1
|
Park JS, Kim KS, Choi HJ. Glucagon-Like Peptide-1 and Hypothalamic Regulation of Satiation: Cognitive and Neural Insights from Human and Animal Studies. Diabetes Metab J 2025; 49:333-347. [PMID: 40367985 PMCID: PMC12086555 DOI: 10.4093/dmj.2025.0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have emerged as blockbuster drugs for treating metabolic diseases. Glucagon-like peptide-1 (GLP-1) plays a pivotal role in glucose homeostasis by enhancing insulin secretion, suppressing glucagon release, delaying gastric emptying, and acting on the central nervous system to regulate satiation and satiety. This review summarizes the discovery of GLP-1 and the development of GLP-1RAs, with a particular focus on their central mechanisms of action. Human neuroimaging studies demonstrate that GLP-1RAs influence brain activity during food cognition, supporting a role in pre-ingestive satiation. Animal studies on hypothalamic feed-forward regulation of hunger suggest that cognitive hypothalamic mechanisms may also contribute to satiation control. We highlight the brain mechanisms of GLP-1RA-induced satiation and satiety, including cognitive impacts, with an emphasis on animal studies of hypothalamic glucagon-like peptide-1 receptor (GLP-1R) and GLP-1R-expressing neurons. Actions in non-hypothalamic regions are also discussed. Additionally, we review emerging combination drugs and oral GLP-1RA formulations aimed at improving efficacy and patient adherence. In conclusion, the dorsomedial hypothalamus (DMH)-a key GLP-1RA target-mediates pre-ingestive cognitive satiation, while other hypothalamic GLP-1R neurons regulate diverse aspects of feeding behavior, offering potential therapeutic targets for obesity treatment.
Collapse
Affiliation(s)
- Joon Seok Park
- Department of Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyu Sik Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Hyung Jin Choi
- Department of Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Korea
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
2
|
Lin Z, Xuan Y, Zhang Y, Zhou Q, Qiu W. Hypothalamus and brainstem circuits in the regulation of glucose homeostasis. Am J Physiol Endocrinol Metab 2025; 328:E588-E598. [PMID: 40047236 DOI: 10.1152/ajpendo.00474.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/03/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025]
Abstract
The central nervous system (CNS) senses and integrates blood glucose status, regulating its levels through communication with peripheral organs. Since traditional wisdom holds that the hypothalamus primarily controls glucose homeostasis, the brainstem, although less studied, has been emerging as a key player in blood glucose metabolism. Although the brainstem is reciprocally wired with the hypothalamus, their interactions are crucial for glucose control. Here, we focus on classic discoveries and recent advancements of hypothalamic and brainstem nodes that regulate glucose homeostasis. Based on the current progress and development for central regulation of blood sugar, we propose that the circuitry and cellular mechanisms for how hypothalamus and brainstem coordinate in blood sugar regulation are crucial; hence, a deeper understanding of both nuclei could shed light on a future cure for diabetes.
Collapse
Affiliation(s)
- Zitian Lin
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yunxin Xuan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yingshi Zhang
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Qirui Zhou
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Weiwei Qiu
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| |
Collapse
|
3
|
Zandvakili I, Perez-Tilve D. The unexpected role of GIP in transforming obesity treatment. Trends Endocrinol Metab 2025; 36:330-338. [PMID: 39198118 DOI: 10.1016/j.tem.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Despite sharing incretin activity with glucagon-like peptide 1 (GLP-1), the development of gastric inhibitory polypeptide (GIP)-based drugs has been hindered by the minor effects of native GIP on appetite and body weight and genetic studies associating loss-of-function with reduced obesity. Yet, pharmacologically optimized GIP-based molecules have demonstrated profound weight lowering benefits of GIPR agonism when combined with GLP-1-based therapies, which has re-energized deeper exploration of the molecular mechanisms and downstream signaling of GIPR. Interestingly, both GIPR agonism and antagonism offer metabolic benefits, leading to differing viewpoints on how to target GIPR therapeutically. Here we summarize the emerging evidence about the tissue-specific mechanisms that positions GIP-based therapies as important targets for the next generation of anti-obesity and metabolic therapies.
Collapse
Affiliation(s)
- Inuk Zandvakili
- Division of Digestive Diseases, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Diego Perez-Tilve
- Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
4
|
Xu Z, Wen S, Dong M, Zhou L. Targeting central pathway of Glucose-Dependent Insulinotropic Polypeptide, Glucagon and Glucagon-like Peptide-1 for metabolic regulation in obesity and type 2 diabetes. Diabetes Obes Metab 2025; 27:1660-1675. [PMID: 39723473 DOI: 10.1111/dom.16146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Obesity and type 2 diabetes are significant public health challenges that greatly impact global well-being. The development of effective therapeutic strategies has become more and more concentrated on the central nervous system and metabolic regulation. The primary pharmaceutical interventions for the treatment of obesity and uncontrolled hyperglycemia are now generally considered to be incretin-based anti-diabetic treatments, particularly glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide receptor agonists. This is a result of their substantial influence on the central nervous system and the consequent effects on energy balance and glucose regulation. It is increasingly crucial to understand the neural pathways of these pharmaceuticals. The purpose of this review is to compile and present the most recent central pathways regarding glucagon-like peptide-1, glucose-dependent insulinotropic polypeptide and glucagon receptors, with a particular emphasis on central metabolic regulation.
Collapse
Affiliation(s)
- Zhimin Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Fudan Zhangjiang Institute, Fudan University, Shanghai, China
| | - Meiyuan Dong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
5
|
Qiu T, Fu O. GPCRs in hypothalamic neurons and their roles in controlling food intake and metabolism. Front Mol Neurosci 2025; 18:1536577. [PMID: 39974186 PMCID: PMC11835808 DOI: 10.3389/fnmol.2025.1536577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
G-protein coupled receptor (GPCR) subtypes within the hypothalamus play a pivotal role in maintaining body homeostasis, particularly in the regulation of food intake and energy metabolism. This review provides an overview of classical loss and gain-of-function studies on GPCRs related to feeding and metabolism, with a focus on emerging cell-type-specific investigations. These studies reveal that diverse GPCR-expressing neuronal populations are intricately linked to feeding and energy balance. We also discuss recent findings that highlight the interaction of distinct peptide-GPCR systems in modulating complex feeding behaviors.
