1
|
Arefin TM, Börchers S, Olekanma D, Cramer SR, Sotzen MR, Zhang N, Skibicka KP. Sex-specific signatures of GLP-1 and amylin on resting state brain activity and functional connectivity in awake rats. Neuropharmacology 2025; 269:110348. [PMID: 39914619 PMCID: PMC11926989 DOI: 10.1016/j.neuropharm.2025.110348] [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: 10/08/2024] [Revised: 12/16/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Gut-produced glucagon-like peptide-1 (GLP-1) and pancreas-made amylin robustly reduce food intake by directly or indirectly affecting brain activity. While for both peptides a direct action in the hindbrain and the hypothalamus is likely, few studies examined their impact on whole brain activity in rodents and did so evaluating male rodents under anesthesia. However, both sex and anesthesia may significantly alter the influence of feeding controlling molecules on brain activity. Therefore, we investigated the effect of GLP-1 and amylin on brain activity and functional connectivity (FC) in awake adult male and female rats using resting-state functional magnetic resonance imaging (rsfMRI). We further examined the relationship between the altered brain activity or connectivity and subsequent food intake in response to amylin or GLP-1. We observed sex divergent effects of amylin and GLP-1 on the brain activity and FC patterns. Most importantly correlation analysis between FC and feeding behavior revealed that different brain areas potentially drive reduced food intake in male and female rats. Our findings underscore the distributed and distinctly sex divergent neural network engaged by each of these anorexic peptides and suggest that different brain areas may be the primary drivers of the feeding outcome in male and female rats. Moreover, prominent activity and connectivity alterations observed in brain areas not typically associated with feeding behavior in both sexes may either indicate novel feeding centers or alternatively suggest the involvement of these substances in behaviors beyond feeding and metabolism. The latter question is of potential translational significance as analogues of both amylin and GLP-1 are clinically utilized.
Collapse
Affiliation(s)
- Tanzil M Arefin
- Huck Institutes of Life Science, Pennsylvania State University, State College, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, USA; Center for Neurotechnology in Mental Health Research, Pennsylvania State University, University Park, USA; Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA; Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Stina Börchers
- Nutritional Sciences, Pennsylvania State University, University Park, PA, USA; Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Doris Olekanma
- Nutritional Sciences, Pennsylvania State University, University Park, PA, USA; Huck Institutes of Life Science, Pennsylvania State University, State College, PA, USA; The Neuroscience Graduate Program, Pennsylvania State University, University Park, USA
| | - Samuel R Cramer
- Huck Institutes of Life Science, Pennsylvania State University, State College, PA, USA; The Neuroscience Graduate Program, Pennsylvania State University, University Park, USA
| | - Morgan R Sotzen
- Nutritional Sciences, Pennsylvania State University, University Park, PA, USA; Huck Institutes of Life Science, Pennsylvania State University, State College, PA, USA
| | - Nanyin Zhang
- Huck Institutes of Life Science, Pennsylvania State University, State College, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, USA; Center for Neurotechnology in Mental Health Research, Pennsylvania State University, University Park, USA
| | - Karolina P Skibicka
- Nutritional Sciences, Pennsylvania State University, University Park, PA, USA; Huck Institutes of Life Science, Pennsylvania State University, State College, PA, USA; Institute of Neuroscience and Physiology, University of Gothenburg, Sweden.
| |
Collapse
|
2
|
Shen J, Wang M, Pang G, Zhang Y, Zhang J, Shi Y, Liu J, Zhan C. GLP-1 receptor agonist exendin-4 suppresses food intake by inhibiting hindbrain orexigenic NPY neurons. Am J Physiol Endocrinol Metab 2025; 328:E661-E674. [PMID: 40126941 DOI: 10.1152/ajpendo.00528.2024] [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: 12/31/2024] [Revised: 02/01/2025] [Accepted: 03/07/2025] [Indexed: 03/26/2025]
Abstract
Peripherally delivered glucagon-like peptide-1 (GLP-1)-based drugs suppress eating through their action in the brain. However, the specific neuronal mechanisms, especially their impacts on the orexigenic circuit, remain largely elusive. Neuropeptide Y (NPY) neurons in the nucleus tractus solitarius (NTS) are newly identified as orexigenic neurons with a potent eating-stimulating effect, but their responses to GLP-1 drugs are unknown. Through ex vivo electrophysiological recordings, we study the impacts of GLP-1 receptor (GLP-1R) agonist exendin-4 on NTSNPY neurons. We discovered that the GLP-1R agonist exendin-4 inhibits NTSNPY neuronal activity via GABAb receptors by augmenting presynaptic GABA release. We also explored the contribution of NTSNPY neurons to exendin-4-mediated eating suppression. Interestingly, chemogenetic activation of NTSNPY neurons effectively counteracted exendin-4-induced anorexigenic effect. Moreover, chemogenetic inhibition of NTSNPY neurons mimicked the eating-suppressing effect of exendin-4. Collectively, our findings highlight a population of orexigenic NTSNPY neurons that may be targeted by a GLP-1R agonist to suppress food intake, suggesting that this neuronal population has translational importance as a potential therapeutic target for obesity treatment.NEW & NOTEWORTHY This study discovers that the glucagon-like peptide-1 (GLP-1) receptor agonist exendin-4 indirectly inhibits the majority of orexigenic hindbrain NPY neurons via GABAb receptors by augmenting presynaptic GABA release. Chemogenetic activation of these NPY neurons effectively counteracts exendin-4 (Exn-4)-induced anorexigenic effect, whereas chemogenetic inhibition of them mimics the eating-suppressing effect of exendin-4. This study uncovers a mechanism by which Exn-4 inhibits orexigenic hindbrain NPY neurons, thereby providing new insights into how GLP-1 drugs suppress food intake.
Collapse
Affiliation(s)
- Jiayi Shen
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, People's Republic of China
| | - Mengtian Wang
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- School of Biomedical Engineering, School of Information Science and Technology, University of Science and Technology of China, Hefei, People's Republic of China
| | - Guodong Pang
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yan Zhang
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, People's Republic of China
| | - Jian Zhang
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- School of Biomedical Engineering, School of Information Science and Technology, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yuyan Shi
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, People's Republic of China
| | - Ji Liu
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- School of Biomedical Engineering, School of Information Science and Technology, University of Science and Technology of China, Hefei, People's Republic of China
| | - Cheng Zhan
- Department of Hematology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- Department of Endocrinology, The First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, People's Republic of China
| |
Collapse
|
3
|
Keel PK, Bodell LP, Ali SI, Starkey A, Trotta J, Luxama JW, Halfhide C, Hill NG, Appelbaum J, Williams DL. Examining Weight Suppression, Leptin Levels, Glucagon-Like Peptide 1 Response, and Reward-Related Constructs in Severity and Maintenance of Bulimic Syndromes: Protocol and Sample Characteristics for a Cross-Sectional and Longitudinal Study. JMIR Res Protoc 2025; 14:e66554. [PMID: 40198107 PMCID: PMC12015349 DOI: 10.2196/66554] [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: 09/16/2024] [Revised: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Bulimia nervosa and related syndromes (BN-S) characterized by binge eating vary considerably in illness severity and course. Using the Research Domain Criteria framework of the National Institute of Mental Health, we developed a model positing that the same set of physiological consequences of weight suppression (WS; defined as the difference between the highest and current adult body weight) contribute to binge-eating severity and maintenance by (1) increasing the drive or motivation to consume food (reward valuation effort [RVE]) and (2) decreasing the ability for food consumption to lead to a state of satiation or satisfaction (reward satiation). OBJECTIVE Our funded project aimed to test concurrent associations among WS, physiological factors (leptin concentrations and postprandial glucagon-like peptide 1 [GLP-1] response), behavioral indicators of RVE (breakpoint on progressive ratio tasks) and reward satiation (ad-lib test meal intake), self-report of these core constructs, and binge-eating severity in BN-S (aim 1); test prospective associations to determine whether WS predicts BN-S maintenance in longitudinal models and whether posited mediators also predict BN-S maintenance (aim 2); and determine whether associations between WS and BN-S severity and maintenance are mediated by alterations in leptin levels, GLP-1 response, RVE, and reward satiation (aim 3). METHODS We aimed to recruit a sample of 320 women with BN-S or noneating disorder controls, with BMI from 16 kg/m2 to 35 kg/m2, for our study. The study included diagnostic interviews; questionnaires; height, weight, and percentage of body fat measurements; weight history; fasting leptin level; postprandial GLP-1 and insulin responses to a fixed meal; and ad-lib meal and progressive ratio tasks to behaviorally measure reward satiation and RVE, respectively, at baseline, with at least 78.1% (250/320) of the participants providing data at 6- and 12-month follow-up visits. Data will be analyzed using structural equation models to test posited pathways. RESULTS Data collection began in November 2016 and ended in April 2023, pausing in-person data collection from March 2020 to February 2021 due to the COVID-19 pandemic. Of 399 eligible women enrolled, 290 (72.7%) provided clinical, behavioral, and biological data at baseline, and 249 (62.4%) provided follow-up data. Measures demonstrated strong psychometric properties. CONCLUSIONS We seek to identify biobehavioral predictors to inform treatments that target key factors influencing the severity and course of binge eating. These data, supported solely through federal funding, can inform questions emerging from recent interest and controversy surrounding the use of GLP-1 agonists for binge eating. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) RR1-10.2196/66554.
Collapse
Affiliation(s)
- Pamela K Keel
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Lindsay P Bodell
- Department of Psychology, Western University, London, ON, Canada
| | - Sarrah I Ali
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Austin Starkey
- Department of Psychology, Louisiana State University, Baton Rouge, LA, United States
| | - Jenna Trotta
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - J Woody Luxama
- College of Medicine, University of Central Florida, Orlando, FL, United States
| | | | - Naomi G Hill
- Department of Psychology, Ohio University, Athens, OH, United States
| | - Jonathan Appelbaum
- College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Diana L Williams
- Kravis Department of Integrated Sciences, Claremont McKenna College, Claremont, CA, United States
| |
Collapse
|
4
|
Holst JJ, Rosenkilde MM. Oxyntomodulin - past, present and future. Peptides 2025; 188:171393. [PMID: 40187415 DOI: 10.1016/j.peptides.2025.171393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
Almost since its discovery, glucagon was suspected to be formed in the gastrointestinal tract, and the L-cells were shown to contain glucagon-like immunoreactivity. This was due to the presence of two peptides that both contained the full glucagon sequence:glicentin of 69 amino acids and oxyntomodulin of 37 amino acids. While glicentin is a part of the glucagon precursor, proglucagon, and probably is inactive, oxyntomodulin, a fragment of glicentin, interacts although weakly with the glucagon as well as the GLP-1 receptor. However, in agreement with these activities, oxyntomodulin inhibited appetite and food intake in humans and inspired development of long acting, potent glucagon-GLP-1 co-agonists. Several such co-agonists are currently in clinical development and show promise because they combine GLP-1 like activities with those of glucagon agonism: additive weight loss and a stimulation of hepatic lipid metabolism with unique effectiveness on hepatic steatosis. They may therefore be effective in the treatment of metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- Jens Juul Holst
- The NovoNordisk Foundation Center for Basic Metabolic Research, Denmark; Department of Biomedical Sciences, the Panum institute, University of Copenhagen, Denmark.
