neuroprotective properties, improving memory retention and total hippocampus pyramidal neuronal population in mice. The purpose of this investigation was to determine the anti-seizure and anti-oxidative effects of liraglutide in a pentylenetetrazole (PTZ)-induced rat model of epilepsy. 48 rats were randomly assigned to two groups: those who had electroencephalography (EEG) recordings and those who underwent behavioral assessment. Rats received either intraperitoneal (IP) liraglutide at two different dosages (3-6 mg/kg) or a placebo, followed by pentylenetetrazole (IP). To determine if liraglutide has anti-seizure characteristics, we examined seizure activity in rats using EEG, the Racine convulsion scale (RCS), the time of first myoclonic jerk (FMJ), and MDA, SOD, TNF-α, IL-1β and GAD-67 levels. The mean EEG spike wave percentage score was reduced from 75.8% (placebo) to 59.4% (lower-dose) and 41.5% (higher-dose). FMJ had increased from a mean of 70.6 s (placebo) to 181.2 s (lower-dose) and 205.2 s (higher-dose). RCS was reduced from a mean of 5.5 (placebo) to 2.7 (lower-dose) and 2.4 (higher-dose). Liraglutide (3 and 6 mg/kg i.p.) successfully decreased the spike percentages and RCS associated with PTZ induced epilepsy, as well as considerably decreased MDA, TNF-α, IL-1β and elevated SOD, GAD-67 levels in rat brain. Liraglutide significantly decreased seizure activity at both dosages when compared to control, most likely due to its anti-oxidant and anti-inflammatory properties. The potential clinical role of liraglutide as an anti-seizure medication should be further explored.
Collapse
|
6
|
Abot A, Fried S, Cani PD, Knauf C. Reactive Oxygen Species/Reactive Nitrogen Species as Messengers in the Gut: Impact on Physiology and Metabolic Disorders. Antioxid Redox Signal 2022; 37:394-415. [PMID: 34714099 DOI: 10.1089/ars.2021.0100] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: The role of reactive oxygen/nitrogen species as "friend" or "foe" messengers in the whole body is well characterized. Depending on the concentration in the tissue considered, these molecular actors exert beneficial or deleterious impacts leading to a pathological state, as observed in metabolic disorders such as type 2 diabetes and obesity. Recent Advances: Among the tissues impacted by oxidation and inflammation in this pathological state, the intestine is a site of dysfunction that can establish diabetic symptoms, such as alterations in the intestinal barrier, gut motility, microbiota composition, and gut/brain axis communication. In the intestine, reactive oxygen/nitrogen species (from the host and/or microbiota) are key factors that modulate the transition from physiological to pathological signaling. Critical Issues: Controlling the levels of intestinal reactive oxygen/nitrogen species is a complicated balance between positive and negative impacts that is in constant equilibrium. Here, we describe the synthesis and degradation of intestinal reactive oxygen/nitrogen species and their interactions with the host. The development of novel redox-based therapeutics that alter these processes could restore intestinal health in patients with metabolic disorders. Future Directions: Deciphering the mode of action of reactive oxygen/nitrogen species in the gut of obese/diabetic patients could result in a future therapeutic strategy that combines nutritional and pharmacological approaches. Consequently, preventive and curative treatments must take into account one of the first sites of oxidative and inflammatory dysfunctions in the body, that is, the intestine. Antioxid. Redox Signal. 37, 394-415.
Collapse
Affiliation(s)
- Anne Abot
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| | - Steven Fried
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| | - Patrice D Cani
- International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France.,UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, WELBIO, Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Claude Knauf
- Université Paul Sabatier, Toulouse III, INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan, Toulouse, France.,International Research Project (IRP), European Lab "NeuroMicrobiota," Brussels, Belgium and Toulouse, France
| |
Collapse
|
7
|
Wemelle E, Carneiro L, Abot A, Lesage J, Cani PD, Knauf C. Glucose Stimulates Gut Motility in Fasted and Fed Conditions: Potential Involvement of a Nitric Oxide Pathway. Nutrients 2022; 14:nu14102176. [PMID: 35631317 PMCID: PMC9143273 DOI: 10.3390/nu14102176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Type 2 diabetes (T2D) is associated with a duodenal hypermotility in postprandial conditions that favors hyperglycemia and insulin resistance via the gut-brain axis. Enterosynes, molecules produced within the gut with effects on the enteric nervous system, have been recently discovered and pointed to as potential key modulators of the glycemia. Indeed, targeting the enteric nervous system that controls gut motility is now considered as an innovative therapeutic way in T2D to limit intestinal glucose absorption and restore the gut-brain axis to improve insulin sensitivity. So far, little is known about the role of glucose on duodenal contraction in fasted and fed states in normal and diabetic conditions. The aim of the present study was thus to investigate these effects in adult mice. (2) Methods: Gene-expression level of glucose transporters (SGLT-1 and GLUT2) were quantified in the duodenum and jejunum of normal and diabetic mice fed with an HFD. The effect of glucose at different concentrations on duodenal and jejunal motility was studied ex vivo using an isotonic sensor in fasted and fed conditions in both normal chow and HFD mice. (3) Results: Both SGLT1 and GLUT2 expressions were increased in the duodenum (47 and 300%, respectively) and jejunum (75% for GLUT2) of T2D mice. We observed that glucose stimulates intestinal motility in fasted (200%) and fed (400%) control mice via GLUT2 by decreasing enteric nitric oxide release (by 600%), a neurotransmitter that inhibits gut contractions. This effect was not observed in diabetic mice, suggesting that glucose sensing and mechanosensing are altered during T2D. (4) Conclusions: Glucose acts as an enterosyne to control intestinal motility and glucose absorption through the enteric nervous system. Our data demonstrate that GLUT2 and a reduction of NO production could both be involved in this stimulatory contracting effect.
Collapse
Affiliation(s)
- Eve Wemelle
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Place du Docteur Baylac, CS 60039, CEDEX 3, 31024 Toulouse, France; (E.W.); (L.C.)
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, 31024 Toulouse, France
| | - Lionel Carneiro
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Place du Docteur Baylac, CS 60039, CEDEX 3, 31024 Toulouse, France; (E.W.); (L.C.)
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, 31024 Toulouse, France
| | - Anne Abot
- Enterosys SAS, 31670 Labège, France;
| | - Jean Lesage
- Université de Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France;
| | - Patrice D. Cani
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, 31024 Toulouse, France
- UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, WELBIO, Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, 1200 Brussels, Belgium
- Correspondence: (P.D.C.); (C.K.)
| | - Claude Knauf
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Place du Docteur Baylac, CS 60039, CEDEX 3, 31024 Toulouse, France; (E.W.); (L.C.)
- NeuroMicrobiota, International Research Program (IRP) INSERM/UCLouvain, 31024 Toulouse, France
- Correspondence: (P.D.C.); (C.K.)
| |
Collapse
|
8
|
Colon-Perez L, Montesinos J, Monsivais M. The Future of Neuroimaging and Gut-Brain Axis Research for Substance Use Disorders. Brain Res 2022; 1781:147835. [DOI: 10.1016/j.brainres.2022.147835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 12/19/2022]
|
9
|
Wemelle E, Marousez L, de Lamballerie M, Knauf C, Lesage J. High Hydrostatic Pressure Processing of Human Milk Increases Apelin and GLP-1 Contents to Modulate Gut Contraction and Glucose Metabolism in Mice Compared to Holder Pasteurization. Nutrients 2022; 14:nu14010219. [PMID: 35011094 PMCID: PMC8747192 DOI: 10.3390/nu14010219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/29/2021] [Accepted: 01/02/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND High hydrostatic pressure (HHP) processing is a non-thermal method proposed as an alternative to Holder pasteurization (HoP) for the sterilization of human breast milk (BM). HHP preserves numerous milk bioactive factors that are degraded by HoP, but no data are available for milk apelin and glucagon-like peptide 1 (GLP-1), two hormones implicated in the control of glucose metabolism directly and via the gut-brain axis. This study aims to determine the effects of HoP and HHP processing on apelin and GLP-1 concentrations in BM and to test the effect of oral treatments with HoP- and HHP-BM on intestinal contractions and glucose metabolism in adult mice. METHODS Mice were treated by daily oral gavages with HoP- or HHP-BM during one week before intestinal contractions, and glucose tolerance was assessed. mRNA expression of enteric neuronal enzymes known to control intestinal contraction was measured. RESULTS HoP-BM displayed a reduced concentration of apelin and GLP-1, whereas HHP processing preserved these hormones close to their initial levels in raw milk. Chronic HHP-BM administration to mice increased ileal mRNA nNos expression level leading to a decrease in gut contraction associated with improved glucose tolerance. CONCLUSION In comparison to HoP, HPP processing of BM preserves both apelin and GLP-1 and improves glucose tolerance by acting on gut contractions. This study reinforces previous findings demonstrating that HHP processing provides BM with a higher biological value than BM treated by HoP.
Collapse
Affiliation(s)
- Eve Wemelle
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Place du Docteur Baylac, CS 60039, CEDEX 3, 31024 Toulouse, France;
- European Associated Laboratory (EAL) «NeuroMicrobiota», International Research Projects (IRP) INSERM, 1000 Brussels, Belgium
- European Associated Laboratory (EAL) «NeuroMicrobiota», International Research Projects (IRP) INSERM, 31024 Toulouse, France
| | - Lucie Marousez
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France;
| | | | - Claude Knauf
- INSERM U1220, Institut de Recherche en Santé Digestive (IRSD), Université Paul Sabatier, Toulouse III, CHU Purpan, Place du Docteur Baylac, CS 60039, CEDEX 3, 31024 Toulouse, France;
- European Associated Laboratory (EAL) «NeuroMicrobiota», International Research Projects (IRP) INSERM, 1000 Brussels, Belgium
- European Associated Laboratory (EAL) «NeuroMicrobiota», International Research Projects (IRP) INSERM, 31024 Toulouse, France
- Correspondence: (C.K.); (J.L.)