Collapse
Affiliation(s)
- Tian Qiu
- School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
- Laboratory of Food Perception Science, Science Center for Future Foods, Jiangnan University, Wuxi, Jiangsu, China
| | - Ou Fu
- School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
- Laboratory of Food Perception Science, Science Center for Future Foods, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
6
|
Yin K, Zhang T, Lu X, Shen Q, Gu K, Huang Y, Li C, Hou J, Li J, Zhang G. Tak1 licenses mitochondrial transfer from astrocytes to POMC neurons to maintain glucose and cholesterol homeostasis. Cell Rep 2024; 43:114983. [PMID: 39565693 DOI: 10.1016/j.celrep.2024.114983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/24/2024] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
It remains incompletely understood how the astrocytes in the mediobasal hypothalamus (MBH) regulate systemic glucose and cholesterol metabolism. Here, we show that MBH astrocytic Tak1 (transforming growth factor β [TGF-β]-activated kinase 1) controls the metabolism of glucose and cholesterol. Tak1 is expressed in MBH astrocytes and activated after a short-term nutritional excess. In chow-fed mice, astrocytic deletion of Tak1 across the brain or its suppression in the MBH impairs glucose tolerance, reduces insulin sensitivity, and results in hypercholesterolemia. Astrocytic Tak1 activation in the MBH alleviates these symptoms in mice fed a high-fat diet (HFD). We show that astrocytic Tak1 modulates the activity of proopiomelanocortin (POMC) neurons and enables the transport of mitochondria from astrocytes to POMC neurons. In astrocytic Tak1 knockout mice, supplementation of CD38, a molecule that is crucial in mitochondrial transfer, restores glucose and cholesterol homeostasis. Overall, these findings highlight an important role of MBH astrocytic Tak1 in glucose and cholesterol metabolism.
Collapse
Affiliation(s)
- Kaili Yin
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tingting Zhang
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiyuan Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qing Shen
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Kaiyue Gu
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Ying Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Chaonan Li
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Jingyi Hou
- Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Guo Zhang
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Institute of Metabolism and Health, Henan University, Kaifeng, Henan 475004, China; Zhongzhou Laboratory for Integrative Biology, Zhengzhou, Henan 450046, China.
| |
Collapse
|
7
|
Wei J, Wu H, Zheng Y, Wang N, Benedict C, Chen W, Tan X. Adequate sleep duration accentuates the effect of glucagon-like peptide-1 receptor variant on HbA1c: A gene-environment interaction study. Diabetes Res Clin Pract 2024; 218:111927. [PMID: 39536975 DOI: 10.1016/j.diabres.2024.111927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/24/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Both glucagon-like peptide-1 receptor (GLP1R) agonists and lifestyle modifications are widely adopted in managing glycemia. However, the joint effects of GLP1R agonists with lifestyle on glycemic traits have not been evaluated. METHODS This gene-environment study tested the interaction between GLP1R-rs10305492 variant, consistent with the effect of GLP1R agonist therapies, and four lifestyle factors (diet, physical activity, sleep duration, and chronotype) for glucose and glycated hemoglobin (HbA1c) levels among 263,846 UK Biobank participants. Linear regression models were conducted to evaluate the effects of the rs10305492 and lifestyle factors on glucose and HbA1c levels. RESULTS GLP1R-rs10305492-AA/AG genotype combined a healthy diet, regular physical activity, adequate sleep duration, or morning chronotype were associated with lower glucose and HbA1c levels (all P for trend < 0.001). A synergistic effect was found between rs10305492 and sleep duration on HbA1c, suggesting a recommended adequate sleep duration (7-8 h/day) may amplify the HbA1c lowering effect of GLP1R agonists. Joint effects of the rs10305492 and adequate sleep were associated with a 26 % reduced risk of hyperglycemia (>7.8 mmol/L) risk and a 22 % lower of high HbA1c (>39 mmol/mol or 5.7 %). CONCLUSIONS Combining GLP1R agonists with adequate sleep may provide additional benefits for glycemic control in clinical practice.
Collapse
Affiliation(s)
- Jiahe Wei
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Hanzhang Wu
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Ying Zheng
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Ningjian Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Christian Benedict
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, 75124, Uppsala, Sweden
| | - Wei Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiao Tan
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China; Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
8
|
Mirzadeh Z, Faber C. Brain Defense of Glycemia in Health and Diabetes. Diabetes 2024; 73:1952-1966. [PMID: 39401393 PMCID: PMC11579547 DOI: 10.2337/dbi24-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 11/22/2024]
Abstract
The brain coordinates the homeostatic defense of multiple metabolic variables, including blood glucose levels, in the context of ever-changing external and internal environments. The biologically defended level of glycemia (BDLG) is the net result of brain modulation of insulin-dependent mechanisms in cooperation with the islet, and insulin-independent mechanisms through direct innervation and neuroendocrine control of glucose effector tissues. In this article, we highlight evidence from animal and human studies to develop a framework for the brain's core homeostatic functions-sensory/afferent, integration/processing, and motor/efferent-that contribute to the normal BDLG in health and its elevation in diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Zaman Mirzadeh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | - Chelsea Faber
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| |
Collapse
|
9
|
Yang S, Tang Q, Zhang Y, Du Y, Zhao X, Mei F, Li Y. Neuronostatin regulates neuronal function and energetic metabolism in Alzheimer's disease in a GPR107-dependent manner. Neuropharmacology 2024; 258:110090. [PMID: 39048031 DOI: 10.1016/j.neuropharm.2024.110090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, which is characterized by the accumulation and aggregation of amyloid in brain. Neuronostatin (NST) is an endogenous peptide hormone that participates in many fundamental neuronal processes. However, the metabolism and function of NST in neurons of AD mice are not known. In this study, by combining the structural analyses, primary cultures, knockout cells, and various assessments, the behavior, histopathology, brain-wide expression and cellular signaling pathways in the APP/PS1 mice were investigated. It was found that NST directly bound to GPR107, which was primarily expressed in neurons. NST modulated the neuronal survivability and neurite outgrowth induced by Aβ via GPR107 in neurons. Intracerebroventricular (i.c.v.) administration of NST attenuated learning and memory abilities, reduced the synaptic protein levels of hippocampus, but improved amyloid plaques in the cortex and hippocampus of APP/PS1 mice. NST modulated glucose metabolism of hypothalamus-hippocampus-cortex axis in APP/PS1 mice and decreased ATP levels via the regulation of reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) in response to Aβ, suppressed energetic metabolism, and mitochondrial function in neurons via GPR107/protein kinase A (PKA) signaling pathway. In summary, our findings suggest that NST regulates neuronal function and brain energetic metabolism in AD mice via the GPR107/PKA signaling pathway, which can be a promising target for the treatment of AD.
Collapse
Affiliation(s)
- Shaobin Yang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China.