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, the Panum institute, University of Copenhagen, Denmark.
| |
Collapse
|
5
|
Chilamakuri SN, N M, Thalla M, Velayutham R, Lee Y, Cho SM, Jung H, Natesan S. Role of Microneedles for Improved Treatment of Obesity: Progress and Challenges. Mol Pharm 2025. [PMID: 40167034 DOI: 10.1021/acs.molpharmaceut.4c01115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Obesity is a global metabolic health epidemic characterized by excessive lipid and fat accumulation, leading to severe conditions such as diabetes, cancer, and cardiovascular disease. Immediate attention and management of obesity-related health risks are most warranted. The imbalance between fat absorption, metabolic rate, and environmental and genetic factors is responsible for obesity. Treatment typically involves lifestyle modifications, pharmacotherapy, and surgery. While lifestyle changes are crucial, effective treatment often necessitates medication as a preferred adjunct strategy. However, medications commonly used, such as oral pharmacotherapy, often show side effects due to systemic exposure and, thus, may not effectively target the intended areas, leading to drug loss. On the other hand, transdermal administration of drugs with microneedle (MN)-based technologies, a painless drug delivery approach with patient compliance, is gaining interest as an alternative obesity treatment, as it directly targets adipose tissue via local delivery, minimizing system exposure and dose reduction. This Review addresses the pathophysiology of obesity, current treatment strategies, challenges in the treatment of obesity using conventional formulations, the importance of the use of nano-based medications through transdermal delivery, and the use of MNs as a promising platform for the effective delivery of nanoparticle-based anti-obesity medications. The potential of combining MNs with stimuli-responsive and non-responsive adjuvant therapies to enhance treatment efficacy and patient outcomes is explored. In addition, the limitations and future perspectives related to the use of MNs for obesity are addressed to highlight the transformative potential of this technology for obesity management. MNs hold promise in precisely delivering anti-obesity drugs while requiring lower dosages and minimizing side effects compared to conventional oral or injectable therapies and ultimately improving the quality of life for individuals struggling with obesity and its associated comorbidities.
Collapse
Affiliation(s)
- Sudarshan Naidu Chilamakuri
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| | - Manasa N
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| | - Maharshi Thalla
- Department of Pharmaceutical Sciences, Texas A&M University, Kingsville, Texas 78363, United States
| | - Ravichandiran Velayutham
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| | - Youjin Lee
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Republic of Korea
| | - Sung Min Cho
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Republic of Korea
| | - Hyungil Jung
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seoul 08389, Republic of Korea
- Department of Integrative Biotechnology, Yonsei University, 85 Songdogwahak-ro, Incheon 21983, Republic of Korea
| | - Subramanian Natesan
- Department of Pharmaceutics, Advance Formulation Laboratory, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| |
Collapse
|
6
|
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
|
7
|
Vear A, Heneka MT, Clemmensen C. Incretin-based therapeutics for the treatment of neurodegenerative diseases. Nat Metab 2025; 7:679-696. [PMID: 40211045 DOI: 10.1038/s42255-025-01263-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025]
Abstract
Neurodegenerative diseases (NDDs) represent a heterogeneous group of disorders characterized by progressive neuronal loss, which results in significant deficits in memory, cognition, motor skills, and sensory functions. As the prevalence of NDDs rises, there is an urgent unmet need for effective therapies. Current drug development approaches primarily target single pathological features of the disease, which could explain the limited efficacy observed in late-stage clinical trials. Originally developed for the treatment of obesity and diabetes, incretin-based therapies, particularly long-acting GLP-1 receptor (GLP-1R) agonists and GLP-1R-gastric inhibitory polypeptide receptor (GIPR) dual agonists, are emerging as promising treatments for NDDs. Despite limited conclusive preclinical evidence, their pleiotropic ability to reduce neuroinflammation, enhance neuronal energy metabolism and promote synaptic plasticity positions them as potential disease-modifying NDD interventions. In anticipation of results from larger clinical trials, continued advances in next-generation incretin mimetics offer the potential for improved brain access and enhanced neuroprotection, paving the way for incretin-based therapies as a future cornerstone in the management of NDDs.
Collapse
Affiliation(s)
- Anika Vear
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Stefater-Richards MA, Jhe G, Zhang YJ. GLP-1 Receptor Agonists in Pediatric and Adolescent Obesity. Pediatrics 2025; 155:e2024068119. [PMID: 40031990 DOI: 10.1542/peds.2024-068119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/11/2024] [Indexed: 03/05/2025] Open
Abstract
Obesity remains highly prevalent among children in the United States and is associated with an ever-increasing burden of obesity-related diseases. Effective pediatric obesity prevention and treatment will require both societal interventions and health care system innovation. One recent advancement is the approval of glucagon-like peptide-1 receptor agonists (GLP-1RAs) for use in adolescents. GLP-1RAs are notable for their effectiveness in weight management and in their ability to ameliorate obesity-related diseases. GLP-1RAs can be an important part of a comprehensive treatment plan for pediatric patients seeking obesity care, and we will review the pediatric clinician's considerations for their effective use. We discuss the history of obesity pharmacology and development of GLP-1RAs. We review the indications for use and common adverse reactions. We highlight the importance of mental health care for obesity treatment, with a focus on disordered eating behaviors and their intersection with obesity and pharmacologic treatment of obesity. Nutrition remains an important issue for obesity prevention and management, and we highlight nutritional concerns during GLP-1RA therapy. Finally, we discuss health inequities in obesity, the dangers of perpetuating these inequities if GLP-1RA access remains biased, and the opportunities for improvement.
Collapse
Affiliation(s)
- Margaret A Stefater-Richards
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Grace Jhe
- Division of Adolescent/Young Adult Medicine, Boston Children's Hospital, Boston, Massachusetts
- Department of Psychiatry and Behavioral Sciences, Boston Children's Hospital, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Yanjia Jason Zhang
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
9
|
J C, Me C, Mt C. Renoprotective mechanisms of glucagon-like peptide-1 receptor agonists. DIABETES & METABOLISM 2025; 51:101641. [PMID: 40127835 DOI: 10.1016/j.diabet.2025.101641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
Glucagon-like peptide-1 (GLP-1) is an incretin hormone, secreted from gut endocrine cells, which acts to potentiate nutrient-induced insulin secretion. Activation of its receptor, GLP-1R, decreases glucagon secretion and gastric emptying, thereby decreasing blood glucose and body weight. It is largely through these mechanisms that Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have transformed the treatment of type 2 diabetes. More recently, preclinical and clinical studies have reported that these agents have potent extra-pancreatic effects, exhibiting cardioprotective and renoprotective actions. The recent FLOW trial was the first multicentre clinical trial investigating the effect of GLP-1RAs on a primary renal outcome and reported robust evidence that GLP-1RAs are renoprotective. Studies in rodent models of renal injury have shown that gain and loss of GLP-1R signalling improves or deteriorates kidney function. However, the precise mechanisms responsible for renal benefits of GLP-1RAs are not yet fully understood. While prolonged activation of GLP-1 receptors (GLP-1R) has been shown to reverse diabetes-related disruptions in gene expression across various renal cell populations, GLP-1R expression in both rodent and human kidneys is thought to be primarily confined to certain vascular smooth muscle cells. This review discusses recent advances in our understanding of the effects of GLP-1 medicines on the kidney with a focus on indirect and direct mechanisms of action.
Collapse
Affiliation(s)
- Chen J
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, 3004, Victoria, Australia
| | - Cooper Me
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, 3004, Victoria, Australia
| | - Coughlan Mt
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, 3004, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University Parkville Campus, 381 Royal Parade, Parkville, 3052, Victoria, Australia.
| |
Collapse
|
10
|
James-Okoro PP, Lewis JE, Gribble FM, Reimann F. The role of GIPR in food intake control. Front Endocrinol (Lausanne) 2025; 16:1532076. [PMID: 40166681 PMCID: PMC11955450 DOI: 10.3389/fendo.2025.1532076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is one of two incretin hormones playing key roles in the control of food intake, nutrient assimilation, insulin secretion and whole-body metabolism. Recent pharmacological advances and clinical trials show that unimolecular co-agonists that target the receptors for the incretins - GIP and glucagon-like peptide 1 (GLP-1) - offer more effective treatment strategies for obesity and type 2 diabetes mellitus (T2D) compared with GLP-1 receptor (GLP1R) agonists alone, suggesting previously underappreciated roles of GIP in regulating food intake and body weight. The mechanisms by which GIP regulates energy balance remain controversial as both agonism and antagonism of the GIP receptor (GIPR) produce weight loss and improve metabolic outcomes in preclinical models. Recent studies have shown that GIPR signalling in the central nervous system (CNS), especially in regions of the brain that regulate energy balance, is essential for its action on appetite regulation. This finding has sparked interest in understanding the mechanisms by which GIP engages brain circuits to reduce food intake and body weight. In this review, we present key knowledge around the actions of GIP on food intake regulation and the potential mechanisms by which GIPR and GIPR/GLP1R agonists may regulate energy balance.
Collapse
Affiliation(s)
| | | | - Fiona Mary Gribble
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Frank Reimann
- Institute of Metabolic-Science-Metabolic Research Laboratories and MRC-Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Li S, Chen W, Ma S, Zhou X, Li J, Li B. Expandable konjac fiber modulates appetite and chyme digestion in vivo by stomach-intestine-brain axis. Int J Biol Macromol 2025; 307:142089. [PMID: 40090644 DOI: 10.1016/j.ijbiomac.2025.142089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Appetite regulation is a lifestyle intervention strategy to maintain health. The regulatory effects of dietary fiber (especially insoluble dietary fiber), as a crucial element of the nutritional composition, on appetite remain poorly understood. This study investigated modulatory effects of konjac fiber (KF, with high and low expansion) and konjac powder (KP) on chyme digestion, gastrointestinal hormones, intestinal microbiota, appetite genes in hypothalamus, GLP-1 receptor (GLP-1R) protein in various tissues of rats by dietary intervention. The results showed that highly-expanded konjac fiber (HKF) significantly delayed gastric emptying and inhibited hydrolysis of chyme. Konjac fiber (KF), especially HKF, and KP increased short-chain fatty acid (SCFA) content and plasma glucagon-like peptide-1 (GLP-1) levels. HKF upregulated the expression of GLP-1R protein in rat stomachs, nucleus tractus solitaries (NTS), and area postrema (AP) of rat brain, but down-regulated the expression of appetite gene AgRP/NPY in hypothalamus, thus, inhibiting appetite, reducing daily food intake and weight gain. Overall, this study reveals the mechanism through which expandable konjac fiber modulates appetite and chyme digestion in vivo by stomach-intestine-brain axis. Our findings provide an insight into the regulatory effects of insoluble dietary fiber on appetite and offered a valuable reference for the development of satiety-enhancing functional foods.