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France;
- Correspondence: (C.K.); (J.L.)
| |
Collapse
|
10
|
Ilchmann-Diounou H, Buleon M, Bacquie V, Theodorou V, Denis C, Menard S. Revisiting definition and assessment of intestinal trans-epithelial passage. Cell Mol Life Sci 2021; 78:8157-8164. [PMID: 34731253 PMCID: PMC8629865 DOI: 10.1007/s00018-021-04000-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/27/2021] [Accepted: 10/18/2021] [Indexed: 10/26/2022]
Abstract
This study aims to remind that Intestinal Passage (IP) measurement is a complex task that cannot be achieved by a unique measure of an orally given exogenous marker in blood or urine. This will be illustrated in the case of NOD mice. Indeed, various methods have been proposed to measure IP. Among them ex vivo measurement in Ussing chambers of luminal to serosal fluxes of exogenous markers and in vivo measurement of exogenous markers in blood or urine after oral gavage are the more commonly used. Even though they are commonly used indifferently, they do not give the same information and can provide contradictory results. Published data showed that diabetic status in female Non Obese Diabetic (NOD) mice increased FD4 concentration in blood after gavage but did not modify FD4 fluxes in Ussing chamber. We observed the same results in our experimental conditions and tracked FD4 concentrations in blood over a kinetic study (Area Under the Curve-AUC). In vivo measurements are a dynamic process and address not only absorption (IP and intestinal surface) but also distribution, metabolism and excretion (ADME). Diabetic status in NOD mice was associated with an increase of intestinal length (absorptive surface), itself positively correlated with AUC of FD4 in blood. We concluded that increased intestinal length induced by diabetic status will extend the absorptive surface and increase FD4 concentration in plasma (in vivo measurement) despite no modification on IP of FD4 (ex vivo measurement). In addition, this study characterized intestinal function in diabetic NOD mice. Diabetic status in NOD female mice increases intestinal length and decreases paracellular IP (FSS) without affecting transcellular IP (HRP, FD4). Histological studies of small and large intestine did not show any modification of intestinal circumference nor villi and crypt size. Finally, diabetic status was not associated with intestinal inflammation (ELISA).
Collapse
Affiliation(s)
- Hanna Ilchmann-Diounou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Marie Buleon
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Disease, Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Valérie Bacquie
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Vassilia Theodorou
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Colette Denis
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Disease, Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Sandrine Menard
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France. .,IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, 31024, Toulouse, France.
| |
Collapse
|
11
|
Aygun H. Exendin-4 increases absence-like seizures and anxiety-depression-like behaviors in WAG/Rij rats. Epilepsy Behav 2021; 123:108246. [PMID: 34385055 DOI: 10.1016/j.yebeh.2021.108246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022]
Abstract
AIM Epilepsy is a neurological condition affecting millions of people worldwide. Glucagon-like peptide-1 (GLP-1) is a gut hormone, and its neuroprotective effect was investigated in previous studies. In this study, the effects of exendin-4, a GLP-1 receptor agonist, were studied in genetic absence epileptic Wistar Albino Glaxo/Rijswijk rats (WAG/Rij). WAG/Rij rat is a genetic model of the absence epilepsy and depression-like comorbidity. METHOD We examined the effects of exendin-4 (10, 50 and 100 µg/kg) on the absence seizures (Electrocorticography [ECoG] recordings), anxiety level (open-field test [OF]), and depression-like levels (forced swimming test [FST]) in the WAG/Rij rats. Basal ECoG recording was performed for all rats. Then, exendin-4 (10, 50 or 100 µg/kg) was administered intraperitoneally and ECoG recording was made for 180 min. After ECoG recording, forced swimming test and open-field test were applied. RESULTS Administration of 10, 50, or 100 µg/kg exendin-4 increased the duration and number of spike-wave discharges (SWDs) considerably without changing the amplitude. The 100 µg/kg dose of exendin-4 was the most effective in increasing the total duration of SWDs. Additionally, all exendin-4 doses increased anxiety level in OF and depression-like level in FST. CONCLUSION Our results showed that exendin-4 increased SWD incidence and anxiety- and depression-like behaviors in the WAG/Rij rats. Besides, it was also found that high doses caused the most proabsence effect.
Collapse
Affiliation(s)
- Hatice Aygun
- Department of Physiology, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Turkey.
| |
Collapse
|
12
|
Pomié C, Servant F, Garidou L, Azalbert V, Waget A, Klopp P, Garret C, Charpentier J, Briand F, Sulpice T, Lelouvier B, Douin-Echinard V, Burcelin R. CX3CR1 regulates gut microbiota and metabolism. A risk factor of type 2 diabetes. Acta Diabetol 2021; 58:1035-1049. [PMID: 33754166 DOI: 10.1007/s00592-021-01682-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/25/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The intestinal microbiota to immune system crosstalk is a major regulator of metabolism and hence metabolic diseases. An impairment of the chemokine receptor CX3CR1, as a key regulator shaping intestinal microbiota under normal chow feeding, could be one of the early events of dysglycemia. METHODS We studied the gut microbiota ecology by sequencing the gut and tissue microbiota. We studied its role in energy metabolism in CX3CR1-deficent and control mice using various bioassays notably the glycemic regulation during fasting and the respiratory quotient as two highly sensitive physiological features. We used antibiotics and prebiotics treatments, and germ free mouse colonization. RESULTS We identify that CX3CR1 disruption impairs gut microbiota ecology and identified a specific signature associated to the genotype. The glycemic control during fasting and the respiratory quotient throughout the day are deeply impaired. A selected four-week prebiotic treatment modifies the dysbiotic microbiota and improves the fasting state glycemic control of the CX3CR1-deficent mice and following a glucose tolerance test. A 4 week antibiotic treatment also improves the glycemic control as well. Eventually, germ free mice colonized with the microbiota from CX3CR1-deficent mice developed glucose intolerance. CONCLUSIONS CX3CR1 is a molecular mechanism in the control of the gut microbiota ecology ensuring the maintenance of a steady glycemia and energy metabolism. Its impairment could be an early mechanism leading to gut microbiota dysbiosis and the onset of metabolic disease.
Collapse
Affiliation(s)
- Celine Pomié
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France
- Evotec, Toulouse, France
| | - Florence Servant
- VAIOMER, Prologue Biotech, Rue Pierre et Marie Curie, Labège Innopole, France
| | - Lucile Garidou
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France
| | - Vincent Azalbert
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France
| | - Aurélie Waget
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France
| | - Pascale Klopp
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France
| | - Céline Garret
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France
| | - Julie Charpentier
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France
| | - Francois Briand
- PHYSIOGENEX, Prologue Biotech, Rue Pierre et Marie Curie, Labège Innopole, France
| | - Thierry Sulpice
- PHYSIOGENEX, Prologue Biotech, Rue Pierre et Marie Curie, Labège Innopole, France
| | - Benjamin Lelouvier
- VAIOMER, Prologue Biotech, Rue Pierre et Marie Curie, Labège Innopole, France
| | - Victorine Douin-Echinard
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France.
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France.
| | - Rémy Burcelin
- Institut National de La Santé et de la Recherche Médicale (INSERM), Toulouse, France.
- Unité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia', Université Paul Sabatier (UPS), 31432, Toulouse Cedex 4, France.
| |
Collapse
|
13
|
The Microbiota and the Gut-Brain Axis in Controlling Food Intake and Energy Homeostasis. Int J Mol Sci 2021; 22:ijms22115830. [PMID: 34072450 PMCID: PMC8198395 DOI: 10.3390/ijms22115830] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity currently represents a major societal and health challenge worldwide. Its prevalence has reached epidemic proportions and trends continue to rise, reflecting the need for more effective preventive measures. Hypothalamic circuits that control energy homeostasis in response to food intake are interesting targets for body-weight management, for example, through interventions that reinforce the gut-to-brain nutrient signalling, whose malfunction contributes to obesity. Gut microbiota-diet interactions might interfere in nutrient sensing and signalling from the gut to the brain, where the information is processed to control energy homeostasis. This gut microbiota-brain crosstalk is mediated by metabolites, mainly short chain fatty acids, secondary bile acids or amino acids-derived metabolites and subcellular bacterial components. These activate gut-endocrine and/or neural-mediated pathways or pass to systemic circulation and then reach the brain. Feeding time and dietary composition are the main drivers of the gut microbiota structure and function. Therefore, aberrant feeding patterns or unhealthy diets might alter gut microbiota-diet interactions and modify nutrient availability and/or microbial ligands transmitting information from the gut to the brain in response to food intake, thus impairing energy homeostasis. Herein, we update the scientific evidence supporting that gut microbiota is a source of novel dietary and non-dietary biological products that may beneficially regulate gut-to-brain communication and, thus, improve metabolic health. Additionally, we evaluate how the feeding time and dietary composition modulate the gut microbiota and, thereby, the intraluminal availability of these biological products with potential effects on energy homeostasis. The review also identifies knowledge gaps and the advances required to clinically apply microbiome-based strategies to improve the gut-brain axis function and, thus, combat obesity.
Collapse
|
14
|
Shandilya A, Mehan S. Dysregulation of IGF-1/GLP-1 signaling in the progression of ALS: potential target activators and influences on neurological dysfunctions. Neurol Sci 2021; 42:3145-3166. [PMID: 34018075 DOI: 10.1007/s10072-021-05328-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/17/2021] [Indexed: 12/31/2022]
Abstract
The prominent causes for motor neuron diseases like ALS are demyelination, immune dysregulation, and neuroinflammation. Numerous research studies indicate that the downregulation of IGF-1 and GLP-1 signaling pathways plays a significant role in the progression of ALS pathogenesis and other neurological disorders. In the current review, we discussed the dysregulation of IGF-1/GLP-1 signaling in neurodegenerative manifestations of ALS like a genetic anomaly, oligodendrocyte degradation, demyelination, glial overactivation, immune deregulation, and neuroexcitation. In addition, the current review reveals the IGF-1 and GLP-1 activators based on the premise that the restoration of abnormal IGF-1/GLP-1 signaling could result in neuroprotection and neurotrophic effects for the clinical-pathological presentation of ALS and other brain diseases. Thus, the potential benefits of IGF-1/GLP-1 signal upregulation in the development of disease-modifying therapeutic strategies may prevent ALS and associated neurocomplications.
Collapse
Affiliation(s)
- Ambika Shandilya
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
15
|
Suzuki S, Aoe S. High β-Glucan Barley Supplementation Improves Glucose Tolerance by Increasing GLP-1 Secretion in Diet-Induced Obesity Mice. Nutrients 2021; 13:nu13020527. [PMID: 33561965 PMCID: PMC7915888 DOI: 10.3390/nu13020527] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/30/2021] [Accepted: 02/04/2021] [Indexed: 01/17/2023] Open
Abstract
The aim of this study was to investigate the underlying mechanism for the improvement of glucose tolerance following intake of high β-glucan barley (HGB) in terms of intestinal metabolism. C57BL/6J male mice were fed a fatty diet supplemented with HGB corresponding to 5% of dietary fiber for 83 days. An oral glucose tolerance test was performed at the end of the experimental period. The concentration of short-chain fatty acids (SCFAs) in the cecum was analyzed by GC–MS (gas chromatography–mass spectrometry). The mRNA expression levels related to L cell function in the ileum were measured by real-time PCR. Glucagon-like peptide-1 (GLP-1) levels in the portal vein and cecal content were assessed by enzyme-linked immunosorbent assay. GLP-1-producing L cells of the ileum were quantified by immunohistochemistry. HGB intake improved glucose tolerance and increased the cecal levels of SCFAs, acetate, and propionate. The number of GLP-1-positive L cells in the HGB group was significantly higher than in the control group. GLP-1 levels in the portal vein and cecal GLP-1 pool size in the HGB group were significantly higher than the control group. In conclusion, we report improved glucose tolerance after HGB intake induced by an increase in L cell number and subsequent rise in GLP-1 secretion.