| | - Qi Tang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Yimeng Zhang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Yaqin Du
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Xiaoqian Zhao
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Fangting Mei
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Yanhong Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| |
Collapse
|
10
|
Krishnan G, Bagath M, Devaraj C, Soren NM. The signalling association of glucagon-like peptide-1 and its receptors in the gastrointestinal tract and GPR40 and insulin receptor in the pancreas of sheep. Gen Comp Endocrinol 2024; 358:114602. [PMID: 39226991 DOI: 10.1016/j.ygcen.2024.114602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/19/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024]
Abstract
The present study was aimed at gaining insight into the signalling relationship between glucagon-like peptide-1 (GLP-1) and its receptor (GLP-1R) in the regulation of glucose metabolism. Further, to assess the role of G-protein-coupled receptor 40 (GPR40) and insulin receptor (INSR) in the pancreas of sheep that were supplemented with calcium salts of long-chain fatty acids (CSFAs). An experiment was carried out over a period of 60 days with eighteen sheep, and they were fed with a standard basal diet. The sheep were divided into three groups: CSFA0 (without CSFAs), while CSFA3 and CSFA5 were supplemented with 3 % and 5 % of CSFAs, respectively. Plasma concentrations of GLP-1, insulin, glucagon, and glucose were assessed every two weeks. At the end of the experiment, sheep were slaughtered, and samples of gastrointestinal tract (GIT) epithelial tissues and pancreas were collected to assess the relative expression of mRNA of GPR40, GLP-1R, and INSR. Postprandial GLP-1 and insulin were increased by 3.7-4.1 and 1.45-1.5 times, respectively, in the CSFAs-supplemented groups compared to CSFA0. Post-feeding, glucagon and glucose levels decreased in CSFA3 and CSFA5 compared to CSFA0. The results indicated that the supplementation of LCFAs increased the expression of GLP-1R in the GIT and pancreas, as well as the mRNA of GPR40 and INSR in the pancreas. Chemosensing of LCFAs by GPR40 in the pancreas triggers signalling transduction, and enhanced GLP-1 and GLP-1R resulted in moderately increased insulin secretion and reduced glucagon levels. These combined effects, along with the glucose-lowering effect of GLP-1, effectively lowered glucose levels in normoglycemic sheep.
Collapse
Affiliation(s)
- G Krishnan
- Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India.
| | - M Bagath
- Animal Nutrition Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| | - C Devaraj
- Bioenergetics and Environmental Sciences Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| | - N M Soren
- Animal Nutrition Division, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore 560030, India
| |
Collapse
|
11
|
Guo X, Feng H, Cai L, Zheng J, Li Y. DPP-IV as a potential candidate in anti-obesity and obesity-related diseases treatment. Biomed Pharmacother 2024; 180:117464. [PMID: 39326107 DOI: 10.1016/j.biopha.2024.117464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Along with social development and lifestyle changes, the number of overweight and obese patients worldwide is rising annually. Obesity is a chronic metabolic disease with complex etiology. Dipeptidyl peptidase IV (DPP-IV) is a novel adipokine with significantly elevated expression in the visceral fat of obese patients. DPP-IV is a molecule that regulates metabolic homeostasis and inflammatory processes. Through its enzymatic activity, it plays a significant part in achieving hypoglycemic and weight loss effects through various pathways. DPP-IV and DPP-IV inhibitors also have pleiotropic effects in modulating obesity-related diseases by reducing obesity-related inflammation, ameliorating inflammatory bowel disease (IBD), improving hepatic steatosis and lowering cardiovascular risk, and even decreasing the risk of novel coronavirus disease-19 (COVID-19). This paper reviews the mechanisms of action based on DPP-IV targets in obesity and metabolic homeostasis, as well as their active role in the treatment of chronic diseases associated with obesity.
Collapse
Affiliation(s)
- Xin Guo
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| | - Huolun Feng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China.
| | - Liyang Cai
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| | - Jiabin Zheng
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| | - Yong Li
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
12
|
Liu L, Huang Z, Zhang J, Wang M, Yue T, Wang W, Wu Y, Zhang Z, Xiong W, Wang C, Wu F, Zhan C, Bi G, Liu J. Hypothalamus-sympathetic-liver axis mediates the early phase of stress-induced hyperglycemia in the male mice. Nat Commun 2024; 15:8632. [PMID: 39366937 PMCID: PMC11452707 DOI: 10.1038/s41467-024-52815-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024] Open
Abstract
Rapid glucose supply is crucial for animal survival during stress response. How the timescale of stress-induced glucose release precisely controlled by hypothalamic corticotropin-releasing hormone (CRH) neurons remains unclear. Here, we show that stress-induced hyperglycemia can be divided into at least two stages in male mice: the first fast stage is mediated by hypothalamus (paraventricular to ventromedial hypothalamus)-sympathetic (raphe pallidus nucleus to intermediolateral nucleus)-liver (HSL) axis activity; the second delayed stage is mediated by adrenal activity. Blocking the activity of HSL axis impairs predatory evoked flight responses, indicating that the HSL pathway activity is necessary for stress coping. We further reveal the intracellular signal cascade for CRH signal in the hypothalamus, which is mediated by GABAA receptor β3 subunit phosphorylation at S408/409, results in prevention of GABAA receptor membrane recruitment. Thus, we uncovered the precise timescale of glucose supply during stress which is mediated by adrenal independent HSL and adrenal dependent pathway respectively.
Collapse
Affiliation(s)
- Ling Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Zhaohuan Huang
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
| | - Jian Zhang
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Mengtian Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Ting Yue
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Wei Wang
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Yue Wu
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Zhi Zhang
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Wei Xiong
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Chao Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Feng Wu
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Cheng Zhan
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, LuJiang Road 17, Hefei, 230001, China
| | - Guoqiang Bi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China
| | - Ji Liu
- Department of Endocrinology, The First Affiliated Hospital of USTC, National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, West WangJiang Road 5089, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Diseases, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
- MoE Key Laboratory of Brain-inspired Intelligent Perception and Cognition, University of Science and Technology of China, Huangshan Road 443, Hefei, 230027, China.
| |
Collapse
|
13
|
Chen B, Yu X, Horvath-Diano C, Ortuño MJ, Tschöp MH, Jastreboff AM, Schneeberger M. GLP-1 programs the neurovascular landscape. Cell Metab 2024; 36:2173-2189. [PMID: 39357509 DOI: 10.1016/j.cmet.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Readily available nutrient-rich foods exploit our inherent drive to overconsume, creating an environment of overnutrition. This transformative setting has led to persistent health issues, such as obesity and metabolic syndrome. The development of glucagon-like peptide-1 receptor (GLP-1R) agonists reveals our ability to pharmacologically manage weight and address metabolic conditions. Obesity is directly linked to chronic low-grade inflammation, connecting our metabolic environment to neurodegenerative diseases. GLP-1R agonism in curbing obesity, achieved by impacting appetite and addressing associated metabolic defects, is revealing additional benefits extending beyond weight loss. Whether GLP-1R agonism directly impacts brain health or does so indirectly through improved metabolic health remains to be elucidated. In exploring the intricate connection between obesity and neurological conditions, recent literature suggests that GLP-1R agonism may have the capacity to shape the neurovascular landscape. Thus, GLP-1R agonism emerges as a promising strategy for addressing the complex interplay between metabolic health and cognitive well-being.
Collapse
Affiliation(s)
- Bandy Chen
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Xiaofei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Claudia Horvath-Diano
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - María José Ortuño
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | - Ania M Jastreboff
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA; Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
14
|
D'Ávila M, Hall S, Horvath TL. GLP-1, GIP, and Glucagon Agonists for Obesity Treatment: A Hunger Perspective. Endocrinology 2024; 165:bqae128. [PMID: 39301751 DOI: 10.1210/endocr/bqae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 09/22/2024]
Abstract
For centuries, increasingly sophisticated methods and approaches have been brought to bear to promote weight loss. Second only to the Holy Grail of research on aging, the idea of finding a single and simple way to lose weight has long preoccupied the minds of laymen and scientists alike. The effects of obesity are far-reaching and not to be minimized; the need for more effective treatments is obvious. Is there a single silver bullet that addresses this issue without effort on the part of the individual? The answer to this question has been one of the most elusive and sought-after in modern history. Now and then, a miraculous discovery propagates the illusion that a simple solution is possible. Now there are designer drugs that seem to accomplish the task: we can lose weight without effort using mono, dual, and triple agonists of receptors for glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and glucagon. There are, however, fundamental biological principles that raise intriguing questions about these therapies beyond the currently reported side-effects. This perspective reflects upon these issues from the angle of complex goal-oriented behaviors, and systemic and cellular metabolism associated with satiety and hunger.