Collapse
Affiliation(s)
- Sha Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Wenjing Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Shaohua Ma
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Xiaorui Zhou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Jing Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China.
| |
Collapse
|
12
|
Ceulemans D, Deleus E, Benhalima K, van der Schueren B, Lannoo M, Devlieger R. Pregnancy After Metabolic Bariatric Surgery: Risks and Rewards for Mother and Child. BJOG 2025; 132:401-413. [PMID: 39663779 DOI: 10.1111/1471-0528.18032] [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: 02/12/2024] [Revised: 07/29/2024] [Accepted: 11/20/2024] [Indexed: 12/13/2024]
Abstract
As the prevalence of obesity increases worldwide, and lifestyle modification or pharmaceutical treatment yields insufficient results for patients with severe obesity, an increasing number of patients opt for metabolic bariatric surgery as an effective and durable treatment of this disease. Seeing as 80% of these patients are women, many of whom are of reproductive age, pregnancies after metabolic bariatric surgery become increasingly common. Metabolic bariatric surgery has many benefits for overall health and pregnancy outcomes, but certain risks are also reported. This leads to the rise of a new population of patients with their own specific needs regarding follow-up. This review discusses the various benefits and risks of these types of surgery for pregnancy. We provide an overview of the current state of the evidence and look into future research goals.
Collapse
Affiliation(s)
- Dries Ceulemans
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Ellen Deleus
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Katrien Benhalima
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Bart van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Matthias Lannoo
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Roland Devlieger
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
- Department of Obstetrics, Gynecology and Reproduction, St-Augustinus Hospital, Wilrijk, Belgium
| |
Collapse
|
13
|
Meiring S, Aydin Ö, van Baar ACG, van der Vossen EWJ, Rampanelli E, van Grieken NCT, Holleman F, Nieuwdorp M, Bergman JJGHM. From Endoscopic Inspection to Gene-Expression: A Thorough Assessment of the Duodenal Mucosa After Resurfacing-A Prospective Study. Dig Dis Sci 2025; 70:1052-1063. [PMID: 39779586 PMCID: PMC11920325 DOI: 10.1007/s10620-024-08710-4] [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: 02/08/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025]
Abstract
AIMS Duodenal Mucosal Resurfacing (DMR) is an endoscopic ablation technique aimed at improving glycemia in patients with type 2 diabetes mellitus (T2DM). Although the exact underlying mechanism is still unclear, it is postulated that the DMR-induced improvements are the result of changes in the duodenal mucosa. For this reason, we assessed macroscopic and microscopic changes in the duodenal mucosa induced by DMR + GLP-1RA. METHODS We included 16 patients with T2DM using basal insulin that received a combination treatment of a single DMR and GLP-1RA. Endoscopic evaluation was performed before the DMR procedure and 3 month after, and duodenal biopsies were obtained. Histological evaluation was performed and L and K cell density was calculated. In addition, gene-expression analysis and Western blotting was performed. RESULTS Endoscopic evaluation at 3 month showed duodenal mucosa with a normal appearance. In line, microscopic histological evaluation showed no signs of villous atrophy or inflammation and unchanged L and K cell density. Unbiased transcriptome profiling and western blotting revealed that PDZK1 expression was higher in responders at baseline and after DMR. GATA6 expression was significantly increased in responders after DMR compared to non-responders. CONCLUSION The absence of macroscopic and microscopic changes after 3 month suggest that improvements in glycemic parameters after DMR do not result from significant histological changes in duodenal mucosa. It is more likely that these improvements result from more subtle changes in enteroendocrine signaling. PDZK1 and GATA6 expression might play a role in DMR; this needs to be confirmed in pre-clinical studies.
Collapse
Affiliation(s)
- S Meiring
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Ö Aydin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
- Department of Bariatric Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - A C G van Baar
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - E W J van der Vossen
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - E Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - N C T van Grieken
- Department of Pathology, Amsterdam University Medical Centres, Location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - F Holleman
- Department of Internal Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - M Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - J J G H M Bergman
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Ma W, Zhang B, Chen X. Glucagon-like pepetide-1 receptor agonist suggests novel therapeutic options for hypothalamic obesity. J Diabetes Investig 2025; 16:357-359. [PMID: 39661011 PMCID: PMC11871401 DOI: 10.1111/jdi.14372] [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: 10/31/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
We briefly summarizes the mechanism of GLP-1RA therapy in HO both in rodents and in humans. We also summarized the clinical trials and case reports of GLP-1RA therapy in HO, especially the more and more often used semaglutide. We are hoping the therapy of GLP-1RA in HO will arouse more attention from clinicians in the future.
Collapse
Affiliation(s)
- Wanlu Ma
- Department of EndocrinologyChina‐Japan Friendship HospitalBeijingChina
| | - Bo Zhang
- Department of EndocrinologyChina‐Japan Friendship HospitalBeijingChina
| | - Xiaoping Chen
- Department of EndocrinologyChina‐Japan Friendship HospitalBeijingChina
| |
Collapse
|
15
|
Affiliation(s)
- Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto
| |
Collapse
|
16
|
Duran M, Willis JR, Dalvi N, Fokakis Z, Virkus SA, Hardaway JA. Integration of Glucagon-Like Peptide 1 Receptor Actions Through the Central Amygdala. Endocrinology 2025; 166:bqaf019. [PMID: 39888375 PMCID: PMC11850305 DOI: 10.1210/endocr/bqaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Understanding the detailed mechanism of action of glucagon-like peptide 1 receptor (GLP-1R) agonists on distinct topographic and genetically defined brain circuits is critical for improving the efficacy and mitigating adverse side effects of these compounds. In this mini-review, we propose that the central nucleus of the amygdala (CeA) is a critical mediator of GLP-1R agonist-driven hypophagia. Here, we review the extant literature demonstrating CeA activation via GLP-1R agonists across multiple species and through multiple routes of administration. The precise role of GLP-1Rs within the CeA is unclear but the site-specific GLP-1Rs may mediate distinct behavioral and physiological hallmarks of GLP-1R agonists on food intake. Thus, we propose important novel directions and methods to test the role of the CeA in mediating GLP-1R actions.
Collapse
Affiliation(s)
- Miguel Duran
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer R Willis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nilay Dalvi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zoe Fokakis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sonja A Virkus
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J Andrew Hardaway
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
17
|
Douros JD, Flak JN, Knerr PJ. The agony and the efficacy: central mechanisms of GLP-1 induced adverse events and their mitigation by GIP. Front Endocrinol (Lausanne) 2025; 16:1530985. [PMID: 39963285 PMCID: PMC11830610 DOI: 10.3389/fendo.2025.1530985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
| | - Jonathan N. Flak
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Patrick J. Knerr
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
| |
Collapse
|
18
|
Jin T, Chen YE. International Accolades for GLP-1 Research: Recognizing Pioneers in Diabetes and Obesity Treatment Across Five Prestigious Awards. Cardiovasc Drugs Ther 2025; 39:9-12. [PMID: 39470894 DOI: 10.1007/s10557-024-07630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 11/01/2024]
Affiliation(s)
- Tianru Jin
- Banting and Best Diabetes Centre, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada.
| | - Y Eugene Chen
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
19
|
Urbanik LA, Booth JL, Acharya NK, Evans BB, Grigson PS. Effect of acute treatment with the glucagon-like peptide-1 receptor agonist, liraglutide, and estrus phase on cue- and drug-induced fentanyl seeking in female rats. Behav Pharmacol 2025; 36:16-29. [PMID: 39718042 PMCID: PMC12013456 DOI: 10.1097/fbp.0000000000000805] [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] [Indexed: 12/25/2024]
Abstract
Opioid use disorder (OUD) is a crisis in the USA. Despite advances with medications for OUD, overdose deaths have continued to rise and are largely driven by fentanyl. We have previously found that male rats readily self-administer fentanyl, with evident individual differences in fentanyl taking, seeking, and reinstatement behaviors. We also have shown that acute treatment with the glucagon-like peptide-1 receptor (GLP-1R) agonist, liraglutide, can reduce fentanyl seeking behavior in male rats. However, given that females are significantly more vulnerable to drug-related cues, drug cravings, and to the development of OUD compared to males, it is imperative that we investigate the biological risk factors on fentanyl use disorder. Further, preclinical models report that females in estrus have increased fentanyl intake, more rapid development of OUD, and enhanced relapse vulnerability compared to those in a non-estrus phase. Thus, we aimed here to understand the effect of estrus phase on our model of OUD and on the effectiveness of acute liraglutide treatment. Herein, we show that female rats readily self-administer fentanyl (1.85 μg/infusion) intravenously, with marked individual differences in fentanyl taking behavior. Additionally, rats in the estrus phase exhibited greater fentanyl intake compared with those in a non-estrus phase, greater cue-induced fentanyl seeking, and greater drug-induced reinstatement of fentanyl seeking. Finally, acute liraglutide treatment (0.3 mg/kg s.c.) reduced cue-induced fentanyl seeking and blocked drug-induced reinstatement of fentanyl seeking, particularly when tested in estrus. Overall, these data support the broad effectiveness of acute GLP-1R agonists as a promising non-opioid treatment for OUD.
Collapse
Affiliation(s)
| | - Jennifer L Booth
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | | | | | | |
Collapse
|
20
|
Krieger JP, Daniels D, Lee S, Mastitskaya S, Langhans W. Glucagon-Like Peptide-1 Links Ingestion, Homeostasis, and the Heart. Compr Physiol 2025; 15:e7. [PMID: 39887844 PMCID: PMC11790259 DOI: 10.1002/cph4.7] [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: 11/02/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 02/01/2025]
Abstract
Glucagon-like peptide-1 (GLP-1), a hormone released from enteroendocrine cells in the distal small and large intestines in response to nutrients and other stimuli, not only controls eating and insulin release, but is also involved in drinking control as well as renal and cardiovascular functions. Moreover, GLP-1 functions as a central nervous system peptide transmitter, produced by preproglucagon (PPG) neurons in the hindbrain. Intestinal GLP-1 inhibits eating by activating vagal sensory neurons directly, via GLP-1 receptors (GLP-1Rs), but presumably also indirectly, by triggering the release of serotonin from enterochromaffin cells. GLP-1 enhances glucose-dependent insulin release via a vago-vagal reflex and by direct action on beta cells. Finally, intestinal GLP-1 acts on the kidneys to modulate electrolyte and water movements, and on the heart, where it provides numerous benefits, including anti-inflammatory, antiatherogenic, and vasodilatory effects, as well as protection against ischemia/reperfusion injury and arrhythmias. Hindbrain PPG neurons receive multiple inputs and project to many GLP-1R-expressing brain areas involved in reward, autonomic functions, and stress. PPG neuron-derived GLP-1 is involved in the termination of large meals and is implicated in the inhibition of water intake. This review details GLP-1's roles in these interconnected systems, highlighting recent findings and unresolved issues, and integrating them to discuss the physiological and pathological relevance of endogenous GLP-1 in coordinating these functions. As eating poses significant threats to metabolic, fluid, and immune homeostasis, the body needs mechanisms to mitigate these challenges while sustaining essential nutrient intake. Endogenous GLP-1 plays a crucial role in this "ingestive homeostasis."