Collapse
Affiliation(s)
- Sachina Suzuki
- The Institute of Human Culture Studies, Otsuma Women’s University, Chiyoda-ku, Tokyo 102-8357, Japan;
| | - Seiichiro Aoe
- The Institute of Human Culture Studies, Otsuma Women’s University, Chiyoda-ku, Tokyo 102-8357, Japan;
- Studies in Human Life Sciences, Graduate School of Studies in Human Culture, Otsuma Women’s University, Chiyoda-ku, Tokyo 102-8357, Japan
- Correspondence: ; Tel.: +81-3-5275-6048
| |
Collapse
|
16
|
Yap YA, Mariño E. Dietary SCFAs Immunotherapy: Reshaping the Gut Microbiota in Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1307:499-519. [PMID: 32193865 DOI: 10.1007/5584_2020_515] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diet-microbiota related inflammatory conditions such as obesity, autoimmune type 1 diabetes (T1D), type 2 diabetes (T2D), cardiovascular disease (CVD) and gut infections have become a stigma in Western societies and developing nations. This book chapter examines the most relevant pre-clinical and clinical studies about diet-gut microbiota approaches as an alternative therapy for diabetes. We also discuss what we and others have extensively investigated- the power of dietary short-chain fatty acids (SCFAs) technology that naturally targets the gut microbiota as an alternative method to prevent and treat diabetes and its related complications.
Collapse
Affiliation(s)
- Yu Anne Yap
- Infection and Immunity Program, Biomedicine Discovery Institute, Department of Biochemistry, Monash University, Melbourne, VIC, Australia
| | - Eliana Mariño
- Infection and Immunity Program, Biomedicine Discovery Institute, Department of Biochemistry, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
17
|
Jo D, Yoon G, Song J. Role of Exendin-4 in Brain Insulin Resistance, Mitochondrial Function, and Neurite Outgrowth in Neurons under Palmitic Acid-Induced Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10010078. [PMID: 33435277 PMCID: PMC7827489 DOI: 10.3390/antiox10010078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 11/17/2022] Open
Abstract
Glucagon like peptide 1 (GLP-1) is an incretin hormone produced by the gut and brain, and is currently being used as a therapeutic drug for type 2 diabetes and obesity, suggesting that it regulates abnormal appetite patterns, and ameliorates impaired glucose metabolism. Many researchers have demonstrated that GLP-1 agonists and GLP-1 receptor agonists exert neuroprotective effects against brain damage. Palmitic acid (PA) is a saturated fatty acid, and increases the risk of neuroinflammation, lipotoxicity, impaired glucose metabolism, and cognitive decline. In this study, we investigated whether or not Exentin-4 (Ex-4; GLP-1 agonist) inhibits higher production of reactive oxygen species (ROS) in an SH-SY5Y neuronal cell line under PA-induced apoptosis conditions. Moreover, pre-treatment with Ex-4 in SH-SY5Y neuronal cells prevents neural apoptosis and mitochondrial dysfunction through several cellular signal pathways. In addition, insulin sensitivity in neurons is improved by Ex-4 treatment under PA-induced insulin resistance. Additionally, our imaging data showed that neuronal morphology is improved by EX-4 treatment, in spite of PA-induced neuronal damage. Furthermore, we identified that Ex-4 inhibits neuronal damage and enhanced neural complexity, such as neurite length, secondary branches, and number of neurites from soma in PA-treated SH-SY5Y. We observed that Ex-4 significantly increases neural complexity, dendritic spine morphogenesis, and development in PA treated primary cortical neurons. Hence, we suggest that GLP-1 administration may be a crucial therapeutic solution for improving neuropathology in the obese brain.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea; (D.J.); (G.Y.)
- BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-757, Korea
| | - Gwangho Yoon
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea; (D.J.); (G.Y.)
- BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-757, Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea; (D.J.); (G.Y.)
- BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-757, Korea
- Correspondence: ; Tel.:+82-61-379-2706; Fax: +82-61-375-5834
| |
Collapse
|
18
|
Chung JY, Jeong JH, Song J. Resveratrol Modulates the Gut-Brain Axis: Focus on Glucagon-Like Peptide-1, 5-HT, and Gut Microbiota. Front Aging Neurosci 2020; 12:588044. [PMID: 33328965 PMCID: PMC7732484 DOI: 10.3389/fnagi.2020.588044] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Resveratrol is a natural polyphenol that has anti-aging and anti-inflammatory properties against stress condition. It is reported that resveratrol has beneficial functions in various metabolic and central nervous system (CNS) diseases, such as obesity, diabetes, depression, and dementia. Recently, many researchers have emphasized the connection between the brain and gut, called the gut-brain axis, for treating both CNS neuropathologies and gastrointestinal diseases. Based on previous findings, resveratrol is involved in glucagon-like peptide 1 (GLP-1) secreted by intestine L cells, the patterns of microbiome in the intestine, the 5-hydroxytryptamine (5-HT) level, and CNS inflammation. Here, we review recent evidences concerning the relevance and regulatory function of resveratrol in the gut-brain axis from various perspectives. Here, we highlight the necessity for further study on resveratrol's specific mechanism in the gut-brain axis. We present the potential of resveratrol as a natural therapeutic substance for treating both neuropathology and gastrointestinal dysfunction.
Collapse
Affiliation(s)
- Ji Yeon Chung
- Department of Neurology, Chosun University Medical School, Gwangju, South Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, South Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
19
|
Markaki I, Winther K, Catrina SB, Svenningsson P. Repurposing GLP1 agonists for neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:91-112. [PMID: 32854860 DOI: 10.1016/bs.irn.2020.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is a large unmet medical need to find disease modifying therapies against neurodegenerative diseases. This review summarizes data indicating that insulin resistance occurs in neurodegeneration and strategies to normalize insulin sensitivity in neurons may provide neuroprotective actions. In particular, recent preclinical and clinical studies in Parkinson's disease and Alzheimer's disease have indicated that glucagon-like peptide 1 (GLP1) agonism and dipeptidyl peptidase-4 inhibition may exert neuroprotection. Mechanistic insights from these studies and future directions for drug development against neurodegeneration based on GLP1 agonism are discussed.
Collapse
Affiliation(s)
- Ioanna Markaki
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Center of Neurology, Academic Specialist Center, Stockholm, Sweden.
| | - Kristian Winther
- Center of Diabetes, Academic Specialist Center, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Center of Diabetes, Academic Specialist Center, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Center of Neurology, Academic Specialist Center, Stockholm, Sweden; Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
20
|
Role of Glucagon-Like Peptide-1 in Appetite Regulation in Patients with Morbid Obesity and Leptin Resistance. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09864-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
21
|
Knauf C, Abot A, Wemelle E, Cani PD. Targeting the Enteric Nervous System to Treat Metabolic Disorders? "Enterosynes" as Therapeutic Gut Factors. Neuroendocrinology 2020; 110:139-146. [PMID: 31280267 DOI: 10.1159/000500602] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/28/2019] [Indexed: 11/19/2022]
Abstract
The gut-brain axis is of crucial importance for controlling glucose homeostasis. Alteration of this axis promotes the type 2 diabetes (T2D) phenotype (hyperglycaemia, insulin resistance). Recently, a new concept has emerged to demonstrate the crucial role of the enteric nervous system in the control of glycaemia via the hypothalamus. In diabetic patients and mice, modification of enteric neurons activity in the proximal part of the intestine generates a duodenal hyper-contractility that generates an aberrant message from the gut to the brain. In turn, the hypothalamus sends an aberrant efferent message that provokes a state of insulin resistance, which is characteristic of a T2D state. Targeting the enteric nervous system of the duodenum is now recognized as an innovative strategy for treatment of diabetes. By acting in the intestine, bioactive gut molecules that we called "enterosynes" can modulate the function of a specific type of neurons of the enteric nervous system to decrease the contraction of intestinal smooth muscle cells. Here, we focus on the origins of enterosynes (hormones, neurotransmitters, nutrients, microbiota, and immune factors), which could be considered therapeutic factors, and we describe their modes of action on enteric neurons. This unsuspected action of enterosynes is proposed for the treatment of T2D, but it could be applied for other therapeutic solutions that implicate communication between the gut and brain.
Collapse
Affiliation(s)
- Claude Knauf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Toulouse, France,
- NeuroMicrobiota, European Associated Laboratory (EAL) INSERM, Toulouse, France,
| | - Anne Abot
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Toulouse, France
- NeuroMicrobiota, European Associated Laboratory (EAL) INSERM, Toulouse, France
| | - Eve Wemelle
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Toulouse, France
- NeuroMicrobiota, European Associated Laboratory (EAL) INSERM, Toulouse, France
| | - Patrice D Cani
- NeuroMicrobiota, European Associated Laboratory (EAL) INSERM, Toulouse, France
- UCLouvain, Université Catholique de Louvain, WELBIO - Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium
| |
Collapse
|
22
|
Villa-Rodriguez JA, Ifie I, Gonzalez-Aguilar GA, Roopchand DE. The Gastrointestinal Tract as Prime Site for Cardiometabolic Protection by Dietary Polyphenols. Adv Nutr 2019; 10:999-1011. [PMID: 31144710 PMCID: PMC6855987 DOI: 10.1093/advances/nmz038] [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] [Received: 09/18/2018] [Revised: 12/14/2018] [Accepted: 03/19/2019] [Indexed: 02/07/2023] Open
Abstract
Substantial evidence from nutritional epidemiology links polyphenol-rich diets with reduced incidence of chronic disorders; however, biological mechanisms underlying polyphenol-disease relations remain enigmatic. Emerging evidence is beginning to unmask the contribution of the gastrointestinal tract on whole-body energy homeostasis, suggesting that the intestine may be a prime target for intervention and a fundamental site for the metabolic actions of polyphenols. During their transit through the gastrointestinal tract, polyphenols may activate enteric nutrient sensors ensuing appropriate responses from other peripheral organs to regulate metabolic homeostasis. Furthermore, polyphenols can modulate the absorption of glucose, attenuating exaggerated hormonal responses and metabolic imbalances. Polyphenols that escape absorption are metabolized by the gut microbiota and the resulting catabolites may act locally, activating nuclear receptors that control enteric functions such as intestinal permeability. Finally, polyphenols modulate gut microbial ecology, which can have profound effects on cardiometabolic health.