Collapse
Affiliation(s)
- Mateus D'Ávila
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06520, USA
| | - Samantha Hall
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
15
|
Chen Z, Deng X, Shi C, Jing H, Tian Y, Zhong J, Chen G, Xu Y, Luo Y, Zhu Y. GLP-1R-positive neurons in the lateral septum mediate the anorectic and weight-lowering effects of liraglutide in mice. J Clin Invest 2024; 134:e178239. [PMID: 39225090 PMCID: PMC11364389 DOI: 10.1172/jci178239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Liraglutide, a glucagon-like peptide-1 (GLP-1) analog, is approved for obesity treatment, but the specific neuronal sites that contribute to its therapeutic effects remain elusive. Here, we show that GLP-1 receptor-positive (GLP-1R-positive) neurons in the lateral septum (LSGLP-1R) play a critical role in mediating the anorectic and weight-loss effects of liraglutide. LSGLP-1R neurons were robustly activated by liraglutide, and chemogenetic activation of these neurons dramatically suppressed feeding. Targeted knockdown of GLP-1 receptors within the LS, but not in the hypothalamus, substantially attenuated liraglutide's ability to inhibit feeding and lower body weight. The activity of LSGLP-1R neurons rapidly decreased during naturalistic feeding episodes, while synaptic inactivation of LSGLP-1R neurons diminished the anorexic effects triggered by liraglutide. Together, these findings offer critical insights into the functional role of LSGLP-1R neurons in the physiological regulation of energy homeostasis and delineate their instrumental role in mediating the pharmacological efficacy of liraglutide.
Collapse
Affiliation(s)
- Zijun Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaofei Deng
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cuijie Shi
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Haiyang Jing
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yu Tian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Jiafeng Zhong
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Gaowei Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yunlong Xu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Yixiao Luo
- Hunan Province People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
16
|
Kim KS, Park JS, Hwang E, Park MJ, Shin HY, Lee YH, Kim KM, Gautron L, Godschall E, Portillo B, Grose K, Jung SH, Baek SL, Yun YH, Lee D, Kim E, Ajwani J, Yoo SH, Güler AD, Williams KW, Choi HJ. GLP-1 increases preingestive satiation via hypothalamic circuits in mice and humans. Science 2024; 385:438-446. [PMID: 38935778 PMCID: PMC11961025 DOI: 10.1126/science.adj2537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/31/2024] [Indexed: 06/29/2024]
Abstract
Glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RAs) are effective antiobesity drugs. However, the precise central mechanisms of GLP-1RAs remain elusive. We administered GLP-1RAs to patients with obesity and observed a heightened sense of preingestive satiation. Analysis of human and mouse brain samples pinpointed GLP-1 receptor (GLP-1R) neurons in the dorsomedial hypothalamus (DMH) as candidates for encoding preingestive satiation. Optogenetic manipulation of DMHGLP-1R neurons caused satiation. Calcium imaging demonstrated that these neurons are actively involved in encoding preingestive satiation. GLP-1RA administration increased the activity of DMHGLP-1R neurons selectively during eating behavior. We further identified that an intricate interplay between DMHGLP-1R neurons and neuropeptide Y/agouti-related peptide neurons of the arcuate nucleus (ARCNPY/AgRP neurons) occurs to regulate food intake. Our findings reveal a hypothalamic mechanism through which GLP-1RAs control preingestive satiation, offering previously unexplored neural targets for obesity and metabolic diseases.
Collapse
Affiliation(s)
- Kyu Sik Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Joon Seok Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Eunsang Hwang
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Min Jung Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hwa Yun Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Young Hee Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Kyung Min Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Laurent Gautron
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Bryan Portillo
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kyle Grose
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sang-Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - So Lin Baek
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Young Hyun Yun
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Doyeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Eunseong Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Jason Ajwani
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Seong Ho Yoo
- Department of Forensic Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Seoul, Republic of Korea
| | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Kevin W. Williams
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Hyung Jin Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Wide River Institute of Immunology, Seoul National University, 101 Dabyeonbat-gil, Hwachon-myeon, Gangwon-do 25159, Republic of Korea
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
17
|
Spezani R, Marinho TS, Reis TS, Aguila MB, Mandarim-de-Lacerda CA. Cotadutide (GLP-1/Glucagon dual receptor agonist) modulates hypothalamic orexigenic and anorexigenic neuropeptides in obese mice. Peptides 2024; 173:171138. [PMID: 38147963 DOI: 10.1016/j.peptides.2023.171138] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/15/2023] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
The hypothalamic neuropeptides linked to appetite and satiety were investigated in obese mice treated with cotadutide (a dual receptor agonist of glucagon-like peptide 1 (GLP-1R)/Glucagon (GCGR)). Twelve-week-old male C57BL/6 mice were fed a control diet (C group, n = 20) or a high-fat diet (HF group, n = 20) for ten weeks. Each group was further divided, adding cotadutide treatment and forming groups C, CC, HF, and HFC for four additional weeks. The hypothalamic arcuate neurons were labeled by immunofluorescence, and protein expressions (Western blotting) for neuropeptide Y (NPY), proopiomelanocortin (POMC), agouti-related protein (AgRP), and cocaine- and amphetamine-regulated transcript (CART). Cotadutide enhanced POMC and CART neuropeptides and depressed NPY and AGRP neuropeptides. In addition, gene expressions (RT-qPCR) determined that Lepr (leptin receptor) and Calcr (calcitonin receptor) were diminished in HF compared to C but enhanced in CC compared to C and HFC compared to HF. Besides, Socs3 (suppressor of cytokine signaling 3) was decreased in HFC compared to HF, while Sst (somatostatin) was higher in HFC compared to HF; Tac1 (tachykinin 1) and Mc4r (melanocortin-4-receptor) were lower in HF compared to C but increased in HFC compared to HF. Also, Glp1r and Gcgr were higher in HFC compared to HF. In conclusion, the findings are compelling, demonstrating the effects of cotadutide on hypothalamic neuropeptides and hormone receptors of obese mice. Cotadutide modulates energy balance through the gut-brain axis and its associated signaling pathways. The study provides insights into the mechanisms underlying cotadutide's anti-obesity effects and its possible implications for obesity treatment.