Collapse
Affiliation(s)
- Jean-Philippe Krieger
- Jean-Philippe Krieger, Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Winterthurerstr. 260, 8057 Zurich
| | - Derek Daniels
- Department of Biological Sciences and the Center for Ingestive Behavior Research, University at Buffalo, the State University of New York, Buffalo NY 14260 USA
| | - Shin Lee
- Shin J. Lee, Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Svetlana Mastitskaya
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Dept. of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
21
|
Estato V, Obadia N, Chateaubriand PH, Figueiredo V, Curty M, Costa Silva M, Ferreira RGL, Santa-Ritta J, Campos Baroni M, Aragão A, Neno JOG, Vasconcellos CAM, Costa D'Avila J, Gomes Granja M, Caire de Castro Faria-Neto H. Semaglutide restores astrocyte-vascular interactions and blood-brain barrier integrity in a model of diet-induced metabolic syndrome. Diabetol Metab Syndr 2025; 17:2. [PMID: 39754250 PMCID: PMC11699651 DOI: 10.1186/s13098-024-01528-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/13/2024] [Indexed: 01/06/2025] Open
Abstract
INTRODUCTION Metabolic syndrome (MetS) is a metabolic disorder related to obesity and insulin resistance and is the primary determinant of the development of low-intensity chronic inflammation. This continuous inflammatory response culminates in neuroimmune-endocrine dysregulation responsible for the metabolic abnormalities and morbidities observed in individuals with MetS. Events such as the accumulation of visceral adipose tissue, increased plasma concentrations of free fatty acids, tissue hypoxia, and sympathetic hyperactivity in individuals with MetS may contribute to the activation of the innate immune response, which compromises cerebral microcirculation and the neurovascular unit, leading to the onset or progression of neurodegenerative diseases. OBJECTIVE This study aimed to evaluate the effects of chronic treatment with a GLP-1 receptor agonist (semaglutide) on cerebral microcirculation and neurovascular unit (NVU) integrity. METHODS C57BL/6 mice were fed a standard normolipidic diet or a high-fat diet (HFD) for 24 weeks and then treated for 4 weeks with semaglutide (HFD SEMA) or saline solution (HFD SAL). At the end of pharmacological treatment, biochemical analyses, immunohistochemistry analysis, and intravital microscopy of the brain microcirculation were carried out to quantify leukocyte-endothelium interactions and to assess structural capillary density, astrocyte coverage on cerebral vessels and microglial activation. RESULTS We observed that SEMA attenuates high-fat diet-induced metabolic alterations in mice fed with HFD for 24 weeks. SEMA also reversed cerebral microcirculation effects of HFD by reducing capillary rarefaction and the interaction of leukocytes in postcapillary brain venules. The HFD-SEMA group exhibited improved astrocyte coverage on vessels. However, SEMA did not reverse microglial activation. CONCLUSIONS Semaglutide can reverse microvascular rarefaction in metabolic syndrome by restoring the integrity of the neurovascular unit. Adverse dietary stimuli can compromise microglial homeostasis that is not reversed by semaglutide.
Collapse
Affiliation(s)
- Vanessa Estato
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation-Fiocruz, Campus Maré. Centro de Pesquisa, Inovação e Vigilância em Covid-19 e Emergências Sanitárias. Endereço: Av. Brasil, 4036-Bloco 2. Manguinhos, Rio de Janeiro, RJ, CEP 21040-361, Brazil.
- Medical School, Estácio-IDOMED, Rio de Janeiro, Brazil.
| | - Nathalie Obadia
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation-Fiocruz, Campus Maré. Centro de Pesquisa, Inovação e Vigilância em Covid-19 e Emergências Sanitárias. Endereço: Av. Brasil, 4036-Bloco 2. Manguinhos, Rio de Janeiro, RJ, CEP 21040-361, Brazil
- Pharmacy School, Universidade Estácio de Sá, Rio de Janeiro, Brazil
| | | | | | - Marcela Curty
- Medical School, Estácio-IDOMED, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | - Joana Costa D'Avila
- Laboratory of Pre-clinical Research, Iguaçu University, Rio de Janeiro, Brazil
| | - Marcelo Gomes Granja
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation-Fiocruz, Campus Maré. Centro de Pesquisa, Inovação e Vigilância em Covid-19 e Emergências Sanitárias. Endereço: Av. Brasil, 4036-Bloco 2. Manguinhos, Rio de Janeiro, RJ, CEP 21040-361, Brazil
- Medical School, Estácio-IDOMED, Rio de Janeiro, Brazil
| | - Hugo Caire de Castro Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation-Fiocruz, Campus Maré. Centro de Pesquisa, Inovação e Vigilância em Covid-19 e Emergências Sanitárias. Endereço: Av. Brasil, 4036-Bloco 2. Manguinhos, Rio de Janeiro, RJ, CEP 21040-361, Brazil
| |
Collapse
|
22
|
Alluri AA, Mohan Kurien M, Pokar NP, Madarapu A, Sadam S, Puvvala N, Seetharaman R. Exploring the therapeutic potential of GLP-1 receptor agonists in the management of obstructive sleep apnea: a comprehensive review. J Basic Clin Physiol Pharmacol 2025; 36:13-25. [PMID: 39804718 DOI: 10.1515/jbcpp-2024-0193] [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: 10/13/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025]
Abstract
Obstructive Sleep Apnea (OSA) is a prevalent sleep disorder marked by repeated episodes of partial or complete upper airway obstruction during sleep, which leads to intermittent hypoxia and fragmented sleep. These disruptions negatively impact cardiovascular health, metabolic function, and overall quality of life. Obesity is a major modifiable risk factor for OSA, as it contributes to both anatomical and physiological mechanisms that increase the likelihood of airway collapse during sleep. While continuous positive airway pressure (CPAP) therapy remains the gold standard for OSA treatment, its limitations - particularly issues with patient adherence - underscore the need for alternative or adjunct therapeutic options. One such option is the use of glucagon-like peptide-1 receptor agonists (GLP-1 RAs), which are widely recognized for their ability to reduce body weight and improve metabolic health. Emerging evidence suggests that GLP-1 RAs may offer therapeutic benefits in managing OSA, particularly by addressing obesity, a key contributor to the condition. This narrative review seeks to explore the role of GLP-1 RAs in the treatment of OSA, evaluating their efficacy in reducing OSA severity and discussing their broader clinical implications for future research and practice.
Collapse
Affiliation(s)
- Amruth Akhil Alluri
- Internal Medicine, American University of the Caribbean School of Medicine, Cupecoy, Sint Maarten
| | - Merin Mohan Kurien
- Acute Medicine, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, UK
| | - Nikhil Patel Pokar
- Internal Medicine, Malla Reddy Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Alekhya Madarapu
- Internal Medicine, Government Medical College Nizamabad, Nizamabad, Telangana, India
| | - Sreeja Sadam
- Internal Medicine, Government Medical College Mahabubnagar, Mahabubnagar, Telangana, India
| | - Nikhitha Puvvala
- Internal Medicine, Malla Reddy Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Rajmohan Seetharaman
- Pharmacology, MGM Medical College and Hospital, MGM Institute of Health Sciences, Nerul, Navi Mumbai, Maharashtra, India
| |
Collapse
|
23
|
D'Alessio DA, Kahn SE. The Development of Glucagon-Like Peptide 1 as a Therapeutic: The Triumph of the Lasker Award for Obesity Is a Victory for Diabetes Research. Diabetes 2025; 74:1-3. [PMID: 39705538 DOI: 10.2337/dbi24-0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Affiliation(s)
- David A D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC
- Division of Endocrinology, Department of Medicine, Duke University, Durham, NC
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, WA
| |
Collapse
|
24
|
D'Alessio DA, Kahn SE. The Development of Glucagon-Like Peptide 1 as a Therapeutic: The Triumph of the Lasker Award for Obesity Is a Victory for Diabetes Research. Diabetes Care 2025; 48:3-5. [PMID: 39705551 DOI: 10.2337/dci24-0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Affiliation(s)
- David A D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, NC
- Division of Endocrinology, Department of Medicine, Duke University, Durham, NC
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, WA
| |
Collapse
|
25
|
Sloop KW, Cox AL, Wainscott DB, White A, Droz BA, Stutsman C, Showalter AD, Suter TM, Dunbar JD, Snider BM, O'Farrell LS, Hewitt N, Ruble JC, Padgett LR, Woerly EM, Peterson JA, Coskun T, Liu Z, Coutant DE, Ai M, Emmerson PJ, Sangwung P, Willard FS. The pharmacological basis for nonpeptide agonism of the GLP-1 receptor by orforglipron. Sci Transl Med 2024; 16:eadp5765. [PMID: 39693407 DOI: 10.1126/scitranslmed.adp5765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/24/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Orally bioavailable, synthetic nonpeptide agonists (NPAs) of the glucagon-like peptide-1 receptor (GLP-1R) may offer an effective, scalable pharmacotherapy to address the metabolic disease epidemic. One of the first molecules in the emerging class of GLP-1R NPAs is orforglipron, which is in clinical development for treating type 2 diabetes and obesity. Here, we characterized the pharmacological properties of orforglipron in comparison with peptide-based GLP-1R agonists and other NPAs. Competition binding experiments using either [125I]GLP-1(7-36)NH2 or [3H]orforglipron indicated that orforglipron is a high-affinity [inhibition constant (Ki) = 1 nM], selective ligand of the human GLP-1R. Signal transduction assays showed that orforglipron has low intrinsic efficacy for effector activation and negligible β-arrestin recruitment. To evaluate GLP-1R engagement in vivo, mice expressing the human GLP-1R were administered orforglipron and subjected to a glucose tolerance test. Predicted receptor occupancy was calculated using the receptor Ki value of orforglipron and its unbound concentration in vivo that reduces hyperglycemia. These experiments revealed that low GLP-1R occupancy by orforglipron is sufficient to yield a full biological response. Moreover, in a model where CRISPR-Cas9 gene editing was used to sensitize the rat GLP-1R (Glp1rS33W) to GLP-1R NPAs, target engagement by orforglipron in the pancreas and brain was consistent with peptide-based GLP-1R agonists. Diet-induced obesity in Glp1rS33W rats enabled studies showing weight loss in animals orally administered orforglipron versus subcutaneous injection of GLP-1R agonist semaglutide. Furthermore, crossover studies indicated oral orforglipron can sustain efficacy initiated by parenteral semaglutide. The pharmacological properties of orforglipron may inform targeting of other peptide receptors with NPAs.