Collapse
Affiliation(s)
- Jose A Villa-Rodriguez
- Institute for Food, Nutrition, and Health, Center for Nutrition, Microbiome, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ,Address correspondence to JAV-R (e-mail: )
| | - Idolo Ifie
- Department of Food Science and Technology, Delta State University, Abraka, Nigeria
| | - Gustavo A Gonzalez-Aguilar
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Sonora, Mexico
| | - Diana E Roopchand
- Institute for Food, Nutrition, and Health, Center for Nutrition, Microbiome, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ,Address correspondence to DER (e-mail: )
| |
Collapse
|
23
|
Effects of GLP-1 Receptor Activation on a Pentylenetetrazole-Kindling Rat Model. Brain Sci 2019; 9:brainsci9050108. [PMID: 31091715 PMCID: PMC6562858 DOI: 10.3390/brainsci9050108] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives: To study the possible anti-seizure and neuroprotective effect of glucagon like peptide 1 (GLP1) analogue (liraglutide) in a pentylenetetrazole (PTZ) induced kindled rat model and its underlying mechanisms. Methods: Thirty Sprague Dawley rats were allocated into 3 equal groups; i) Normal group: normal rats received normal saline, ii) PTZ (kindling) group: received PTZ (50 mg/Kg intraperitoneally (i.p.)) every other day for 2 weeks and iii) PTZ + GLP1 group: same as the PTZ group but rats received liraglutide (75 µg/kg i.p. daily) for 2 weeks before PTZ injection. Seizure severity score, seizure latency and duration were assessed. Also, the expression of caspase-3 (apoptotic marker) and β-catenin (Wnt pathway) by western blotting, markers of oxidative stress (GSH, CAT and MDA) by biochemical assay and the expression of LC3 (marker of autophagy) and heat shock protein 70 (Hsp70) by immunostaining were assessed in hippocampal regions of brain tissues. Results: PTZ caused a significant increase in Racine score and seizure duration with a significant decrease in seizure latency. These effects were associated with a significant increase in MDA, β-catenin, caspase-3, Hsp70 and LC3 in brain tissues (p < 0.05). Meanwhile, liraglutide treatment caused significant attenuation in PTZ-induced seizures, which were associated with significant improvement in markers of oxidative stress, reduction in LC3, caspase-3 and β-catenin and marked increase in Hsp70 in hippocampal regions (p < 0.05). Conclusion: Activation of GLP1R might have anticonvulsant and neuroprotective effects against PTZ-induced epilepsy. These effects could be due to suppression of oxidative stress, apoptosis and autophagy and upregulation of Hsp70.
Collapse
|
24
|
Wen Y, Wu K, Xie Y, Dan W, Zhan Y, Shi Q. Inhibitory effects of glucagon-like peptide-1 receptor on epilepsy. Biochem Biophys Res Commun 2019; 511:79-86. [DOI: 10.1016/j.bbrc.2019.02.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/06/2019] [Indexed: 12/20/2022]
|
25
|
Zhao X, Yan J, Chen K, Song L, Sun B, Wei X. Effects of saccharin supplementation on body weight, sweet receptor mRNA expression and appetite signals regulation in post-weanling rats. Peptides 2018; 107:32-38. [PMID: 30055207 DOI: 10.1016/j.peptides.2018.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/19/2018] [Accepted: 07/22/2018] [Indexed: 01/16/2023]
Abstract
Non-nutritive sweeteners have been considered to promote diet healthfulness by delivering a pleasant sweet taste without calories. We investigated the effects of long term supplementation with drinks containing saccharin on body weight and possible mechanisms of the effects in post-weanling rats. Our results showed that saccharin solution intake increased food intake and energy intake in male rats. In males, saccharin solution intake increased TIR3 mRNA expression in the taste buds and ghrelin receptor mRNA expression both in the taste buds and hypothalamus, whereas no effects were observed in females. These results suggest the effects of saccharin solution exposure on food intake and body weight gain may be different in developmental males and females. In males, peripheral sweet taste receptors and both peripheral and central ghrelin receptors may be involved in the effect of saccharin solution intake to promote food intake and weight gain.
Collapse
Affiliation(s)
- Xiaolin Zhao
- Department of Neonatology, Northwest Women's and Children's Hospital, 1616# Yanxiang Road, Xi'an 710061, PR China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76# W. Yanta Road, Xi'an 710061, PR China
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76# W. Yanta Road, Xi'an 710061, PR China.
| | - Ke Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76# W. Yanta Road, Xi'an 710061, PR China
| | - Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76# W. Yanta Road, Xi'an 710061, PR China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76# W. Yanta Road, Xi'an 710061, PR China
| | - Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76# W. Yanta Road, Xi'an 710061, PR China
| |
Collapse
|
26
|
Sitagliptin enhances the neuroprotective effect of pregabalin against pentylenetetrazole-induced acute epileptogenesis in mice: Implication of oxidative, inflammatory, apoptotic and autophagy pathways. Neurochem Int 2018; 115:11-23. [DOI: 10.1016/j.neuint.2017.10.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/27/2017] [Accepted: 10/10/2017] [Indexed: 12/16/2022]
|
27
|
Abot A, Lucas A, Bautzova T, Bessac A, Fournel A, Le-Gonidec S, Valet P, Moro C, Cani PD, Knauf C. Galanin enhances systemic glucose metabolism through enteric Nitric Oxide Synthase-expressed neurons. Mol Metab 2018; 10:100-108. [PMID: 29428595 PMCID: PMC5985240 DOI: 10.1016/j.molmet.2018.01.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/12/2018] [Accepted: 01/23/2018] [Indexed: 12/14/2022] Open
Abstract
Objective Decreasing duodenal contraction is now considered as a major focus for the treatment of type 2 diabetes. Therefore, identifying bioactive molecules able to target the enteric nervous system, which controls the motility of intestinal smooth muscle cells, represents a new therapeutic avenue. For this reason, we chose to study the impact of oral galanin on this system in diabetic mice. Methods Enteric neurotransmission, duodenal contraction, glucose absorption, modification of gut–brain axis, and glucose metabolism (glucose tolerance, insulinemia, glucose entry in tissue, hepatic glucose metabolism) were assessed. Results We show that galanin, a neuropeptide expressed in the small intestine, decreases duodenal contraction by stimulating nitric oxide release from enteric neurons. This is associated with modification of hypothalamic nitric oxide release that favors glucose uptake in metabolic tissues such as skeletal muscle, liver, and adipose tissue. Oral chronic gavage with galanin in diabetic mice increases insulin sensitivity, which is associated with an improvement of several metabolic parameters such as glucose tolerance, fasting blood glucose, and insulin. Conclusion Here, we demonstrate that oral galanin administration improves glucose homeostasis via the enteric nervous system and could be considered a therapeutic potential for the treatment of T2D. Targeting the enteric nervous system (ENS) is an innovative solution to treat diabetes. The ENS controls duodenal contractions to modulate glycemia via the gut–brain axis. ENS/contractions are targeted by the neuropeptide galanin in the intestine. Oral galanin treatment decreases duodenal hyper-contractility in diabetic mice. Oral galanin restores the gut–brain axis to improve glycemia in diabetic mice.
Collapse
Affiliation(s)
- Anne Abot
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), CHU Purpan, Place du Docteur Baylac, CS 60039, 31024 Toulouse Cedex 3, France; NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL, France
| | - Alexandre Lucas
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Université Paul Sabatier, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), CHU Rangueil, 1 Avenue Jean Poulhès, BP84225, 31432 Toulouse Cedex 4, France
| | - Tereza Bautzova
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), CHU Purpan, Place du Docteur Baylac, CS 60039, 31024 Toulouse Cedex 3, France; NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL, France
| | - Arnaud Bessac
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), CHU Purpan, Place du Docteur Baylac, CS 60039, 31024 Toulouse Cedex 3, France; NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL, France
| | - Audren Fournel
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), CHU Purpan, Place du Docteur Baylac, CS 60039, 31024 Toulouse Cedex 3, France; NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL, France
| | - Sophie Le-Gonidec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Université Paul Sabatier, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), CHU Rangueil, 1 Avenue Jean Poulhès, BP84225, 31432 Toulouse Cedex 4, France
| | - Philippe Valet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Université Paul Sabatier, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), CHU Rangueil, 1 Avenue Jean Poulhès, BP84225, 31432 Toulouse Cedex 4, France
| | - Cédric Moro
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Université Paul Sabatier, UPS, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), CHU Rangueil, 1 Avenue Jean Poulhès, BP84225, 31432 Toulouse Cedex 4, France
| | - Patrice D Cani
- NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL, France; Université Catholique de Louvain (UCL), Louvain Drug Research Institute, LDRI, Metabolism and Nutrition Research Group, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Avenue E. Mounier, 73 B1.73.11, B-1200, Brussels, Belgium.
| | - Claude Knauf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier, UPS, Institut de Recherche en Santé Digestive et Nutrition (IRSD), CHU Purpan, Place du Docteur Baylac, CS 60039, 31024 Toulouse Cedex 3, France; NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL, France.
| |
Collapse
|
28
|
Qin N, Hu X, Li S, Wang J, Li Z, Li D, Xu F, Gao M, Hua H. Hypoglycemic effect of silychristin A from Silybum marianum fruit via protecting pancreatic islet β cells from oxidative damage and inhibiting α -glucosidase activity in vitro and in rats with type 1 diabetes. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.09.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
29
|
Recombinant Incretin-Secreting Microbe Improves Metabolic Dysfunction in High-Fat Diet Fed Rodents. Sci Rep 2017; 7:13523. [PMID: 29051554 PMCID: PMC5648875 DOI: 10.1038/s41598-017-14010-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/02/2017] [Indexed: 01/16/2023] Open
Abstract
The gut hormone glucagon-like peptide (GLP)-1 and its analogues represent a new generation of anti-diabetic drugs, which have also demonstrated propensity to modulate host lipid metabolism. Despite this, drugs of this nature are currently limited to intramuscular administration routes due to intestinal degradation. The aim of this study was to design a recombinant microbial delivery vector for a GLP-1 analogue and assess the efficacy of the therapeutic in improving host glucose, lipid and cholesterol metabolism in diet induced obese rodents. Diet-induced obese animals received either Lactobacillus paracasei NFBC 338 transformed to express a long-acting analogue of GLP-1 or the isogenic control microbe which solely harbored the pNZ44 plasmid. Short-term GLP-1 microbe intervention in rats reduced serum low-density lipoprotein cholesterol, triglycerides and triglyceride-rich lipoprotein cholesterol substantially. Conversely, extended GLP-1 microbe intervention improved glucose-dependent insulin secretion, glucose metabolism and cholesterol metabolism, compared to the high-fat control group. Interestingly, the microbe significantly attenuated the adiposity associated with the model and altered the serum lipidome, independently of GLP-1 secretion. These data indicate that recombinant incretin-secreting microbes may offer a novel and safe means of managing cholesterol metabolism and diet induced dyslipidaemia, as well as insulin sensitivity in metabolic dysfunction.