Collapse
Affiliation(s)
- Renata Spezani
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Thiago Santos Reis
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Popova D, Sun J, Chow HM, Hart RP. A critical review of ethanol effects on neuronal firing: A metabolic perspective. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:450-458. [PMID: 38217065 PMCID: PMC10966925 DOI: 10.1111/acer.15266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
Ethanol metabolism is relatively understudied in neurons, even though changes in neuronal metabolism are known to affect their activity. Recent work demonstrates that ethanol is preferentially metabolized over glucose as a source of carbon and energy, and it reprograms neurons to a state of reduced energy potential and diminished capacity to utilize glucose once ethanol is exhausted. Ethanol intake has been associated with changes in neuronal firing and specific brain activity (EEG) patterns have been linked with risk for alcohol use disorder (AUD). Furthermore, a haplotype of the inwardly rectifying potassium channel subunit, GIRK2, which plays a critical role in regulating excitability of neurons, has been linked with AUD and shown to be directly regulated by ethanol. At the same time, overexpression of GIRK2 prevents ethanol-induced metabolic changes. Based on the available evidence, we conclude that the mechanisms underlying the effects of ethanol on neuronal metabolism are a novel target for developing therapies for AUD.
Collapse
Affiliation(s)
- Dina Popova
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway NJ USA
- Present address: Neuroscience Institute, NYU Langone Grossman School of Medicine, New York, NY USA
| | - Jacquelyne Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ronald P. Hart
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway NJ USA
| |
Collapse
|
19
|
Manuel J, Halbe E, Ewald AC, Hoff A, Jordan J, Tank J, Heusser K, Gerlach DA. Glucose-sensitive hypothalamic nuclei traced through functional magnetic resonance imaging. Front Neurosci 2023; 17:1297197. [PMID: 38146542 PMCID: PMC10749345 DOI: 10.3389/fnins.2023.1297197] [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: 09/19/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Introduction Hypothalamic glucose-sensitive neural circuits, which regulate energy metabolism and can contribute to diseases such as obesity and type 2 diabetes, have been difficult to study in humans. We developed an approach to assess hypothalamic functional connectivity changes during glucose loading using functional magnetic resonance imaging (fMRI). Methods To do so, we conducted oral glucose tolerance tests while acquiring functional images before, and 10 and 45 min after glucose ingestion in a healthy male and cross-sectionally in 20 healthy participants on two different diets. Results At group level, 39 fMRI sessions were not sufficient to detect glucose-mediated connectivity changes. However, 10 repeated sessions in a single subject revealed significant intrinsic functional connectivity increases 45 min after glucose intake in the arcuate, paraventricular, and dorsomedial nuclei, as well as in the posterior hypothalamic area, median eminence, and mammillary bodies. Discussion Our methodology allowed to outline glucose-sensitive hypothalamic pathways in a single human being and holds promise in delineating individual pathophysiology mechanisms in patients with dysglycemia.
Collapse
Affiliation(s)
- Jorge Manuel
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Institute for Neuroradiology, Hannover Medical School, Hanover, Germany
| | - Eva Halbe
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Ann Charlotte Ewald
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Alex Hoff
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
| | - Jens Tank
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Karsten Heusser
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Darius A. Gerlach
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| |
Collapse
|
20
|
Wu W, Zheng J, Wang R, Wang Y. Ion channels regulate energy homeostasis and the progression of metabolic disorders: Novel mechanisms and pharmacology of their modulators. Biochem Pharmacol 2023; 218:115863. [PMID: 37863328 DOI: 10.1016/j.bcp.2023.115863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
The progression of metabolic diseases, featured by dysregulated metabolic signaling pathways, is orchestrated by numerous signaling networks. Among the regulators, ion channels transport ions across the membranes and trigger downstream signaling transduction. They critically regulate energy homeostasis and pathogenesis of metabolic diseases and are potential therapeutic targets for treating metabolic disorders. Ion channel blockers have been used to treat diabetes for decades by stimulating insulin secretion, yet with hypoglycemia and other adverse effects. It calls for deeper understanding of the largely elusive regulatory mechanisms, which facilitates the identification of new therapeutic targets and safe drugs against ion channels. In the article, we critically assess the two principal regulatory mechanisms, protein-channel interaction and post-translational modification on the activities of ion channels to modulate energy homeostasis and metabolic disorders through multiple novel mechanisms. Moreover, we discuss the multidisciplinary methods that provide the tools for elucidation of the regulatory mechanisms mediating metabolic disorders by ion channels. In terms of translational perspective, the mechanistic analysis of recently validated ion channels that regulate insulin resistance, body weight control, and adverse effects of current ion channel antagonists are discussed in details. Their small molecule modulators serve as promising new drug candidates to combat metabolic disorders.
Collapse
Affiliation(s)
- Wenyi Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Jianan Zheng
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, China
| | - Yibing Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, China.
| |
Collapse
|
21
|
Lai TT, Liou CW, Tsai YH, Lin YY, Wu WL. Butterflies in the gut: the interplay between intestinal microbiota and stress. J Biomed Sci 2023; 30:92. [PMID: 38012609 PMCID: PMC10683179 DOI: 10.1186/s12929-023-00984-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
Psychological stress is a global issue that affects at least one-third of the population worldwide and increases the risk of numerous psychiatric disorders. Accumulating evidence suggests that the gut and its inhabiting microbes may regulate stress and stress-associated behavioral abnormalities. Hence, the objective of this review is to explore the causal relationships between the gut microbiota, stress, and behavior. Dysbiosis of the microbiome after stress exposure indicated microbial adaption to stressors. Strikingly, the hyperactivated stress signaling found in microbiota-deficient rodents can be normalized by microbiota-based treatments, suggesting that gut microbiota can actively modify the stress response. Microbiota can regulate stress response via intestinal glucocorticoids or autonomic nervous system. Several studies suggest that gut bacteria are involved in the direct modulation of steroid synthesis and metabolism. This review provides recent discoveries on the pathways by which gut microbes affect stress signaling and brain circuits and ultimately impact the host's complex behavior.
Collapse
Affiliation(s)
- Tzu-Ting Lai
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Chia-Wei Liou
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yu-Hsuan Tsai
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Yuan-Yuan Lin
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan
| | - Wei-Li Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|
22
|
Masliukov PM. Changes of Signaling Pathways in Hypothalamic Neurons with Aging. Curr Issues Mol Biol 2023; 45:8289-8308. [PMID: 37886966 PMCID: PMC10605528 DOI: 10.3390/cimb45100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
The hypothalamus is an important regulator of autonomic and endocrine functions also involved in aging regulation. The aging process in the hypothalamus is accompanied by disturbed intracellular signaling including insulin/insulin-like growth factor-1 (IGF-1)/growth hormone (GH), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/the mammalian target of rapamycin (mTOR), mitogen activated protein kinase (MAPK), janus kinase (JAK)/signal transducer and activator of transcription (STAT), AMP-activated protein kinase (AMPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB), and nitric oxide (NO). In the current review, I have summarized the current understanding of the changes in the above-mentioned pathways in aging with a focus on hypothalamic alterations.