Collapse
Affiliation(s)
- Kyle W Sloop
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Amy L Cox
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - David B Wainscott
- Molecular Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indianapolis, IN 46285, USA
| | - Alex White
- Molecular Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indianapolis, IN 46285, USA
| | - Brian A Droz
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Cynthia Stutsman
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Aaron D Showalter
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Todd M Suter
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - James D Dunbar
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Brandy M Snider
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Libbey S O'Farrell
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Natalie Hewitt
- Molecular Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indianapolis, IN 46285, USA
| | - J Craig Ruble
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Leah R Padgett
- Molecular Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indianapolis, IN 46285, USA
| | - Eric M Woerly
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Jeffrey A Peterson
- Synthetic Molecule Design and Development, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Tamer Coskun
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Zhaomin Liu
- Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - David E Coutant
- Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Minrong Ai
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Paul J Emmerson
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Panjamaporn Sangwung
- Molecular Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indianapolis, IN 46285, USA
| | - Francis S Willard
- Molecular Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indianapolis, IN 46285, USA
| |
Collapse
|
26
|
Jalleh RJ, Plummer MP, Marathe CS, Umapathysivam MM, Quast DR, Rayner CK, Jones KL, Wu T, Horowitz M, Nauck MA. Clinical Consequences of Delayed Gastric Emptying With GLP-1 Receptor Agonists and Tirzepatide. J Clin Endocrinol Metab 2024; 110:1-15. [PMID: 39418085 DOI: 10.1210/clinem/dgae719] [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: 06/10/2024] [Revised: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
CONTEXT Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) are established therapeutics for type 2 diabetes and obesity. Among other mechanisms, they slow gastric emptying and motility of the small intestine. This helps to limit postprandial glycemic excursions and reduce chylomicron formation and triglyceride absorption. Conversely, motility effects may have detrimental consequences, eg, retained gastric contents at endoscopy or general anesthesia, potentially complicated by pulmonary aspiration or bowel obstruction. DATA ACQUISITION We searched the PubMed database for studies involving GLP-1RA therapy and adverse gastrointestinal/biliary events. DATA SYNTHESIS Retained gastric contents at the time of upper gastrointestinal endoscopy are found more frequently with GLP-1 RAs but rarely are associated with pulmonary aspiration. Well-justified recommendations for the periprocedural management of GLP-1RAs (eg, whether to withhold these medications and for how long) are compromised by limited evidence. Important aspects to be considered are (1) their long half-lives, (2) the capacity of GLP-1 receptor agonism to slow gastric emptying even at physiological GLP-1 concentrations, (c) tachyphylaxis observed with prolonged treatment, and (d) the limited effect on gastric emptying in individuals with slow gastric emptying before initiating treatment. Little information is available on the influence of diabetes mellitus itself (ie, in the absence of GLP-1 RA treatment) on retained gastric contents and pulmonary aspiration. CONCLUSION Prolonged fasting periods regarding solid meal components, point-of-care ultrasound examination for retained gastric content, and the use of prokinetic medications like erythromycin may prove helpful and represent an important area needing further study to increase patient safety for those treated with GLP-1 RAs.
Collapse
Affiliation(s)
- Ryan J Jalleh
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Mark P Plummer
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Intensive Care Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Chinmay S Marathe
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Mahesh M Umapathysivam
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Southern Adelaide Diabetes and Endocrine Service, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Daniel R Quast
- Diabetes, Endocrinology, Metabolism Section, Medical Department I, Katholisches Klinikum Bochum gGmbH, Sankt Josef-Hospital, Ruhr-University, D-44791 Bochum, Germany
| | - Christopher K Rayner
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Karen L Jones
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Tongzhi Wu
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Michael Horowitz
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Michael A Nauck
- Diabetes, Endocrinology, Metabolism Section, Medical Department I, Katholisches Klinikum Bochum gGmbH, Sankt Josef-Hospital, Ruhr-University, D-44791 Bochum, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, D-17475 Greifswald, Germany
| |
Collapse
|
27
|
Zarei M, Sahebi Vaighan N, Farjoo MH, Talebi S, Zarei M. Incretin-based therapy: a new horizon in diabetes management. J Diabetes Metab Disord 2024; 23:1665-1686. [PMID: 39610543 PMCID: PMC11599551 DOI: 10.1007/s40200-024-01479-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/22/2024] [Indexed: 11/30/2024]
Abstract
Diabetes mellitus, a metabolic syndrome characterized by hyperglycemia and insulin dysfunction, often leads to serious complications such as neuropathy, nephropathy, retinopathy, and cardiovascular disease. Incretins, gut peptide hormones released post-nutrient intake, have shown promising therapeutic effects on these complications due to their wide-ranging biological impacts on various body systems. This review focuses on the role of incretin-based therapies, particularly Glucagon-like peptide-1 (GLP-1) agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors, in managing diabetes and its complications. We also discuss the potential of novel agents like semaglutide, a recently approved oral compound, and dual/triple agonists targeting GLP-1/GIP, GLP-1/glucagon, and GLP-1/GIP/glucagon receptors, which are currently under investigation. The review aims to provide a comprehensive understanding of the beneficial impacts of natural incretins and the therapeutic potential of incretin-based therapies in diabetes management.
Collapse
Affiliation(s)
- Malek Zarei
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navideh Sahebi Vaighan
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Farjoo
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soosan Talebi
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Zarei
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
- John B. Little Center for Radiation Sciences, Harvard T.H Chan School of Public Health, Boston, MA USA
| |
Collapse
|
28
|
Aguggia J, Fernandez G, Cassano D, Mustafá ER, Rodríguez SS, Cantel S, Fehrentz JA, Raingo J, Schiöth HB, Habib AM, De Francesco PN, Perello M. Selective Colocalization of GHSR and GLP-1R in a Subset of Hypothalamic Neurons and Their Functional Interaction. Endocrinology 2024; 166:bqae160. [PMID: 39737802 DOI: 10.1210/endocr/bqae160] [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: 09/23/2024] [Indexed: 01/01/2025]
Abstract
The GH secretagogue receptor (GHSR) and the glucagon-like peptide-1 receptor (GLP-1R) are G protein-coupled receptors with critical, yet opposite, roles in regulating energy balance. Interestingly, these receptors are expressed in overlapping brain regions. However, the extent to which they target the same neurons and engage in molecular crosstalk remains unclear. To explore the potential colocalization of GHSR and GLP-1R in specific neurons, we performed detailed mapping of cells positive for both receptors using GHSR-eGFP reporter mice or wild-type mice infused with fluorescent ghrelin, alongside an anti-GLP-1R antibody. We found that GHSR+ and GLP-1R+ cells are largely segregated in the mouse brain. The highest overlap was observed in the hypothalamic arcuate nucleus, where 15% to 20% of GHSR+ cells were also GLP-1R+ cells. Additionally, we examined RNA-sequencing datasets from mouse and human brains to assess the fraction and distribution of neurons expressing both receptors, finding that double-positive Ghsr+/Glp1r+ cells are highly segregated, with a small subset of double-positive Ghsr+/Glp1r+ cells representing <10% of all Ghsr+ or Glp1r+ cells, primarily enriched in the hypothalamus. Furthermore, we conducted functional studies using patch-clamp recordings in a heterologous expression system to assess potential crosstalk in regulating presynaptic calcium channels. We provide the first evidence that liraglutide-evoked GLP-1R activity inhibits presynaptic channels, and that the presence of one GPCR attenuates the inhibitory effects of ligand-evoked activity mediated by the other on presynaptic calcium channels. In conclusion, while GHSR and GLP-1R can engage in molecular crosstalk, they are largely segregated across most neuronal types within the brain.
Collapse
Affiliation(s)
- Julieta Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Silvia S Rodríguez
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jesica Raingo
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| |
Collapse
|
29
|
Martinelli S, Mazzotta A, Longaroni M, Petrucciani N. Potential role of glucagon-like peptide-1 (GLP-1) receptor agonists in substance use disorder: A systematic review of randomized trials. Drug Alcohol Depend 2024; 264:112424. [PMID: 39288591 DOI: 10.1016/j.drugalcdep.2024.112424] [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: 05/10/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Increasing evidence suggests that GLP-1 receptor agonists (GLP-1RA) have a potential use in addiction treatment. Few studies have assessed the impact of GLP-1RA on substance use disorder (SUD), particularly in humans. The study aimed to do systematic review of clinical trials to assess GLP-1RA's effect on reducing SUD in patients. METHODS The scientific literature was reviewed using the MEDLINE, Scopus and Cochrane Library databases, following PRISMA guidelines. Studies including patients with a diagnosis of SU who were treated with GLP-1RA were selected. The primary outcome was GLP-1RA's therapeutic effect on SUD, and the secondary outcomes were therapeutic effects of GLP-1RA on weight, BMI and HbA1c. RESULTS 1218 studies were retrieved, resulting in 507 papers after title and abstract screening. Following full-text review, only 5 articles met inclusion criteria. We incorporated a total of 630 participants utilizing Exenatide (n=3) and Dulaglutide (n=2) as GLP-1RAs. Therapeutic effect of GLP-1RA on SUD was assessed in 5 studies, with 3 demonstrating a significant decrease in SUD (alcohol and nicotine). GLP-1RA's impact on body weight, BMI, and HbA1c, was reported in 3 studies. These revealed a notable reduction in these parameters among the GLP-1RA treated group. CONCLUSION This review will give an overview of current new findings in human studies; we suggest that the effects of GLP-1RA in SUD is a possible new option of therapy in addiction medicine.
Collapse
Affiliation(s)
- Silvia Martinelli
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy; Department of Mental Health, Local Health Authority Viterbo, Viterbo, Italy
| | - Alessandro Mazzotta
- Department of Surgery, M.G. General Vannini Hospital, Istituto Figlie Di San Camillo, Rome, Italy
| | - Mattia Longaroni
- Department of Surgery, Santa Maria della Misericordia Hospital, University of Perugia, Italy
| | - Niccolò Petrucciani
- Department of Medical and Surgical Sciences and Translational Medicine, Division of General and Hepatobiliary Surgery, St. Andrea Hospital, Sapienza University of Rome, Italy.
| |
Collapse
|
30
|
Mahishi D, Agrawal N, Jiang W, Yapici N. From Mammals to Insects: Exploring the Genetic and Neural Basis of Eating Behavior. Annu Rev Genet 2024; 58:455-485. [PMID: 39585905 DOI: 10.1146/annurev-genet-111523-102202] [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] [Indexed: 11/27/2024]
Abstract
Obesity and anorexia are life-threatening diseases that are still poorly understood at the genetic and neuronal levels. Patients suffering from these conditions experience disrupted regulation of food consumption, leading to extreme weight gain or loss and, in severe situations, death from metabolic dysfunction. Despite the development of various behavioral and pharmacological interventions, current treatments often yield limited and short-lived success. To address this, a deeper understanding of the genetic and neural mechanisms underlying food perception and appetite regulation is essential for identifying new drug targets and developing more effective treatment methods. This review summarizes the progress of past research in understanding the genetic and neural mechanisms controlling food consumption and appetite regulation, focusing on two key model organisms: the fruit fly Drosophila melanogaster and the mouse Mus musculus. These studies investigate how the brain senses energy and nutrient deficiency, how sensory signals trigger appetitive behaviors, and how food intake is regulated through interconnected neural circuits in the brain.
Collapse
Affiliation(s)
- Deepthi Mahishi
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Naman Agrawal
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Wenshuai Jiang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| |
Collapse
|
31
|
Li H, Fang Y, Wang D, Shi B, Thompson GJ. Impaired brain glucose metabolism in glucagon-like peptide-1 receptor knockout mice. Nutr Diabetes 2024; 14:86. [PMID: 39389952 PMCID: PMC11466955 DOI: 10.1038/s41387-024-00343-w] [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: 01/26/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Quantitative mapping of the brain's metabolism is a critical tool in studying and diagnosing many conditions, from obesity to neurodegenerative diseases. In particular, noninvasive approaches are urgently required. Recently, there have been promising drug development approaches for the treatment of disorders related to glucose metabolism in the brain and, therefore, against obesity-associated diseases. One of the most important drug targets to emerge has been the Glucagon-like peptide-1 (GLP-1) and its receptor (GLP-1R). GLP and GLP-1R play an important role in regulating blood sugar and maintaining energy homeostasis. However, the macroscopic effects on brain metabolism and function due to the presence of GLP-1R are unclear. METHODS To explore the physiological role of GLP-1R in mouse brain glucose metabolism, and its relationship to brain function, we used three methods. We used deuterium magnetic resonance spectroscopy (DMRS) to provide quantitative information about metabolic flux, fluorodeoxyglucose positron emission tomography (FDG-PET) to measure brain glucose metabolism, and resting state-functional MRI (rs-fMRI) to measure brain functional connectivity. We used these methods in both mice with complete GLP-1R knockout (GLP-1R KO) and wild-type C57BL/6N (WT) mice. RESULTS The metabolic rate of GLP-1R KO mice was significantly slower than that of WT mice (p = 0.0345, WT mice 0.02335 ± 0.057 mM/min, GLP-1R KO mice 0.01998 ± 0.07 mM/min). Quantification of the mean [18F]FDG signal in the whole brain also showed significantly reduced glucose uptake in GLP-1R KO mice versus control mice (p = 0.0314). Observing rs-fMRI, the functional brain connectivity in GLP-1R KO mice was significantly lower than that in the WT group (p = 0.0032 for gFCD, p = 0.0002 for whole-brain correlation, p < 0.0001 for ALFF). CONCLUSIONS GLP-1R KO mice exhibit impaired brain glucose metabolism to high doses of exogenous glucose, and they also have reduced functional connectivity. This suggests that the GLP-1R KO mouse model may serve as a model for correlated metabolic and functional connectivity loss.