Collapse
|
30
|
Fenugreek Compound (N55) Lowers Plasma Glucose through the Enhancement of Response of Physiological Glucagon-like peptide-1. Sci Rep 2017; 7:12265. [PMID: 28947828 PMCID: PMC5613011 DOI: 10.1038/s41598-017-12290-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 09/04/2017] [Indexed: 12/01/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) analogues are approved for treating type 2 diabetes, but are known to activate GLP-1R signaling globally and constitutively. Active compound N55, previously isolated from fenugreek, enhances the potency of GLP-1 without activating GLP-1R. Here we investigated if N55 lowers plasma glucose base on physiological levels of GLP-1. N55 was found to dose-dependently lower plasma glucose in non-fasted mice but not in the fasted mice, with the effect attenuated by GLP-1R antagonist exendin-(9–39) (Ex-9). On the other hand, when co-administered with dipeptidyl peptidase-IV (DPP4) -resistant [Aib8]-GLP-1(7–36) amide (GLP-1′), hypoglycemic response to N55 was observed in the fasted mice. This enhancement was also found to display dose dependency. N55 enhancement of the hypoglycemic and insulinotropic action of GLP-1′ was eliminated upon Ex-9 treatment. Both exendin-4 (Ex-4) and DPP4-resistant GLP-1 mutant peptide ([Aib8, E22, E30]-GLP-1(7–36) amide) activated GLP-1R and improved glucose tolerance but the enhancement effect of N55 was not observed in vivo or in vitro. In conclusions, N55 lowers plasma glucose according to prandial status by enhancing the response of physiological levels of GLP-1 and is much less likely to disrupt tight regulation of GLP-1R signaling as compare to GLP-1 analogues.
Collapse
|
31
|
Beta-glucans and cancer: The influence of inflammation and gut peptide. Eur J Med Chem 2017; 142:486-492. [PMID: 28964548 DOI: 10.1016/j.ejmech.2017.09.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/05/2017] [Accepted: 09/07/2017] [Indexed: 12/13/2022]
Abstract
Dietary β-glucans are soluble fibers with potentially health-promoting effects. Gut peptides are important signals in the regulation of energy and glucose homeostasis. This article reviews the effects of different enriched β-glucan food consumption on immune responses, inflammation, gut hormone and cancer. Gut hormones are influenced by enriched β-glucan food consumption and levels of such peptide as YY, ghrelin, glucagon-like peptide 1 and 2 in humans influence serum glucose concentration as well as innate and adaptive immunity. Cancer cell development is also regulated by obesity and glucose dishomeostasy that are influenced by β-glucan food consumption that in turn regulated gut hormones.
Collapse
|
32
|
Grasset E, Puel A, Charpentier J, Collet X, Christensen JE, Tercé F, Burcelin R. A Specific Gut Microbiota Dysbiosis of Type 2 Diabetic Mice Induces GLP-1 Resistance through an Enteric NO-Dependent and Gut-Brain Axis Mechanism. Cell Metab 2017; 25:1075-1090.e5. [PMID: 28467926 DOI: 10.1016/j.cmet.2017.04.013] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 02/01/2017] [Accepted: 04/13/2017] [Indexed: 12/22/2022]
Abstract
Glucagon-like peptide-1 (GLP-1)-based therapies control glycemia in type 2 diabetic (T2D) patients. However, in some patients the treatment must be discontinued, defining a state of GLP-1 resistance. In animal models we identified a specific set of ileum bacteria impairing the GLP-1-activated gut-brain axis for the control of insulin secretion and gastric emptying. Using prediction algorithms, we identified bacterial pathways related to amino acid metabolism and transport system modules associated to GLP-1 resistance. The conventionalization of germ-free mice demonstrated their role in enteric neuron biology and the gut-brain-periphery axis. Altogether, insulin secretion and gastric emptying require functional GLP-1 receptor and neuronal nitric oxide synthase in the enteric nervous system within a eubiotic gut microbiota environment. Our data open a novel route to improve GLP-1-based therapies.
Collapse
Affiliation(s)
- Estelle Grasset
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: Intestinal Risk Factors, Diabetes, Dyslipidemia, Heart Failure, F-31432 Toulouse, Cedex 4, France
| | - Anthony Puel
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: Intestinal Risk Factors, Diabetes, Dyslipidemia, Heart Failure, F-31432 Toulouse, Cedex 4, France
| | - Julie Charpentier
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: Intestinal Risk Factors, Diabetes, Dyslipidemia, Heart Failure, F-31432 Toulouse, Cedex 4, France
| | - Xavier Collet
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: Intestinal Risk Factors, Diabetes, Dyslipidemia, Heart Failure, F-31432 Toulouse, Cedex 4, France
| | - Jeffrey E Christensen
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: Intestinal Risk Factors, Diabetes, Dyslipidemia, Heart Failure, F-31432 Toulouse, Cedex 4, France
| | - François Tercé
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: Intestinal Risk Factors, Diabetes, Dyslipidemia, Heart Failure, F-31432 Toulouse, Cedex 4, France
| | - Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France; Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: Intestinal Risk Factors, Diabetes, Dyslipidemia, Heart Failure, F-31432 Toulouse, Cedex 4, France.
| |
Collapse
|
33
|
Gaykema RP, Newmyer BA, Ottolini M, Raje V, Warthen DM, Lambeth PS, Niccum M, Yao T, Huang Y, Schulman IG, Harris TE, Patel MK, Williams KW, Scott MM. Activation of murine pre-proglucagon-producing neurons reduces food intake and body weight. J Clin Invest 2017; 127:1031-1045. [PMID: 28218622 DOI: 10.1172/jci81335] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 12/30/2016] [Indexed: 12/14/2022] Open
Abstract
Peptides derived from pre-proglucagon (GCG peptides) act in both the periphery and the CNS to change food intake, glucose homeostasis, and metabolic rate while playing a role in anxiety behaviors and physiological responses to stress. Although the actions of GCG peptides produced in the gut and pancreas are well described, the role of glutamatergic GGC peptide-secreting hindbrain neurons in regulating metabolic homeostasis has not been investigated. Here, we have shown that chemogenetic stimulation of GCG-producing neurons reduces metabolic rate and food intake in fed and fasted states and suppresses glucose production without an effect on glucose uptake. Stimulation of GCG neurons had no effect on corticosterone secretion, body weight, or conditioned taste aversion. In the diet-induced obese state, the effects of GCG neuronal stimulation on gluconeogenesis were lost, while the food intake-lowering effects remained, resulting in reductions in body weight and adiposity. Our work suggests that GCG peptide-expressing neurons can alter feeding, metabolic rate, and glucose production independent of their effects on hypothalamic pituitary-adrenal (HPA) axis activation, aversive conditioning, or insulin secretion. We conclude that GCG neurons likely stimulate separate populations of downstream cells to produce a change in food intake and glucose homeostasis and that these effects depend on the metabolic state of the animal.
Collapse
|
34
|
Nance KD, Days EL, Weaver CD, Coldren A, Farmer TD, Cho HP, Niswender CM, Blobaum AL, Niswender KD, Lindsley CW. Discovery of a Novel Series of Orally Bioavailable and CNS Penetrant Glucagon-like Peptide-1 Receptor (GLP-1R) Noncompetitive Antagonists Based on a 1,3-Disubstituted-7-aryl-5,5-bis(trifluoromethyl)-5,8-dihydropyrimido[4,5-d]pyrimidine-2,4(1H,3H)-dione Core. J Med Chem 2017; 60:1611-1616. [PMID: 28103022 DOI: 10.1021/acs.jmedchem.6b01706] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A duplexed, functional multiaddition high throughput screen and subsequent optimization effort identified the first orally bioavailable and CNS penetrant glucagon-like peptide-1 receptor (GLP-1R) noncompetitive antagonist. Antagonist 5d not only blocked exendin-4-stimulated insulin release in islets but also lowered insulin levels while increasing blood glucose in vivo.
Collapse
Affiliation(s)
- Kellie D Nance
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Emily L Days
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - C David Weaver
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Anastasia Coldren
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Tiffany D Farmer
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Hyekyung P Cho
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Colleen M Niswender
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Anna L Blobaum
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Kevin D Niswender
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Craig W Lindsley
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, ‡Department of Pharmacology, §Vanderbilt Center for Neuroscience Drug Discovery, ∥Vanderbilt Institute of Chemical Biology, and ⊥Vanderbilt Diabetes Center, Vanderbilt University School of Medicine , Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare System, ∇Department of Chemistry, and ○Vanderbilt Kennedy Center, Vanderbilt University , Nashville, Tennessee 37232, United States
| |
Collapse
|
35
|
Fournel A, Drougard A, Duparc T, Marlin A, Brierley SM, Castro J, Le-Gonidec S, Masri B, Colom A, Lucas A, Rousset P, Cenac N, Vergnolle N, Valet P, Cani PD, Knauf C. Apelin targets gut contraction to control glucose metabolism via the brain. Gut 2017; 66:258-269. [PMID: 26565000 PMCID: PMC5284480 DOI: 10.1136/gutjnl-2015-310230] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/02/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The gut-brain axis is considered as a major regulatory checkpoint in the control of glucose homeostasis. The detection of nutrients and/or hormones in the duodenum informs the hypothalamus of the host's nutritional state. This process may occur via hypothalamic neurons modulating central release of nitric oxide (NO), which in turn controls glucose entry into tissues. The enteric nervous system (ENS) modulates intestinal contractions in response to various stimuli, but the importance of this interaction in the control of glucose homeostasis via the brain is unknown. We studied whether apelin, a bioactive peptide present in the gut, regulates ENS-evoked contractions, thereby identifying a new physiological partner in the control of glucose utilisation via the hypothalamus. DESIGN We measured the effect of apelin on electrical and mechanical duodenal responses via telemetry probes and isotonic sensors in normal and obese/diabetic mice. Changes in hypothalamic NO release, in response to duodenal contraction modulated by apelin, were evaluated in real time with specific amperometric probes. Glucose utilisation in tissues was measured with orally administrated radiolabeled glucose. RESULTS In normal and obese/diabetic mice, glucose utilisation is improved by the decrease of ENS/contraction activities in response to apelin, which generates an increase in hypothalamic NO release. As a consequence, glucose entry is significantly increased in the muscle. CONCLUSIONS Here, we identify a novel mode of communication between the intestine and the hypothalamus that controls glucose utilisation. Moreover, our data identified oral apelin administration as a novel potential target to treat metabolic disorders.