Collapse
Affiliation(s)
- Petr M Masliukov
- Department Normal Physiology, Yaroslavl State Medical University, ul. Revoliucionnaya 5, 150000 Yaroslavl, Russia
| |
Collapse
|
23
|
Sanetra AM, Palus-Chramiec K, Chrobok L, Jeczmien-Lazur JS, Klich JD, Lewandowski MH. Proglucagon signalling in the rat Dorsomedial Hypothalamus - Physiology and high-fat diet-mediated alterations. Mol Cell Neurosci 2023; 126:103873. [PMID: 37295578 DOI: 10.1016/j.mcn.2023.103873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023] Open
Abstract
A relatively new pharmacological target in obesity treatment has been the preproglucagon (PPG) signalling, predominantly with glucagon-like peptide (GLP) 1 receptor agonists. As far as the PPG role within the digestive system is well recognised, its actions in the brain remain understudied. Here, we investigated PPG signalling in the Dorsomedial Hypothalamus (DMH), a structure involved in feeding regulation and metabolism, using in situ hybridisation, electrophysiology, and immunohistochemistry. Our experiments were performed on animals fed both control, and high-fat diet (HFD), uncovering HFD-mediated alterations. First, sensitivity to exendin-4 (Exn4, a GLP1R agonist) was shown to increase under HFD, with a higher number of responsive neurons. The amplitude of the response to both Exn4 and oxyntomodulin (Oxm) was also altered, diminishing its relationship with the cells' spontaneous firing rate. Not only neuronal sensitivity, but also GLP1 presence, and therefore possibly release, was influenced by HFD. Immunofluorescent labelling of the GLP1 showed changes in its density depending on the metabolic state (fasted/fed), but this effect was eliminated by HFD feeding. Interestingly, these dietary differences were absent after a period of restricted feeding, allowing for an anticipation of the alternating metabolic states, which suggests possible prevention of such outcome.
Collapse
Affiliation(s)
- A M Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland.
| | - K Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
| | - L Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland; School of Physiology, Pharmacology, and Neuroscience, University of Bristol, University Walk, Biomedical Sciences Building, Bristol BS8 1TD, UK
| | - J S Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
| | - J D Klich
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle Street 10, 13125 Berlin, Germany
| | - M H Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland.
| |
Collapse
|
24
|
Chen Z, Liu XA, Kenny PJ. Central and peripheral actions of nicotine that influence blood glucose homeostasis and the development of diabetes. Pharmacol Res 2023; 194:106860. [PMID: 37482325 DOI: 10.1016/j.phrs.2023.106860] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Cigarette smoking has long been recognized as a risk factor for type 2 diabetes (T2D), although the precise causal mechanisms underlying this relationship remain poorly understood. Recent evidence suggests that nicotine, the primary reinforcing component in tobacco, may play a pivotal role in connecting cigarette smoking and T2D. Extensive research conducted in both humans and animals has demonstrated that nicotine can elevate blood glucose levels, disrupt glucose homeostasis, and induce insulin resistance. The review aims to elucidate the genetic variants of nicotinic acetylcholine receptors associated with diabetes risk and provide a comprehensive overview of the available data on the mechanisms through which nicotine influences blood glucose homeostasis and the development of diabetes. Here we emphasize the central and peripheral actions of nicotine on the release of glucoregulatory hormones, as well as its effects on glucose tolerance and insulin sensitivity. Notably, the central actions of nicotine within the brain, which encompass both insulin-dependent and independent mechanisms, are highlighted as potential targets for intervention strategies in diabetes management.
Collapse
Affiliation(s)
- Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
25
|
Haspula D, Cui Z. Neurochemical Basis of Inter-Organ Crosstalk in Health and Obesity: Focus on the Hypothalamus and the Brainstem. Cells 2023; 12:1801. [PMID: 37443835 PMCID: PMC10341274 DOI: 10.3390/cells12131801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Precise neural regulation is required for maintenance of energy homeostasis. Essential to this are the hypothalamic and brainstem nuclei which are located adjacent and supra-adjacent to the circumventricular organs. They comprise multiple distinct neuronal populations which receive inputs not only from other brain regions, but also from circulating signals such as hormones, nutrients, metabolites and postprandial signals. Hence, they are ideally placed to exert a multi-tier control over metabolism. The neuronal sub-populations present in these key metabolically relevant nuclei regulate various facets of energy balance which includes appetite/satiety control, substrate utilization by peripheral organs and glucose homeostasis. In situations of heightened energy demand or excess, they maintain energy homeostasis by restoring the balance between energy intake and expenditure. While research on the metabolic role of the central nervous system has progressed rapidly, the neural circuitry and molecular mechanisms involved in regulating distinct metabolic functions have only gained traction in the last few decades. The focus of this review is to provide an updated summary of the mechanisms by which the various neuronal subpopulations, mainly located in the hypothalamus and the brainstem, regulate key metabolic functions.
Collapse
Affiliation(s)
- Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Zhenzhong Cui
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
| |
Collapse
|
26
|
Zhang ZY, Yan Q, Wu WH, Zhao Y, Zhang H, Li J. PPAR-alpha/gamma agonists, glucagon-like peptide-1 receptor agonists and metformin for non-alcoholic fatty liver disease: A network meta-analysis. J Int Med Res 2023; 51:3000605231177191. [PMID: 37317834 PMCID: PMC10278420 DOI: 10.1177/03000605231177191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/04/2023] [Indexed: 06/16/2023] Open
Abstract
OBJECTIVE To undertake a network meta-analysis to compare the relative efficacy of a dual peroxisome proliferator-activated receptor (PPAR)α and PPARγ agonist, glucagon-like peptide-1 receptor agonists (GLP-1RAs) and metformin in patients with non-alcoholic fatty liver disease (NAFLD). METHODS Electronic databases, including Embase®, PubMed® and The Cochrane Library, were searched systematically for eligible studies from inception to 20 July 2022. Randomized controlled trials (RCTs) that investigated aspartate aminotransferase, alanine aminotransferase (ALT) and triglyceride levels were considered for inclusion. Data were extracted using a standardized data collection table. A network meta-analysis was performed. Relative risk and 95% confidence interval were calculated for continuous data and I2 was used to assess the heterogeneity of studies. RESULTS A total of 22 RCTs involving 1698 patients were eligible for inclusion in the analysis. Both direct analysis and indirect analysis showed that saroglitazar was significantly superior to GLP-1RAs in improving ALT levels. Metformin improved ALT levels, but the effect was not as good as saroglitazar. CONCLUSION Saroglizatar was the most effective drug for improving NAFLD.INPLASY registration number: INPLASY202340066.