Collapse
Affiliation(s)
- Hui Li
- iHuman Institute, ShanghaiTech University, Shanghai, China.
| | - Yujiao Fang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Da Wang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bowen Shi
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | | |
Collapse
|
32
|
Nestor LJ, Ersche KD. Gut Hormones: Possible Mediators of Addictive Disorders? Eur Addict Res 2024; 30:339-346. [PMID: 39389039 DOI: 10.1159/000540743] [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: 04/16/2024] [Accepted: 08/02/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Alcohol and drug dependence are major health and economic burdens to society. One of the major challenges to reducing this burden will be to develop more effective and better tolerated medications that target alternative mechanisms in the brain. While the dopamine system has been well characterized for mediating the reward value of drugs, there is evidence that the endocrine system also conveys signals to the same neural systems using gut hormones. SUMMARY These gut hormones, produced in the stomach and intestine and that regulate food intake, have also been shown to control the use of other substances, such as alcohol and drugs of abuse. Examples of such hormones are ghrelin and glucagon-like peptide-1, which exert their effects on dopamine transmission in parts of the brain known to be involved in some of the core features of addiction, such as reward sensitivity. KEY MESSAGES This raises the possibility that gut hormone systems may play a pivotal role in addictive disorders. This review will briefly outline emerging evidence that the ghrelin and glucagon-like peptide-1 hormones are contrasting mediators of alcohol and drug use and may present a promising alternative target for treatment intervention in addictive disorders.
Collapse
Affiliation(s)
- Liam J Nestor
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Karen D Ersche
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Systems Neuroscience, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
33
|
Yu J, Gao M, Wang L, Guo X, Liu X, Sheng M, Cheng S, Guo Y, Wang J, Zhao C, Guo W, Zhang Z, Liu Y, Hu C, Ma X, Xie C, Zhang Q, Xu L. An insoluble cellulose nanofiber with robust expansion capacity protects against obesity. Int J Biol Macromol 2024; 277:134401. [PMID: 39097049 DOI: 10.1016/j.ijbiomac.2024.134401] [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: 01/12/2024] [Revised: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
An imbalance between energy intake and energy expenditure predisposes obesity and its related metabolic diseases. Soluble dietary fiber has been shown to improve metabolic homeostasis mainly via microbiota reshaping. However, the application and metabolic effects of insoluble fiber are less understood. Herein, we employed nanotechnology to design citric acid-crosslinked carboxymethyl cellulose nanofibers (CL-CNF) with a robust capacity of expansion upon swelling. Supplementation with CL-CNF reduced food intake and delayed digestion rate in mice by occupying stomach. Besides, CL-CNF treatment mitigated diet-induced obesity and insulin resistance in mice with enhanced energy expenditure, as well as ameliorated inflammation in adipose tissue, intestine and liver and reduced hepatic steatosis, without any discernible signs of toxicity. Additionally, CL-CNF supplementation resulted in enrichment of probiotics such as Bifidobacterium and decreased in the relative abundances of deleterious microbiota expressing bile salt hydrolase, which led to increased levels of conjugated bile acids and inhibited intestinal FXR signaling to stimulate the release of GLP-1. Taken together, our findings demonstrate that CL-CNF administration protects mice from diet-induced obesity and metabolic dysfunction by reducing food intake, enhancing energy expenditure and remodeling gut microbiota, making it a potential therapeutic strategy against metabolic diseases.
Collapse
Affiliation(s)
- Jian Yu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyuan Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaodi Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Maozheng Sheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shimiao Cheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yingying Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiawen Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Cheng Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wenxiu Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhe Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yameng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xinran Ma
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China; Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, China.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Qiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
34
|
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
|
35
|
Holst JJ. GLP-1 physiology in obesity and development of incretin-based drugs for chronic weight management. Nat Metab 2024; 6:1866-1885. [PMID: 39160334 DOI: 10.1038/s42255-024-01113-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024]
Abstract
The introduction of the highly potent incretin receptor agonists semaglutide and tirzepatide has marked a new era in the treatment of type 2 diabetes and obesity. With normalisation of glycated haemoglobin levels and weight losses around 15-25%, therapeutic goals that were previously unrealistic are now within reach, and clinical trials have documented that these effects are associated with reduced risk of cardiovascular events and premature mortality. Here, I review this remarkable development from the earliest observations of glucose lowering and modest weight losses with native glucagon-like peptide (GLP)-1 and short acting compounds, to the recent development of highly active formulations and new molecules. I will classify these agents as GLP-1-based therapies in the understanding that these compounds or combinations may have actions on other receptors as well. The physiology of GLP-1 is discussed as well as its mechanisms of actions in obesity, in particular, the role of sensory afferents and GLP-1 receptors in the brain. I provide details regarding the development of GLP-1 receptor agonists for anti-obesity therapy and discuss the possible mechanism behind their beneficial effects on adverse cardiovascular events. Finally, I highlight new pharmacological developments, including oral agents, and discuss important questions regarding maintenance therapy.
Collapse
Affiliation(s)
- Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences. Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
36
|
Haller N, Lutz TA. Incretin therapy in feline diabetes mellitus - A review of the current state of research. Domest Anim Endocrinol 2024; 89:106869. [PMID: 38870560 DOI: 10.1016/j.domaniend.2024.106869] [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: 03/17/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Incretin hormones potentiate the glucose-induced insulin secretion following enteral nutrient intake. The best characterised incretin hormones are glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) which are produced in and secreted from the gut in response to nutrient ingestion. The property of incretins to enhance endogenous insulin secretion only at elevated blood glucose levels makes them interesting therapeutics for type 2 diabetes mellitus with a better safety profile than exogenous insulin. While incretin therapeutics (especially GLP-1 agonists, and more recently also GLP-1 / GIP dual agonists and other drugs that influence the incretin metabolism (e.g., dipeptidyl peptidase-4 (DPP-4) inhibitors)) are already widely used treatment options for human type 2 diabetes, these drugs are not yet approved for the therapy of feline diabetes mellitus. This review provides an introduction to incretins and feline diabetes mellitus in general and summarises the current study situation on incretins as therapeutics for feline diabetes mellitus to assess their possible future potential in feline medicine. Studies to date on the use of GLP-1 receptor agonists (GLP-1RA) in healthy cats largely confirm their insulinotropic effect known from other species. In diabetic cats, GLP-1RAs appear to significantly reduce glycaemic variability (GV, an indicator for the quality of glycaemic control), which is important for the management of the disease and prevention of long-term complications. However, for widespread use in feline diabetes mellitus, further studies are required that include larger numbers of diabetic cats, and that consider and test a possible need for dose adjustments to overweight and diabetic cats. Also evaluation of the outcome of GLP-1RA monotherapy will be neceessary.
Collapse
Affiliation(s)
- Nina Haller
- Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 204, CH 8057 Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH 8057 Zurich, Switzerland.
| |
Collapse
|
37
|
Nair P. QnAs with Svetlana Mojsov, Joel Habener, and Lotte Bjerre Knudsen: Winners of the 2024 Lasker~DeBakey Clinical Medical Research Award. Proc Natl Acad Sci U S A 2024; 121:e2416868121. [PMID: 39297682 PMCID: PMC11441505 DOI: 10.1073/pnas.2416868121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
|
38
|
Friedman JM. The discovery and development of GLP-1 based drugs that have revolutionized the treatment of obesity. Proc Natl Acad Sci U S A 2024; 121:e2415550121. [PMID: 39297680 PMCID: PMC11441540 DOI: 10.1073/pnas.2415550121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024] Open
Abstract
The 2024 Lasker~DeBakey Clinical Medical Research Award has been given to Joel Habener and Svetlana Mojsov for their discovery of a new hormone GLP-1(7-37) and to Lotte Knudsen for her role in developing sustained acting versions of this hormone as a treatment for obesity. Each of the three had a distinct set of skills that made this advance possible; Habener is an endocrinologist and molecular biologist, Mojsov is a peptide chemist, and Knudsen is a pharmaceutical scientist. Their collective efforts have done what few thought possible-the development of highly effective medicines for reducing weight. Their research has also solved a mystery that began more than a century ago.
Collapse
|
39
|
Burchat N, Vidola J, Pfreundschuh S, Sharma P, Rizzolo D, Guo GL, Sampath H. Intestinal Stearoyl-CoA Desaturase-1 Regulates Energy Balance via Alterations in Bile Acid Homeostasis. Cell Mol Gastroenterol Hepatol 2024; 18:101403. [PMID: 39278403 PMCID: PMC11546130 DOI: 10.1016/j.jcmgh.2024.101403] [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: 01/25/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND & AIMS Stearoyl-CoA desaturase-1 (SCD1) converts saturated fatty acids into monounsaturated fatty acids and plays an important regulatory role in lipid metabolism. Previous studies have demonstrated that mice deficient in SCD1 are protected from diet-induced obesity and hepatic steatosis due to altered lipid assimilation and increased energy expenditure. Previous studies in our lab have shown that intestinal SCD1 modulates intestinal and plasma lipids and alters cholesterol metabolism. Here, we investigated a novel role for intestinal SCD1 in the regulation of systemic energy balance. METHODS To interrogate the role of intestinal SCD1 in modulating whole body metabolism, intestine-specific Scd1 knockout (iKO) mice were maintained on standard chow diet or challenged with a high-fat diet (HFD). Studies included analyses of bile acid content and composition, and metabolic phenotyping, including body composition, indirect calorimetry, glucose tolerance analyses, quantification of the composition of the gut microbiome, and assessment of bile acid signaling pathways. RESULTS iKO mice displayed elevated plasma and hepatic bile acid content and decreased fecal bile acid excretion, associated with increased expression of the ileal bile acid uptake transporter, Asbt. In addition, the alpha and beta diversity of the gut microbiome was reduced in iKO mice, with several alterations in microbe species being associated with the observed increases in plasma bile acids. These increases in plasma bile acids were associated with increased expression of TGR5 targets, including Dio2 in brown adipose tissue and elevated plasma glucagon-like peptide-1 levels. Upon HFD challenge, iKO mice had reduced metabolic efficiency apparent through decreased weight gain despite higher food intake. Concomitantly, energy expenditure was increased, and glucose tolerance was improved in HFD-fed iKO mice. CONCLUSIONS Our results indicate that deletion of intestinal SCD1 has significant impacts on bile acid homeostasis and whole-body energy balance, likely via activation of TGR5.