Collapse
Affiliation(s)
- Audren Fournel
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| | - Anne Drougard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| | - Thibaut Duparc
- NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Catholique de Louvain (UCL), Louvain Drug Research Institute, LDRI, Metabolism and Nutrition research group, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Brussels, Belgium
| | - Alysson Marlin
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| | - Stuart M Brierley
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australia Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia,Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia,Discipline of Physiology, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Joel Castro
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of Adelaide, South Australia Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Sophie Le-Gonidec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| | - Bernard Masri
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1037, Centre de Recherches en Cancérologie de Toulouse (CRCT), CHU Rangueil, Toulouse, Cedex 4, France
| | - André Colom
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| | - Alexandre Lucas
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| | - Perrine Rousset
- Université Paul Sabatier, Toulouse, France,Institut National de la Santé et de la Recherche Médicale (INSERM), U1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), CHU Purpan, Toulouse, Cedex 03, France
| | - Nicolas Cenac
- Université Paul Sabatier, Toulouse, France,Institut National de la Santé et de la Recherche Médicale (INSERM), U1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), CHU Purpan, Toulouse, Cedex 03, France
| | - Nathalie Vergnolle
- Université Paul Sabatier, Toulouse, France,Institut National de la Santé et de la Recherche Médicale (INSERM), U1043, Centre de Physiopathologie de Toulouse Purpan (CPTP), CHU Purpan, Toulouse, Cedex 03, France
| | - Philippe Valet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| | - Patrice D Cani
- NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Catholique de Louvain (UCL), Louvain Drug Research Institute, LDRI, Metabolism and Nutrition research group, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Brussels, Belgium
| | - Claude Knauf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse Cedex 4, France,NeuroMicrobiota, European Associated Laboratory (EAL) INSERM/UCL,Université Paul Sabatier, Toulouse, France
| |
Collapse
|
36
|
Cariou B. Pleiotropic effects of insulin and GLP-1 receptor agonists: Potential benefits of the association. DIABETES & METABOLISM 2016; 41:6S28-6S35. [PMID: 26774017 DOI: 10.1016/s1262-3636(16)30006-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The combination of basal insulin and glucagon-like peptide-1 receptor agonists (GLP-1RAs) is an emerging option for patients with type 2 diabetes (T2D). GLP-1RAs have been shown to improve glycaemic control with a low risk of hypoglycaemia and to promote body weight loss. However, GLP-1 receptors (GLP-1Rs) are widely expressed in extrapancreatic tissues and could sustain pleiotropic actions of GLP-1RAs beyond glycaemic control. The underlying molecular mechanisms maintaining these extrapancreatic actions of GLP-1 are complex, and involve GLP-1R signalling in both the brain and several peripheral tissues. The present review focuses specifically on the role of GLP-1RAs in the cardiovascular system and liver. Preclinical data in rodents and pilot studies in humans suggest that GLP-1RAs may have potential beneficial effects on heart function, blood pressure, postprandial lipaemia, liver steatosis and non-alcoholic steatohepatitis (NASH). Long-term studies are now warranted to determine the safety and clinical relevance of the association between insulin and GLP-1RAs in T2D.
Collapse
Affiliation(s)
- B Cariou
- Clinique d'Endocrinologie, l'institut du thorax, CHU de Nantes, Nantes, F-44000 France; INSERM, UMR1087, l'institut du thorax, Nantes, F-44000 France; Faculté de Médecine, Université de Nantes, Nantes, F-44000 France.
| |
Collapse
|
37
|
Fournel A, Marlin A, Abot A, Pasquio C, Cirillo C, Cani PD, Knauf C. Glucosensing in the gastrointestinal tract: Impact on glucose metabolism. Am J Physiol Gastrointest Liver Physiol 2016; 310:G645-58. [PMID: 26939867 PMCID: PMC4867329 DOI: 10.1152/ajpgi.00015.2016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/25/2016] [Indexed: 01/31/2023]
Abstract
The gastrointestinal tract is an important interface of exchange between ingested food and the body. Glucose is one of the major dietary sources of energy. All along the gastrointestinal tube, e.g., the oral cavity, small intestine, pancreas, and portal vein, specialized cells referred to as glucosensors detect variations in glucose levels. In response to this glucose detection, these cells send hormonal and neuronal messages to tissues involved in glucose metabolism to regulate glycemia. The gastrointestinal tract continuously communicates with the brain, especially with the hypothalamus, via the gut-brain axis. It is now well established that the cross talk between the gut and the brain is of crucial importance in the control of glucose homeostasis. In addition to receiving glucosensing information from the gut, the hypothalamus may also directly sense glucose. Indeed, the hypothalamus contains glucose-sensitive cells that regulate glucose homeostasis by sending signals to peripheral tissues via the autonomous nervous system. This review summarizes the mechanisms by which glucosensors along the gastrointestinal tract detect glucose, as well as the results of such detection in the whole body, including the hypothalamus. We also highlight how disturbances in the glucosensing process may lead to metabolic disorders such as type 2 diabetes. A better understanding of the pathways regulating glucose homeostasis will further facilitate the development of novel therapeutic strategies for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Audren Fournel
- 1NeuroMicrobiota, European Associated Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut de Recherche en Santé Digestive (IRSD), Toulouse, France;
| | - Alysson Marlin
- 1NeuroMicrobiota, European Associated Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut de Recherche en Santé Digestive (IRSD), Toulouse, France;
| | - Anne Abot
- 1NeuroMicrobiota, European Associated Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut de Recherche en Santé Digestive (IRSD), Toulouse, France;
| | - Charles Pasquio
- 1NeuroMicrobiota, European Associated Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut de Recherche en Santé Digestive (IRSD), Toulouse, France;
| | - Carla Cirillo
- 2Laboratory for Enteric NeuroScience (LENS), University of Leuven, Leuven, Belgium; and
| | - Patrice D. Cani
- 3NeuroMicrobiota, European Associated Laboratory, Université Catholique de Louvain (UCL), Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Brussels, Belgium
| | - Claude Knauf
- NeuroMicrobiota, European Associated Laboratory, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut de Recherche en Santé Digestive (IRSD), Toulouse, France;
| |
Collapse
|
38
|
Richards L, Li M, van Esch B, Garssen J, Folkerts G. The effects of short-chain fatty acids on the cardiovascular system. PHARMANUTRITION 2016. [DOI: 10.1016/j.phanu.2016.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Katsurada K, Yada T. Neural effects of gut- and brain-derived glucagon-like peptide-1 and its receptor agonist. J Diabetes Investig 2016; 7 Suppl 1:64-9. [PMID: 27186358 PMCID: PMC4854507 DOI: 10.1111/jdi.12464] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022] Open
Abstract
Glucagon‐like peptide‐1 (GLP‐1) is derived from both the enteroendocrine L cells and preproglucagon‐expressing neurons in the nucleus tractus solitarius (NTS) of the brain stem. As GLP‐1 is cleaved by dipeptidyl peptidase‐4 yielding a half‐life of less than 2 min, it is plausible that the gut‐derived GLP‐1, released postprandially, exerts its effects on the brain mainly by interacting with vagal afferent neurons located at the intestinal or hepatic portal area. GLP‐1 neurons in the NTS widely project in the central nervous system and act as a neurotransmitter. One of the physiological roles of brain‐derived GLP‐1 is restriction of feeding. GLP‐1 receptor agonists have recently been used to treat type 2 diabetic patients, and have been shown to exhibit pleiotropic effects beyond incretin action, which involve brain functions. GLP‐1 receptor agonist administered in the periphery is stable because of its resistance to dipeptidyl peptidase‐4, and is highly likely to act on the brain by passing through the blood–brain barrier (BBB), as well as interacting with vagal afferent nerves. Central actions of GLP‐1 have various roles including regulation of feeding, weight, glucose and lipid metabolism, cardiovascular functions, cognitive functions, and stress and emotional responses. In the present review, we focus on the source of GLP‐1 and the pathway by which peripheral GLP‐1 informs the brain, and then discuss recent findings on the central effects of GLP‐1 and GLP‐1 receptor agonists.
Collapse
Affiliation(s)
- Kenichi Katsurada
- Division of Integrative PhysiologyDepartment of PhysiologyJichi Medical University School of MedicineShimotsukeTochigiJapan; Division of Cardiovascular MedicineDepartment of Internal MedicineJichi Medical University School of MedicineShimotsukeTochigiJapan
| | - Toshihiko Yada
- Division of Integrative Physiology Department of Physiology Jichi Medical University School of Medicine Shimotsuke Tochigi Japan
| |
Collapse
|
40
|
Koshal P, Kumar P. Neurochemical modulation involved in the beneficial effect of liraglutide, GLP-1 agonist on PTZ kindling epilepsy-induced comorbidities in mice. Mol Cell Biochem 2016; 415:77-87. [PMID: 26965494 DOI: 10.1007/s11010-016-2678-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/04/2016] [Indexed: 12/15/2022]
Abstract
Epilepsy is a neurological disorder which occurs due to excessive firing of excitatory neurons in specific region of brain and associated with cognitive impairment and depression. GLP-1 has been reported to maintain hyperexcitability of neurons. Therefore, this study was designed to investigate the neuroprotective effect of liraglutide, GLP-1 analogue in PTZ kindling epilepsy-induced comorbidities and neurochemical alteration in mice. Male albino mice were administered PTZ (35 mg/kg) on every alternate day up to 29th days and challenge test was performed on 33rd day. From 1st day liraglutide (75 and 150 µg/kg) and diazepam (3 mg/kg) were administered up to 33rd day, 30 min prior to PTZ treatment. On 30th day animals were trained on elevated plus maze and passive shock avoidance paradigm and retention was recorded on 31st and 33rd day. On 32nd day tail suspension test was performed. Animals were sacrificed on 34th day for biochemical (LPO, GSH, and nitrite) and neurotransmitters (GABA, glutamate, DA, NE, 5-HT and their metabolites) estimation. Chronic treatment with PTZ developed generalized tonic-clonic seizures, reduced cognitive skills, increased oxidative stress and alteration in the level of neurotransmitters. Pre-treatment with liraglutide (75 and 150 μg/kg) significantly prevented the seizure severity, restored behavioural activity, oxidative defence enzymes, and altered level of neurochemicals in mice brain. The protective effect of liraglutide is attributed to restoration of altered level of GABA, glutamate, DA, NE, and 5-HT by the up-regulation of GLP-1Rs in mice brain.