Collapse
Affiliation(s)
- Zhuo-Ya Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qi Yan
- Department of Endocrinology and Metabolism, Shanxi Medical University Affiliated Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Wen-Hao Wu
- Department of Endocrinology and Metabolism, Shanxi Medical University Affiliated Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yuan Zhao
- Department of Endocrinology and Metabolism, Shanxi Medical University Affiliated Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hua Zhang
- Department of Endocrinology and Metabolism, Shanxi Medical University Affiliated Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jin Li
- Department of Endocrinology and Metabolism, Shanxi Medical University Affiliated Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
27
|
Maity D, Guha Ray P, Buchmann P, Mansouri M, Fussenegger M. Blood-Glucose-Powered Metabolic Fuel Cell for Self-Sufficient Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300890. [PMID: 36893359 DOI: 10.1002/adma.202300890] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Indexed: 05/26/2023]
Abstract
Currently available bioelectronic devices consume too much power to be continuously operated on rechargeable batteries, and are often powered wirelessly, with attendant issues regarding reliability, convenience, and mobility. Thus, the availability of a robust, self-sufficient, implantable electrical power generator that works under physiological conditions would be transformative for many applications, from driving bioelectronic implants and prostheses to programing cellular behavior and patients' metabolism. Here, capitalizing on a new copper-containing, conductively tuned 3D carbon nanotube composite, an implantable blood-glucose-powered metabolic fuel cell is designed that continuously monitors blood-glucose levels, converts excess glucose into electrical power during hyperglycemia, and produces sufficient energy (0.7 mW cm-2 , 0.9 V, 50 mm glucose) to drive opto- and electro-genetic regulation of vesicular insulin release from engineered beta cells. It is shown that this integration of blood-glucose monitoring with elimination of excessive blood glucose by combined electro-metabolic conversion and insulin-release-mediated cellular consumption enables the metabolic fuel cell to restore blood-glucose homeostasis in an automatic, self-sufficient, and closed-loop manner in an experimental model of type-1 diabetes.
Collapse
Affiliation(s)
- Debasis Maity
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Preetam Guha Ray
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Peter Buchmann
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, Basel, CH-4058, Switzerland
| |
Collapse
|
28
|
Alonge KM, Porte D, Schwartz MW. Distinct Roles for Brain and Pancreas in Basal and Postprandial Glucose Homeostasis. Diabetes 2023; 72:547-556. [PMID: 37146276 PMCID: PMC10130484 DOI: 10.2337/db22-0969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/04/2023] [Indexed: 05/07/2023]
Abstract
The glucose homeostasis system ensures that the circulating glucose level is maintained within narrow physiological limits both in the fasting (or basal) state and following a nutrient challenge. Although glucose homeostasis is traditionally conceptualized as a single overarching system, evidence reviewed here suggests that basal glycemia and glucose tolerance are governed by distinct control systems. Specifically, whereas glucose tolerance appears to be determined largely by interactions between insulin secretion and insulin sensitivity, basal-state glucose homeostasis is predominated by insulin-independent mechanisms governed largely by the brain. In addition to a new perspective on how glucose homeostasis is achieved, this "dual control system" hypothesis offers a feasible and testable explanation for observations that are otherwise difficult to reconcile and sheds new light on the integration of central and peripheral metabolic control mechanisms. The implications of this model for the pathogenesis and treatment of impaired fasting glucose, impaired glucose tolerance, and type 2 diabetes are also discussed.
Collapse
Affiliation(s)
- Kimberly M. Alonge
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA
| | - Daniel Porte
- Division of Endocrinology, School of Medicine, University of California San Diego, San Diego, CA
| | - Michael W. Schwartz
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA
| |
Collapse
|
29
|
Han Y, He Y, Harris L, Xu Y, Wu Q. Identification of a GABAergic neural circuit governing leptin signaling deficiency-induced obesity. eLife 2023; 12:e82649. [PMID: 37043384 PMCID: PMC10097419 DOI: 10.7554/elife.82649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
The hormone leptin is known to robustly suppress food intake by acting upon the leptin receptor (LepR) signaling system residing within the agouti-related protein (AgRP) neurons of the hypothalamus. However, clinical studies indicate that leptin is undesirable as a therapeutic regiment for obesity, which is at least partly attributed to the poorly understood complex secondary structure and key signaling mechanism of the leptin-responsive neural circuit. Here, we show that the LepR-expressing portal neurons send GABAergic projections to a cohort of α3-GABAA receptor expressing neurons within the dorsomedial hypothalamic nucleus (DMH) for the control of leptin-mediated obesity phenotype. We identified the DMH as a key brain region that contributes to the regulation of leptin-mediated feeding. Acute activation of the GABAergic AgRP-DMH circuit promoted food intake and glucose intolerance, while activation of post-synaptic MC4R neurons in the DMH elicited exactly opposite phenotypes. Rapid deletion of LepR from AgRP neurons caused an obesity phenotype which can be rescued by blockage of GABAA receptor in the DMH. Consistent with behavioral results, these DMH neurons displayed suppressed neural activities in response to hunger or hyperglycemia. Furthermore, we identified that α3-GABAA receptor signaling within the DMH exerts potent bi-directional regulation of the central effects of leptin on feeding and body weight. Together, our results demonstrate a novel GABAergic neural circuit governing leptin-mediated feeding and energy balance via a unique α3-GABAA signaling within the secondary leptin-responsive neural circuit, constituting a new avenue for therapeutic interventions in the treatment of obesity and associated comorbidities.
Collapse
Affiliation(s)
- Yong Han
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Yang He
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Lauren Harris
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Yong Xu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| | - Qi Wu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
30
|
Zhang L, Jin M, Pan Y, Yang F, Wu Y, Gao J, Chen T, Tan S, Yang T, Chen Y, Huang J. Sustained release of GLP-1 analog from γ-PGA-PAE copolymers for management of type 2 diabetes. BIOMATERIALS ADVANCES 2023; 148:213352. [PMID: 36867980 DOI: 10.1016/j.bioadv.2023.213352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Abstract
GLP-1 has been clinically exploited for treating type 2 diabetes, while its short circulation half-life requires multiple daily injections to maintain effective glycemic control, thus limiting its widespread application. Here we developed a drug delivery system based on self-assembling polymer-amino acid conjugates (γ-PGA-PAE) to provide sustained release of GLP-1 analog (DLG3312). The DLG3312 loaded γ-PGA based nanoparticles (DLG3312@NPs) exhibited a spherical shape with a good monodispersity under transmission electron microscope (TEM) observation. The DLG3312 encapsulation was optimized, and the loading efficiency was as high as 78.4 ± 2.2 %. The transformation of DLG3312@NPs to network structures was observed upon treatment with the fresh serum, resulting in a sustained drug release. The in vivo long-term hypoglycemic assays indicated that DLG3312@NPs significantly reduced blood glucose and glycosylated hemoglobin level. Furthermore, DLG3312@NPs extended the efficacy of DLG3312, leading to a decrease in the dosing schedule that from once a day to once every other day. This approach combined the molecular and materials engineering strategies that offered a unique solution to maximize the availability of anti-diabetic drug and minimize its burdens to type 2 diabetic patients.