Collapse
Affiliation(s)
- Natalie Burchat
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey
| | - Jeanine Vidola
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey
| | - Sarah Pfreundschuh
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey
| | - Priyanka Sharma
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey
| | - Daniel Rizzolo
- Ernest Mario School of Pharmacy, Rutgers University, New Brunswick, New Jersey
| | - Grace L Guo
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey; Ernest Mario School of Pharmacy, Rutgers University, New Brunswick, New Jersey
| | - Harini Sampath
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey; Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey.
| |
Collapse
|
40
|
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
|
41
|
Liu CC, Khan A, Seban N, Littlejohn N, Shah A, Srinivasan S. A homeostatic gut-to-brain insulin antagonist restrains neuronally stimulated fat loss. Nat Commun 2024; 15:6869. [PMID: 39127676 PMCID: PMC11316803 DOI: 10.1038/s41467-024-51077-3] [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: 09/29/2023] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In C. elegans mechanisms by which peripheral organs relay internal state information to the nervous system remain unknown, although strong evidence suggests that such signals do exist. Here we report the discovery of a peptide of the ancestral insulin superfamily called INS-7 that functions as an enteroendocrine peptide and is secreted from specialized cells of the intestine. INS-7 secretion is stimulated by food withdrawal, increases during fasting and acts as a bona fide gut-to-brain peptide that attenuates the release of a neuropeptide that drives fat loss in the periphery. Thus, INS-7 functions as a homeostatic signal from the intestine that gates the neuronal drive to stimulate fat loss during food shortage. Mechanistically, INS-7 functions as an antagonist at the canonical DAF-2 receptor and functions via FOXO and AMPK signaling in ASI neurons. Phylogenetic analysis suggests that INS-7 bears greater resemblance to members of the broad insulin/relaxin superfamily than to conventional mammalian insulin and IGF peptides. The discovery of an endogenous insulin antagonist secreted by specialized intestinal cells with enteroendocrine functions suggests unexpected and important properties of the intestine and its role in directing neuronal functions.
Collapse
Affiliation(s)
- Chung-Chih Liu
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, San Diego, CA, USA
| | - Ayub Khan
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Nicolas Seban
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Nicole Littlejohn
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Aayushi Shah
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA
| | - Supriya Srinivasan
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, San Diego, CA, USA.
| |
Collapse
|
42
|
Hansen LS, Gasbjerg LS, Brønden A, Dalsgaard NB, Bahne E, Stensen S, Hellmann PH, Rehfeld JF, Hartmann B, Wewer Albrechtsen NJ, Holst JJ, Vilsbøll T, Knop FK. The role of glucagon-like peptide 1 in the postprandial effects of metformin in type 2 diabetes: a randomized crossover trial. Eur J Endocrinol 2024; 191:192-203. [PMID: 39049802 DOI: 10.1093/ejendo/lvae095] [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: 04/11/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024]
Abstract
AIMS Although metformin is widely used for treatment of type 2 diabetes (T2D), its glucose-lowering mechanism remains unclear. Using the glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) antagonist exendin(9-39)NH2, we tested the hypothesis that postprandial GLP-1-mediated effects contribute to the glucose-lowering potential of metformin in T2D. METHODS In a randomized, placebo-controlled, double-blind, crossover study, 15 individuals with T2D (median HbA1c 50 mmol/mol [6.7%], body mass index 30.1 kg/m2, age 71 years) underwent, in randomized order, 14 days of metformin and placebo treatment, respectively. Each treatment period was preceded by 14 days without any glucose-lowering medicine and concluded by two 4 h mixed meal tests performed in randomized order and separated by >24 h with either continuous intravenous exendin(9-39)NH2 or saline infusion. RESULTS Compared to placebo, metformin treatment lowered fasting plasma glucose (mean of differences [MD] 1.4 mmol/L × min [95% CI 0.8-2.0]) as well as postprandial plasma glucose excursions during both saline infusion (MD 186 mmol/L × min [95% CI 64-307]) and exendin(9-39)NH2 infusion (MD 268 mmol/L × min [95% CI 108-427]). The metformin-induced improvement in postprandial glucose tolerance was unaffected by GLP-1R antagonization (MD 82 mmol/L × min [95% CI -6564-170]). Metformin treatment increased fasting plasma GLP-1 (MD 1.7 pmol/L × min [95% CI 0.39-2.9]) but did not affect postprandial GLP-1 responses (MD 820 pmol/L × min [95% CI -1750-111]). CONCLUSIONS Using GLP-1R antagonization, we could not detect GLP-1-mediated postprandial glucose-lowering effect of metformin in individuals with T2D. We show that 2 weeks of metformin treatment increases fasting plasma GLP-1, which may contribute to metformin's beneficial effect on fasting plasma glucose in T2D. Trial registration: Clinicaltrials.gov NCT03246451.
Collapse
Affiliation(s)
- Laura S Hansen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Andreas Brønden
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Pharmacology, Copenhagen University Hospital-Bispebjerg and Frederiksberg, DK-2400 Copenhagen NV, Denmark
| | - Niels B Dalsgaard
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Emilie Bahne
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Signe Stensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Pernille H Hellmann
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | | | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Tina Vilsbøll
- Clinical Research, Steno Diabetes Center Copenhagen, University of Copenhagen, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, University of Copenhagen, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
43
|
Ansari S, Khoo B, Tan T. Targeting the incretin system in obesity and type 2 diabetes mellitus. Nat Rev Endocrinol 2024; 20:447-459. [PMID: 38632474 DOI: 10.1038/s41574-024-00979-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/19/2024]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are widespread, non-communicable diseases that are responsible for considerable levels of morbidity and mortality globally, primarily in the form of cardiovascular disease (CVD). Changes to lifestyle and behaviour have insufficient long-term efficacy in most patients with these diseases; metabolic surgery, although effective, is not practically deliverable on the scale that is required. Over the past two decades, therapies based on incretin hormones, spearheaded by glucagon-like peptide 1 (GLP1) receptor agonists (GLP1RAs), have become the treatment of choice for obesity and T2DM, and clinical evidence now suggests that these agents have benefits for CVD. We review the latest advances in incretin-based pharmacotherapy. These include 'GLP1 plus' agents, which combine the known advantages of GLP1RAs with the activity of additional hormones, such as glucose-dependent insulinotropic peptide, glucagon and amylin, to achieve desired therapeutic goals. Second-generation non-peptidic oral GLP1RAs promise to extend the benefits of GLP1 therapy to those who do not want, or cannot have, subcutaneous injection therapy. We conclude with a discussion of the knowledge gaps that must be addressed before incretin-based therapies can be properly deployed for maximum benefit in the treatment of obesity and T2DM.
Collapse
Affiliation(s)
- Saleem Ansari
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Bernard Khoo
- Department of Endocrinology, Division of Medicine, Royal Free Campus, University College London, London, UK
| | - Tricia Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK.
| |
Collapse
|
44
|
Freet CS, Evans B, Brick TR, Deneke E, Wasserman EJ, Ballard SM, Stankoski DM, Kong L, Raja-Khan N, Nyland JE, Arnold AC, Krishnamurthy VB, Fernandez-Mendoza J, Cleveland HH, Scioli AD, Molchanow A, Messner AE, Ayaz H, Grigson PS, Bunce SC. Ecological momentary assessment and cue-elicited drug craving as primary endpoints: study protocol for a randomized, double-blind, placebo-controlled clinical trial testing the efficacy of a GLP-1 receptor agonist in opioid use disorder. Addict Sci Clin Pract 2024; 19:56. [PMID: 39061093 PMCID: PMC11282646 DOI: 10.1186/s13722-024-00481-7] [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: 11/28/2023] [Accepted: 06/07/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Despite continuing advancements in treatments for opioid use disorder (OUD), continued high rates of relapse indicate the need for more effective approaches, including novel pharmacological interventions. Glucagon-like peptide 1 receptor agonists (GLP-1RA) provide a promising avenue as a non-opioid medication for the treatment of OUD. Whereas GLP-1RAs have shown promise as a treatment for alcohol and nicotine use disorders, to date, no controlled clinical trials have been conducted to determine if a GLP-1RA can reduce craving in individuals with OUD. The purpose of the current protocol was to evaluate the potential for a GLP-1RA, liraglutide, to safely and effectively reduce craving in an OUD population in residential treatment. METHOD This preliminary study was a randomized, double-blinded, placebo-controlled clinical trial designed to test the safety and efficacy of the GLP-1RA, liraglutide, in 40 participants in residential treatment for OUD. Along with taking a range of safety measures, efficacy for cue-induced craving was evaluated prior to (Day 1) and following (Day 19) treatment using a Visual Analogue Scale (VAS) in response to a cue reactivity task during functional near-infrared spectroscopy (fNIRS) and for craving. Efficacy of treatment for ambient craving was assessed using Ecological Momentary Assessment (EMA) prior to (Study Day 1), across (Study Days 2-19), and following (Study Days 20-21) residential treatment. DISCUSSION This manuscript describes a protocol to collect clinical data on the safety and efficacy of a GLP-1RA, liraglutide, during residential treatment of persons with OUD, laying the groundwork for further evaluation in a larger, outpatient OUD population. Improved understanding of innovative, non-opioid based treatments for OUD will have the potential to inform community-based interventions and health policy, assist physicians and health care professionals in the treatment of persons with OUD, and to support individuals with OUD in their effort to live a healthy life. TRIAL REGISTRATION ClinicalTrials.gov: NCT04199728. Registered 16 December 2019, https://clinicaltrials.gov/study/NCT04199728?term=NCT04199728 . PROTOCOL VERSION 10 May 2023.