Collapse
Affiliation(s)
- Prashant Koshal
- Department of Pharmacology, ISF College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga, Punjab, 142001, India
| | - Puneet Kumar
- Department of Pharmacology, ISF College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga, Punjab, 142001, India.
| |
Collapse
|
41
|
Patterson E, Ryan PM, Cryan JF, Dinan TG, Ross RP, Fitzgerald GF, Stanton C. Gut microbiota, obesity and diabetes. Postgrad Med J 2016; 92:286-300. [PMID: 26912499 DOI: 10.1136/postgradmedj-2015-133285] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023]
Abstract
The central role of the intestinal microbiota in the progression and, equally, prevention of metabolic dysfunction is becoming abundantly apparent. The symbiotic relationship between intestinal microbiota and host ensures appropriate development of the metabolic system in humans. However, disturbances in composition and, in turn, functionality of the intestinal microbiota can disrupt gut barrier function, a trip switch for metabolic endotoxemia. This low-grade chronic inflammation, brought about by the influx of inflammatory bacterial fragments into circulation through a malfunctioning gut barrier, has considerable knock-on effects for host adiposity and insulin resistance. Conversely, recent evidence suggests that there are certain bacterial species that may interact with host metabolism through metabolite-mediated stimulation of enteric hormones and other systems outside of the gastrointestinal tract, such as the endocannabinoid system. When the abundance of these keystone species begins to decline, we see a collapse of the symbiosis, reflected in a deterioration of host metabolic health. This review will investigate the intricate axis between the microbiota and host metabolism, while also addressing the promising and novel field of probiotics as metabolic therapies.
Collapse
Affiliation(s)
- Elaine Patterson
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Paul M Ryan
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Co. Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Psychiatry and Neurobehavioural Science, University College Cork, Co. Cork, Ireland
| | - R Paul Ross
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland College of Science, Engineering and Food Science, University College Cork, Co. Cork, Ireland
| | - Gerald F Fitzgerald
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
42
|
Tong J, Ma B, Ge L, Mo Q, Zhou G, He J, Wang Y. Dicaffeoylquinic Acid-Enriched Fraction of Cichorium glandulosum Seeds Attenuates Experimental Type 1 Diabetes via Multipathway Protection. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:10791-802. [PMID: 26586022 DOI: 10.1021/acs.jafc.5b04552] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Chicory has a major geographical presence in Europe and Asia. Cichorium glandulosum Boiss. et Huet, a genus Cichorium, is used for medicinal and food purposes in Asia. In this study, a dicaffeoylquinic acid-enriched fraction of C. glandulosum seeds n-BuOH fraction (CGSB) could ameliorate type 1 diabetes mellitus (T1DM) in streptozotocin (STZ)-induced diabetic mice with continuous administration for 2 weeks. CGSB treatment showed significantly higher plasma insulin levels but lower free fatty acids in adipose tissue and liver. Moreover, CGSB improved pancreatic islet mass. In vitro, different fractions of C. glandulosum seed (CGS) induced the differentiation of 3T3-L1 preadipocytes. The mRNA level for peroxisome proliferator-activated receptor alpha increased in high glucose treatment group in HepG2 cells, while CGSB significantly down-regulated the mRNA expression. The main compound of CGSB, 3,5-dicaffeoylquinic acid, was isolated and identified, which exhibited α-glucosidase inhibitory activity. These findings demonstrated that CGSB attenuated experimental T1DM via multipathway protection.
Collapse
Affiliation(s)
- Jing Tong
- Institute of TCM and Natural Products, School of Pharmaceutical Sciences, Wuhan University , Wuhan 430071, P. R. China
| | - Bingxin Ma
- Institute of TCM and Natural Products, School of Pharmaceutical Sciences, Wuhan University , Wuhan 430071, P. R. China
| | - Lanlan Ge
- Institute of TCM and Natural Products, School of Pharmaceutical Sciences, Wuhan University , Wuhan 430071, P. R. China
| | - Qigui Mo
- Institute of TCM and Natural Products, School of Pharmaceutical Sciences, Wuhan University , Wuhan 430071, P. R. China
| | - Gao Zhou
- Institute of TCM and Natural Products, School of Pharmaceutical Sciences, Wuhan University , Wuhan 430071, P. R. China
| | - Jingsheng He
- Institute of TCM and Natural Products, School of Pharmaceutical Sciences, Wuhan University , Wuhan 430071, P. R. China
| | - Youwei Wang
- Institute of TCM and Natural Products, School of Pharmaceutical Sciences, Wuhan University , Wuhan 430071, P. R. China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University , Wuhan 430072, P.R. China
| |
Collapse
|
43
|
Sandoval D, Sisley SR. Brain GLP-1 and insulin sensitivity. Mol Cell Endocrinol 2015; 418 Pt 1:27-32. [PMID: 25724479 PMCID: PMC4547906 DOI: 10.1016/j.mce.2015.02.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/02/2015] [Accepted: 02/17/2015] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes is often treated with a class of drugs referred to as glucagon-like peptide-1 (GLP-1) analogs. GLP-1 is a peptide secreted by the gut that acts through only one known receptor, the GLP-1 receptor. The primary function of GLP-1 is thought to be lowering of postprandial glucose levels. Indeed, medications utilizing this system, including the long-acting GLP-1 analogs liraglutide and exenatide, are beneficial in reducing both blood sugars and body weight. GLP-1 analogs were long presumed to affect glucose control through their ability to increase insulin levels through peripheral action on beta cells. However, multiple lines of data point to the ability of GLP-1 to act within the brain to alter glucose regulation. In this review we will discuss the evidence for a central GLP-1 system and the effects of GLP-1 in the brain on regulating multiple facets of glucose homeostasis including glucose tolerance, insulin production, insulin sensitivity, hepatic glucose production, muscle glucose uptake, and connections of the central GLP-1 system to the gut. Although the evidence indicates that GLP-1 receptors in the brain are not necessary for physiologic control of glucose regulation, we discuss the research showing a strong effect of acute manipulation of the central GLP-1 system on glucose control and how it is relevant to type 2 diabetic patients.
Collapse
Affiliation(s)
- Darleen Sandoval
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Stephanie R Sisley
- Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
44
|
Distribution and characterisation of Glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol Metab 2015; 4:718-31. [PMID: 26500843 PMCID: PMC4588458 DOI: 10.1016/j.molmet.2015.07.008] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 07/24/2015] [Accepted: 07/28/2015] [Indexed: 12/19/2022] Open
Abstract
Objective Although Glucagon-like peptide 1 is a key regulator of energy metabolism and food intake, the precise location of GLP-1 receptors and the physiological relevance of certain populations is debatable. This study investigated the novel GLP-1R-Cre mouse as a functional tool to address this question. Methods Mice expressing Cre-recombinase under the Glp1r promoter were crossed with either a ROSA26 eYFP or tdRFP reporter strain to identify GLP-1R expressing cells. Patch-clamp recordings were performed on tdRFP-positive neurons in acute coronal brain slices from adult mice and selective targeting of GLP-1R cells in vivo was achieved using viral gene delivery. Results Large numbers of eYFP or tdRFP immunoreactive cells were found in the circumventricular organs, amygdala, hypothalamic nuclei and the ventrolateral medulla. Smaller numbers were observed in the nucleus of the solitary tract and the thalamic paraventricular nucleus. However, tdRFP positive neurons were also found in areas without preproglucagon-neuronal projections like hippocampus and cortex. GLP-1R cells were not immunoreactive for GFAP or parvalbumin although some were catecholaminergic. GLP-1R expression was confirmed in whole-cell recordings from BNST, hippocampus and PVN, where 100 nM GLP-1 elicited a reversible inward current or depolarisation. Additionally, a unilateral stereotaxic injection of a cre-dependent AAV into the PVN demonstrated that tdRFP-positive cells express cre-recombinase facilitating virally-mediated eYFP expression. Conclusions This study is a comprehensive description and phenotypic analysis of GLP-1R expression in the mouse CNS. We demonstrate the power of combining the GLP-1R-CRE mouse with a virus to generate a selective molecular handle enabling future in vivo investigation as to their physiological importance. This transgenic mouse allows accurate evaluation of the distribution of GLP-1 receptor expressing cells. GLP-1 depolarises PVN, BNST and hippocampus neurons. GLP-1R expressing cells can be manipulated in vivo using this transgenic mouse.
Collapse
Key Words
- AP, area postrema
- BNST, bed nucleus stria terminalis
- Channelrhodopsin
- DMH, dorsomedial nucleus of the hypothalamus
- DMV, dorsal motor nucleus of the vagus
- Electrophysiology
- Ex-4, Exendin-4
- GFAP, glial fibrillary acidic protein
- GFP, green fluorescent protein
- GLP-1
- GLP-1, Glucagon-like peptide-1
- GLP-1R, Glucagon-like peptide-1 receptor
- Glucagon-like peptide-1 receptor
- NAc, nucleus accumbens
- NTS, nucleus of the solitary tract
- PARV, parvalbumin
- PPG
- PPG, preproglucagon
- PVN, paraventricular nucleus of the hypothalamus
- Preproglucagon
- TH, tyrosine hydroxylase
- VTA, ventral tegmental area
- YFP, yellow fluorescent protein
Collapse
|
45
|
Candeias EM, Sebastião IC, Cardoso SM, Correia SC, Carvalho CI, Plácido AI, Santos MS, Oliveira CR, Moreira PI, Duarte AI. Gut-brain connection: The neuroprotective effects of the anti-diabetic drug liraglutide. World J Diabetes 2015; 6:807-827. [PMID: 26131323 PMCID: PMC4478577 DOI: 10.4239/wjd.v6.i6.807] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/30/2015] [Accepted: 05/18/2015] [Indexed: 02/05/2023] Open
Abstract
Long-acting glucagon-like peptide-1 (GLP-1) analogues marketed for type 2 diabetes (T2D) treatment have been showing positive and protective effects in several different tissues, including pancreas, heart or even brain. This gut secreted hormone plays a potent insulinotropic activity and an important role in maintaining glucose homeostasis. Furthermore, growing evidences suggest the occurrence of several commonalities between T2D and neurodegenerative diseases, insulin resistance being pointed as a main cause for cognitive decline and increased risk to develop dementia. In this regard, it has also been suggested that stimulation of brain insulin signaling may have a protective role against cognitive deficits. As GLP-1 receptors (GLP-1R) are expressed throughout the central nervous system and GLP-1 may cross the blood-brain-barrier, an emerging hypothesis suggests that they may be promising therapeutic targets against brain dysfunctional insulin signaling-related pathologies. Importantly, GLP-1 actions depend not only on the direct effect mediated by its receptor activation, but also on the gut-brain axis involving an exchange of signals between both tissues via the vagal nerve, thereby regulating numerous physiological functions (e.g., energy homeostasis, glucose-dependent insulin secretion, as well as appetite and weight control). Amongst the incretin/GLP-1 mimetics class of anti-T2D drugs with an increasingly described neuroprotective potential, the already marketed liraglutide emerged as a GLP-1R agonist highly resistant to dipeptidyl peptidase-4 degradation (thereby having an increased half-life) and whose systemic GLP-1R activity is comparable to that of native GLP-1. Importantly, several preclinical studies showed anti-apoptotic, anti-inflammatory, anti-oxidant and neuroprotective effects of liraglutide against T2D, stroke and Alzheimer disease (AD), whereas several clinical trials, demonstrated some surprising benefits of liraglutide on weight loss, microglia inhibition, behavior and cognition, and in AD biomarkers. Herein, we discuss the GLP-1 action through the gut-brain axis, the hormone’s regulation of some autonomic functions and liraglutide’s neuroprotective potential.