Collapse
Affiliation(s)
- Li Zhang
- School of Life Science, East China Normal University, Shanghai 200241, PR China
| | - Mingfei Jin
- School of Life Science, East China Normal University, Shanghai 200241, PR China
| | - Yingying Pan
- School of Life Science, East China Normal University, Shanghai 200241, PR China
| | - Fang Yang
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan Wu
- Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jianbo Gao
- Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Tao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shiming Tan
- School of Life Science, East China Normal University, Shanghai 200241, PR China
| | - Ting Yang
- School of Life Science, East China Normal University, Shanghai 200241, PR China
| | - Yazhou Chen
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, PR China; Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Jing Huang
- School of Life Science, East China Normal University, Shanghai 200241, PR China.
| |
Collapse
|
31
|
Sanetra AM, Palus-Chramiec K, Chrobok L, Jeczmien-Lazur JS, Gawron E, Klich JD, Pradel K, Lewandowski MH. High-Fat-Diet-Evoked Disruption of the Rat Dorsomedial Hypothalamic Clock Can Be Prevented by Restricted Nighttime Feeding. Nutrients 2022; 14:nu14235034. [PMID: 36501063 PMCID: PMC9735604 DOI: 10.3390/nu14235034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Obesity is a growing health problem for modern society; therefore, it has become extremely important to study not only its negative implications but also its developmental mechanism. Its links to disrupted circadian rhythmicity are indisputable but are still not well studied on the cellular level. Circadian food intake and metabolism are controlled by a set of brain structures referred to as the food-entrainable oscillator, among which the dorsomedial hypothalamus (DMH) seems to be especially heavily affected by diet-induced obesity. In this study, we evaluated the effects of a short-term high-fat diet (HFD) on the physiology of the male rat DMH, with special attention to its day/night changes. Using immunofluorescence and electrophysiology we found that both cFos immunoreactivity and electrical activity rhythms become disrupted after as few as 4 weeks of HFD consumption, so before the onset of excessive weight gain. This indicates that the DMH impairment is a possible factor in obesity development. The DMH cellular activity under an HFD became increased during the non-active daytime, which coincides with a disrupted rhythm in food intake. In order to explore the relationship between them, a separate group of rats underwent time-restricted feeding with access to food only during the nighttime. Such an approach completely abolished the disruptive effects of the HFD on the DMH clock, confirming its dependence on the feeding schedule of the animal. The presented data highlight the importance of a temporally regulated feeding pattern on the physiology of the hypothalamic center for food intake and metabolism regulation, and propose time-restricted feeding as a possible prevention of the circadian dysregulation observed under an HFD.
Collapse
Affiliation(s)
- Anna Magdalena Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
- Correspondence: (A.M.S.); (M.H.L.); Tel.: +48-12-664-53-56 (A.M.S.); +48-12-664-53-73 (M.H.L.)
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
| | - Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, University Walk, Biomedical Sciences Building, Bristol BS8 1TD, UK
| | - Jagoda Stanislawa Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
| | - Emilia Gawron
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
| | - Jasmin Daniela Klich
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa Street 9, 30-387 Krakow, Poland
- Correspondence: (A.M.S.); (M.H.L.); Tel.: +48-12-664-53-56 (A.M.S.); +48-12-664-53-73 (M.H.L.)
| |
Collapse
|
32
|
Singh I, Wang L, Xia B, Liu J, Tahiri A, El Ouaamari A, Wheeler MB, Pang ZP. Activation of arcuate nucleus glucagon-like peptide-1 receptor-expressing neurons suppresses food intake. Cell Biosci 2022; 12:178. [PMID: 36309763 PMCID: PMC9618215 DOI: 10.1186/s13578-022-00914-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Central nervous system (CNS) control of metabolism plays a pivotal role in maintaining energy balance. In the brain, Glucagon-like peptide 1 (GLP-1), encoded by the proglucagon 'Gcg' gene, produced in a distinct population of neurons in the nucleus tractus solitarius (NTS), has been shown to regulate feeding behavior leading to the suppression of appetite. However, neuronal networks that mediate endogenous GLP-1 action in the CNS on feeding and energy balance are not well understood. RESULTS We analyzed the distribution of GLP-1R-expressing neurons and axonal projections of NTS GLP-1-producing neurons in the mouse brain. GLP-1R neurons were found to be broadly distributed in the brain and specific forebrain regions, particularly the hypothalamus, including the arcuate nucleus of the hypothalamus (ARC), a brain region known to regulate energy homeostasis and feeding behavior, that receives dense NTSGcg neuronal projections. The impact of GLP-1 signaling in the ARC GLP-1R-expressing neurons and the impact of activation of ARC GLP-1R on food intake was examined. Application of GLP-1R specific agonist Exendin-4 (Exn-4) enhanced a proportion of the ARC GLP-1R-expressing neurons and pro-opiomelanocortin (POMC) neuronal action potential firing rates. Chemogenetic activation of the ARC GLP-1R neurons by using Cre-dependent hM3Dq AAV in the GLP-1R-ires-Cre mice, established that acute activation of the ARC GLP-1R neurons significantly suppressed food intake but did not have a strong impact on glucose homeostasis. CONCLUSIONS These results highlight the importance of central GLP-1 signaling in the ARC that express GLP-1R that upon activation, regulate feeding behavior.
Collapse
Affiliation(s)
- Ishnoor Singh
- grid.430387.b0000 0004 1936 8796The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA ,grid.17063.330000 0001 2157 2938Department of Physiology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8 Canada
| | - Le Wang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
| | - Baijuan Xia
- grid.430387.b0000 0004 1936 8796The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA ,grid.413458.f0000 0000 9330 9891School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, 550025 China
| | - Ji Liu
- grid.430387.b0000 0004 1936 8796The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA ,grid.59053.3a0000000121679639National Engineering Laboratory for Brain-Inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei, 230026 Anhui China
| | - Azeddine Tahiri
- grid.430387.b0000 0004 1936 8796The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA
| | - Abdelfattah El Ouaamari
- grid.430387.b0000 0004 1936 8796The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA ,grid.430387.b0000 0004 1936 8796Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA
| | - Michael B. Wheeler
- grid.17063.330000 0001 2157 2938Department of Physiology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8 Canada ,Metabolism Research Group, Division of Advanced Diagnostics, Toronto, ON Canada
| | - Zhiping P. Pang
- grid.430387.b0000 0004 1936 8796The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA ,grid.430387.b0000 0004 1936 8796Department of Neuroscience and Cell Biology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA ,grid.430387.b0000 0004 1936 8796Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901 USA
| |
Collapse
|
33
|
Insights into the Role of Glucagon Receptor Signaling in Metabolic Regulation from Pharmacological Inhibition and Tissue-Specific Knockout Models. Biomedicines 2022; 10:biomedicines10081907. [PMID: 36009454 PMCID: PMC9405517 DOI: 10.3390/biomedicines10081907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
While glucagon has long been recognized as the primary counter hormone to insulin’s actions, it has recently gained recognition as a metabolic regulator with its effects extending beyond control of glycemia. Recently developed models of tissue-specific glucagon receptor knockouts have advanced our understanding of this hormone, providing novel insight into the role it plays within organs as well as its systemic effects. Studies where the pharmacological blockade of the glucagon receptor has been employed have proved similarly valuable in the study of organ-specific and systemic roles of glucagon signaling. Studies carried out employing these tools demonstrate that glucagon indeed plays a role in regulating glycemia, but also in amino acid and lipid metabolism, systemic endocrine, and paracrine function, and in the response to cardiovascular injury. Here, we briefly review recent progress in our understanding of glucagon’s role made through inhibition of glucagon receptor signaling utilizing glucagon receptor antagonists and tissue specific genetic knockout models.
Collapse
|