Collapse
|
46
|
Iyer A, Brown L, Whitehead JP, Prins JB, Fairlie DP. Nutrient and immune sensing are obligate pathways in metabolism, immunity, and disease. FASEB J 2015; 29:3612-25. [PMID: 26065858 DOI: 10.1096/fj.15-271155] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 06/02/2015] [Indexed: 12/13/2022]
Abstract
The growth and survival of multicellular organisms depend upon their abilities to acquire and metabolize nutrients, efficiently store and harness energy, and sense and fight infection. Systems for sensing and using nutrients have consequently coevolved alongside systems for sensing and responding to danger signals, including pathogens, and share many of the same cell signaling proteins and networks. Diets rich in carbohydrates and fats can overload these systems, leading to obesity, metabolic dysfunction, impaired immunity, and cardiovascular disease. Excessive nutrient intake promotes adiposity, typically altering adipocyte function and immune cell distribution, both of which trigger metabolic dysfunction. Here, we discuss novel mechanistic links between metabolism and immunity that underlie metabolic dysfunction in obesity. We aim to stimulate debate about how the endocrine and immune systems are connected through autocrine, paracrine, and neuroendocrine signaling in sophisticated networks that are only now beginning to be resolved. Understanding the expression and action of signaling proteins, together with modulating their receptors or pattern recognition using agonists or antagonists, will enable rational intervention in immunometabolism that may lead to novel treatments for obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Abishek Iyer
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Lindsay Brown
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Jonathan P Whitehead
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - Johannes B Prins
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| | - David P Fairlie
- *Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; School of Health and Wellbeing, University of Southern Queensland, Toowoomba, Queensland, Australia; and Mater Research Institute-University of Queensland, Translational Research Institute, Queensland, Australia
| |
Collapse
|
47
|
Sandoval DA, D'Alessio DA. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease. Physiol Rev 2015; 95:513-48. [PMID: 25834231 DOI: 10.1152/physrev.00013.2014] [Citation(s) in RCA: 310] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preproglucagon gene (Gcg) is expressed by specific enteroendocrine cells (L-cells) of the intestinal mucosa, pancreatic islet α-cells, and a discrete set of neurons within the nucleus of the solitary tract. Gcg encodes multiple peptides including glucagon, glucagon-like peptide-1, glucagon-like peptide-2, oxyntomodulin, and glicentin. Of these, glucagon and GLP-1 have received the most attention because of important roles in glucose metabolism, involvement in diabetes and other disorders, and application to therapeutics. The generally accepted model is that GLP-1 improves glucose homeostasis indirectly via stimulation of nutrient-induced insulin release and by reducing glucagon secretion. Yet the body of literature surrounding GLP-1 physiology reveals an incompletely understood and complex system that includes peripheral and central GLP-1 actions to regulate energy and glucose homeostasis. On the other hand, glucagon is established principally as a counterregulatory hormone, increasing in response to physiological challenges that threaten adequate blood glucose levels and driving glucose production to restore euglycemia. However, there also exists a potential role for glucagon in regulating energy expenditure that has recently been suggested in pharmacological studies. It is also becoming apparent that there is cross-talk between the proglucagon derived-peptides, e.g., GLP-1 inhibits glucagon secretion, and some additive or synergistic pharmacological interaction between GLP-1 and glucagon, e.g., dual glucagon/GLP-1 agonists cause more weight loss than single agonists. In this review, we discuss the physiological functions of both glucagon and GLP-1 by comparing and contrasting how these peptides function, variably in concert and opposition, to regulate glucose and energy homeostasis.
Collapse
Affiliation(s)
- Darleen A Sandoval
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David A D'Alessio
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
48
|
Gagnon J, Baggio LL, Drucker DJ, Brubaker PL. Ghrelin Is a Novel Regulator of GLP-1 Secretion. Diabetes 2015; 64:1513-21. [PMID: 25412624 DOI: 10.2337/db14-1176] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/15/2014] [Indexed: 12/16/2022]
Abstract
GLP-1 is a gastrointestinal L-cell hormone that enhances glucose-stimulated insulin secretion. Hence, strategies that prevent GLP-1 degradation or activate the GLP-1 receptor are used to treat patients with type 2 diabetes. GLP-1 secretion occurs after a meal and is partly regulated by other circulating hormones. Ghrelin is a stomach-derived hormone that plays a key role in whole-body energy metabolism. Because ghrelin levels peak immediately before mealtimes, we hypothesized that ghrelin plays a role in priming the intestinal L-cell for nutrient-induced GLP-1 release. The intraperitoneal injection of ghrelin into mice 15 min before the administration of oral glucose enhanced glucose-stimulated GLP-1 release and improved glucose tolerance, whereas the ghrelin receptor antagonist D-Lys GHRP-6 reduced plasma levels of GLP-1 and insulin and diminished oral glucose tolerance. The ghrelin-mediated improvement in glucose tolerance was lost in mice coinjected with a GLP-1 receptor antagonist as well as in Glp1r(-/-) mice lacking the GLP-1 receptor. The impaired oral glucose tolerance in diet-induced obese mice was also improved by ghrelin preadministration. Importantly, ghrelin directly stimulated GLP-1 release from L-cell lines (murine GLUTag, human NCI-H716) through an extracellular signal-related kinase 1/2-dependent pathway. These studies demonstrate a novel role for ghrelin in enhancing the GLP-1 secretory response to ingested nutrients.
Collapse
Affiliation(s)
- Jeffrey Gagnon
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Laurie L Baggio
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada Lunenfeld-Tanenbaum Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Daniel J Drucker
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada Lunenfeld-Tanenbaum Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
49
|
Dietary sugars: their detection by the gut-brain axis and their peripheral and central effects in health and diseases. Eur J Nutr 2014; 54:1-24. [PMID: 25296886 PMCID: PMC4303703 DOI: 10.1007/s00394-014-0776-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 09/24/2014] [Indexed: 12/25/2022]
Abstract
Background Substantial increases in dietary sugar intake together with the increasing prevalence of obesity worldwide, as well as the parallels found between sugar overconsumption and drug abuse, have motivated research on the adverse effects of sugars on health and eating behaviour. Given that the gut–brain axis depends on multiple interactions between peripheral and central signals, and because these signals are interdependent, it is crucial to have a holistic view about dietary sugar effects on health. Methods Recent data on the effects of dietary sugars (i.e. sucrose, glucose, and fructose) at both peripheral and central levels and their interactions will be critically discussed in order to improve our understanding of the effects of sugars on health and diseases. This will contribute to the development of more efficient strategies for the prevention and treatment for obesity and associated co-morbidities. Results This review highlights opposing effects of glucose and fructose on metabolism and eating behaviour. Peripheral glucose and fructose sensing may influence eating behaviour by sweet-tasting mechanisms in the mouth and gut, and by glucose-sensing mechanisms in the gut. Glucose may impact brain reward regions and eating behaviour directly by crossing the blood–brain barrier, and indirectly by peripheral neural input and by oral and intestinal sweet taste/sugar-sensing mechanisms, whereas those promoted by fructose orally ingested seem to rely only on these indirect mechanisms. Conclusions Given the discrepancies between studies regarding the metabolic effects of sugars, more studies using physiological experimental conditions and in animal models closer to humans are needed. Additional studies directly comparing the effects of sucrose, glucose, and fructose should be performed to elucidate possible differences between these sugars on the reward circuitry.
Collapse
|
50
|
Secher A, Jelsing J, Baquero AF, Hecksher-Sørensen J, Cowley MA, Dalbøge LS, Hansen G, Grove KL, Pyke C, Raun K, Schäffer L, Tang-Christensen M, Verma S, Witgen BM, Vrang N, Bjerre Knudsen L. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest 2014; 124:4473-88. [PMID: 25202980 DOI: 10.1172/jci75276] [Citation(s) in RCA: 586] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Liraglutide is a glucagon-like peptide-1 (GLP-1) analog marketed for the treatment of type 2 diabetes. Besides lowering blood glucose, liraglutide also reduces body weight. It is not fully understood how liraglutide induces weight loss or to what degree liraglutide acts directly in the brain. Here, we determined that liraglutide does not activate GLP-1-producing neurons in the hindbrain, and liraglutide-dependent body weight reduction in rats was independent of GLP-1 receptors (GLP-1Rs) in the vagus nerve, area postrema, and paraventricular nucleus. Peripheral injection of fluorescently labeled liraglutide in mice revealed the presence of the drug in the circumventricular organs. Moreover, labeled liraglutide bound neurons within the arcuate nucleus (ARC) and other discrete sites in the hypothalamus. GLP-1R was necessary for liraglutide uptake in the brain, as liraglutide binding was not seen in Glp1r(-/-) mice. In the ARC, liraglutide was internalized in neurons expressing proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). Electrophysiological measurements of murine brain slices revealed that GLP-1 directly stimulates POMC/CART neurons and indirectly inhibits neurotransmission in neurons expressing neuropeptide Y (NPY) and agouti-related peptide (AgRP) via GABA-dependent signaling. Collectively, our findings indicate that the GLP-1R on POMC/CART-expressing ARC neurons likely mediates liraglutide-induced weight loss.
Collapse
|