Collapse
Affiliation(s)
- Christopher S Freet
- Department of Psychiatry and Behavioral Health, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Brianna Evans
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Timothy R Brick
- Department of Human Development and Family Studies, The Pennsylvania State University, University Park, PA, USA
- Institute for Computational and Data Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Erin Deneke
- Fran and Doug Tieman Center for Research, Caron Treatment Centers, Wernersville, PA, USA
| | - Emily J Wasserman
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Sarah M Ballard
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Dean M Stankoski
- Fran and Doug Tieman Center for Research, Caron Treatment Centers, Wernersville, PA, USA
| | - Lan Kong
- Department of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Nazia Raja-Khan
- Department of Psychiatry and Behavioral Health, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jennifer E Nyland
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Venkatesh Basappa Krishnamurthy
- Department of Medicine and Psychiatry, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julio Fernandez-Mendoza
- Department of Psychiatry and Behavioral Health, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - H Harrington Cleveland
- Department of Human Development and Family Studies, The Pennsylvania State University, University Park, PA, USA
| | - Adam D Scioli
- Fran and Doug Tieman Center for Research, Caron Treatment Centers, Wernersville, PA, USA
| | | | | | - Hasan Ayaz
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Patricia S Grigson
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Scott C Bunce
- Department of Psychiatry and Behavioral Health, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
- Penn State University College of Medicine, Milton S. Hershey Medical Center, H073, 500 University Drive, Hershey, PA, 17033-0850, USA.
| |
Collapse
|
45
|
Li RJ, Yang T, Zeng YH, Natsuyama Y, Ren K, Li J, Nagakawa Y, Yi SQ. Impacts of different pancreatic resection ranges on endocrine function in Suncus murinus. World J Gastrointest Surg 2024; 16:2308-2318. [PMID: 39087135 PMCID: PMC11287669 DOI: 10.4240/wjgs.v16.i7.2308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Surgical intervention involving the pancreas can lead to impaired glucose tolerance and other types of endocrine dysfunction. The scope of pancreatectomy and whether it includes the ventral pancreas are the key factors in the development of postoperative diabetes. The ventral and dorsal pancreases are almost separated in Suncus murinus (S. murinus). AIM To investigate the effects of different extents of pancreatic resection on endocrine function in S. murinus. METHODS Eight-week-old male S. murinus shrews were randomly divided into three experimental groups according to different pancreatic resection ranges as follows: ventral pancreatectomy (VPx) group; partial pancreatectomy (PPx) group; subtotal pancreatectomy (SPx) group; and a sham-operated group. Postprandial serum insulin, glucagon-like peptide-1 (GLP-1), pancreatic polypeptide (PP), and somatostatin (SST) levels, as well as food intake, weight, blood glucose, and glucose tolerance were regularly measured for each animal. RESULTS S. murinus treated with PPx and SPx suffered from varying degrees of impaired glucose tolerance, but only a small proportion of the SPx group developed diabetes. Only S. murinus in the SPx group showed a significant decrease in food intake accompanied by severe weight loss, as well as a significant increase in postprandial serum GLP-1 levels. Postprandial serum PP levels decreased in both the VPx and PPx groups, but not in the SPx group. Postprandial serum SST levels decreased in both VPx and PPx groups, but the decrease was marginal. CONCLUSION Severe weight loss after pancreatectomy may be related to loss of appetite caused by compensatory elevation of GLP-1. PP and GLP-1 may play a role in resisting blood glucose imbalance.
Collapse
Affiliation(s)
- Ru-Jia Li
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo 116-8551, Japan
| | - Ting Yang
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo 116-8551, Japan
| | - Yu-Hao Zeng
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo 116-8551, Japan
| | - Yutaro Natsuyama
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo 116-8551, Japan
| | - Ke Ren
- Faculty of Physical Education, Qujing Normal University, Qujing 655000, Yunnan Province, China
| | - Jun Li
- School of Medicine, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo 160-0023, Japan
| | - Shuang-Qin Yi
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo 116-8551, Japan
| |
Collapse
|
46
|
Liu QK. Mechanisms of action and therapeutic applications of GLP-1 and dual GIP/GLP-1 receptor agonists. Front Endocrinol (Lausanne) 2024; 15:1431292. [PMID: 39114288 PMCID: PMC11304055 DOI: 10.3389/fendo.2024.1431292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are two incretins that bind to their respective receptors and activate the downstream signaling in various tissues and organs. Both GIP and GLP-1 play roles in regulating food intake by stimulating neurons in the brain's satiety center. They also stimulate insulin secretion in pancreatic β-cells, but their effects on glucagon production in pancreatic α-cells differ, with GIP having a glucagonotropic effect during hypoglycemia and GLP-1 exhibiting glucagonostatic effect during hyperglycemia. Additionally, GIP directly stimulates lipogenesis, while GLP-1 indirectly promotes lipolysis, collectively maintaining healthy adipocytes, reducing ectopic fat distribution, and increasing the production and secretion of adiponectin from adipocytes. Together, these two incretins contribute to metabolic homeostasis, preventing both hyperglycemia and hypoglycemia, mitigating dyslipidemia, and reducing the risk of cardiovascular diseases in individuals with type 2 diabetes and obesity. Several GLP-1 and dual GIP/GLP-1 receptor agonists have been developed to harness these pharmacological effects in the treatment of type 2 diabetes, with some demonstrating robust effectiveness in weight management and prevention of cardiovascular diseases. Elucidating the underlying cellular and molecular mechanisms could potentially usher in the development of new generations of incretin mimetics with enhanced efficacy and fewer adverse effects. The treatment guidelines are evolving based on clinical trial outcomes, shaping the management of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Qiyuan Keith Liu
- MedStar Medical Group, MedStar Montgomery Medical Center, Olney, MD, United States
| |
Collapse
|
47
|
Yuan J, Liu W, Jiang X, Huang Y, Zong L, Ding H, Shen X, Sun Y, Feng X, Li X, Song Y, Gu J, Wang Y, Liu H, Zheng Z. Molecular dynamics-guided optimization of BGM0504 enhances dual-target agonism for combating diabetes and obesity. Sci Rep 2024; 14:16680. [PMID: 39030216 PMCID: PMC11271627 DOI: 10.1038/s41598-024-66998-8] [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: 05/05/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
The dual activation of glucagon-like peptide-1 receptor (GLP-1R) and glucose-dependent insulinotropic polypeptide receptor (GIPR) has emerged as a promising therapeutic strategy for managing type 2 diabetes and obesity. Tirzepatide, a dual agonist peptide, has exhibited superior clinical efficacy in glycemic and weight control compared to selective GLP-1R agonists. Nevertheless, the structural basis of Tirzepatide's extended half-life, attributed to an acylation side chain on the parent peptide, raises questions regarding its partial agonistic activity. Employing molecular dynamics simulations, we explored the dynamic processes of peptide-receptor interactions. We uncovered a crucial salt bridge between parent peptide and GLP-1R/GIPR at K20, a feature not discernible in cryo-electron microscopy structures. Building upon these insights, we developed an optimization strategy based on the parent peptide which involved repositioning the acylation side chain. The results of both in vitro and in vivo experiments demonstrated that the optimized peptide has twofold to threefold increase in agonistic activity compared to Tirzepatide while maintaining its extended half-life in plasma. This led to the design of BGM0504, which proved to be more effective than its predecessor, Tirzepatide, in both laboratory and animal studies.
Collapse
Affiliation(s)
- Jiandong Yuan
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China.
| | - Wenlang Liu
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | | | - Yangqing Huang
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China
| | - Leilei Zong
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China
| | - Haifeng Ding
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China
| | - Xinyi Shen
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Yujia Sun
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Xiangyang Feng
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China
| | - Xionghao Li
- Divamics Inc., Suzhou, 215000, People's Republic of China
| | - Yunsong Song
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China
| | - Jianing Gu
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China
| | - Yuhuai Wang
- BrightGene Bio-Medical Technology Co., Ltd, Suzhou, 215000, People's Republic of China
| | - Hao Liu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, Hubei, People's Republic of China.
| | - Zheng Zheng
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
48
|
Stratina E, Stanciu C, Nastasa R, Zenovia S, Stafie R, Rotaru A, Cuciureanu T, Muzica C, Sfarti C, Girleanu I, Minea H, Petrea O, Huiban L, Chiriac S, Singeap AM, Vlad O, Cojocariu C, Trifan A. New Insights on Using Oral Semaglutide versus Dapagliflozin in Patients with Type 2 Diabetes and Metabolic Dysfunction-Associated Steatotic Liver Disease. Diagnostics (Basel) 2024; 14:1475. [PMID: 39061612 PMCID: PMC11275343 DOI: 10.3390/diagnostics14141475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIMS Increases in both the prevalence and severity of metabolic dysfunction-associated steatotic liver disease (MASLD) and obesity are closely related. Type 2 diabetes (T2DM) has been associated with metabolic dysfunction-associated steatohepatitis (MASH)-related cirrhosis and hepatocellular carcinoma. Semaglutide is a glucagon-like peptide-1 (GLP-1) receptor agonist approved for the treatment of T2DM and has an important role in weight loss. Also, it may represent a new therapeutic option for the treatment of MASH in obese diabetic patients. The main outcomes were changes from baseline in liver steatosis and fibrosis at week 24. MATERIAL AND METHODS A total of one hundred eighty-seven patients with T2DM were eligible for this prospective study; ninety-five subjects were treated with oral semaglutide, and ninety-two patients were treated with dapagliflozin as an add-on to metformin. All the subjects were evaluated using Vibration Controlled Transient Elastography (VCTE) from June to December 2022. RESULTS From our cohort, 54% of the patients were females, with a mean age of 59.92 ± 11.89 years and a mean body mass index (BMI) of 29.53 ± 5.33 kg/m2. Following a six-month medication period, we observed a substantial reduction in anthropometric measurements, including the BMI, waist circumference (WC), and waist-to-hip ratio (WtHr), in both groups. Regarding HbA1c, a notable decrease was observed in the semaglutide group (p < 0.001) when compared to the dapagliflozin group (p = 0.011). In addition, the liver stiffness measurement (LSM) according to VCTE improved significantly in the semaglutide group participants from 8.07 ± 2.90 kPa at baseline to 6.51 ± 3.09 kPa after medication (p < 0.001). CONCLUSION The superior metabolic effects of semaglutide, correlated to dapagliflozin, may contribute to a more efficient decrease in hepatic stress and injury, leading to a substantial enhancement of liver function in T2DM patients. Further investigations conducted over an ideal timeframe are necessary to confirm the evidence presented in this study.
Collapse
Affiliation(s)
- Ermina Stratina
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Carol Stanciu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Robert Nastasa
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Sebastian Zenovia
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Remus Stafie
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Adrian Rotaru
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Tudor Cuciureanu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Cristina Muzica
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Catalin Sfarti
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Irina Girleanu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Horia Minea
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Oana Petrea
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Laura Huiban
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Stefan Chiriac
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Ana-Maria Singeap
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Oana Vlad
- Unit of Diabetes, Nutrition and Metabolic Diseases, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Camelia Cojocariu
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| | - Anca Trifan
- Department of Gastroenterology, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (E.S.); (C.S.); (S.Z.); (R.S.); (A.R.); (T.C.); (C.M.); (C.S.); (I.G.); (H.M.); (O.P.); (L.H.); (S.C.); (A.-M.S.); (C.C.); (A.T.)
- “St. Spiridon” Emergency Hospital, Institute of Gastroenterology and Hepatology, 700111 Iasi, Romania
| |
Collapse
|
49
|
Astrup A. Reflections on the discovery GLP-1 as a satiety hormone: Implications for obesity therapy and future directions. Eur J Clin Nutr 2024; 78:551-556. [PMID: 38890501 PMCID: PMC11230893 DOI: 10.1038/s41430-024-01460-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Scientists were chasing an incretin hormone, and when GLP-1 was finally discovered, we found that it had a pronounced satiety effect, slowed down gastric emptying, and actually reduced postprandial insulin response. These mechanisms are the basis for the highly efficacious GLP-1 analogues that today offer safe and effective treatment in millions of people living with obesity. Moreover, the combined GLP-1 mechanisms of weight loss and delayed carbohydrate absorption may also be the key drivers of remission of type 2 diabetes and reduced cardiovascular events found by GLP-1 analogues.
Collapse
Affiliation(s)
- Arne Astrup
- Department of Obesity and Nutritional Sciences, Novo Nordisk Foundation, Hellerup, Denmark.
| |
Collapse
|
50
|
Thorens B, Hodson DJ. Building the Glucagon-Like Peptide-1 Receptor Brick by Brick: Revisiting a 1993 Diabetes Classic by Thorens et al. Diabetes 2024; 73:1027-1031. [PMID: 38900951 PMCID: PMC11189827 DOI: 10.2337/dbi24-0025] [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: 03/05/2024] [Accepted: 04/14/2024] [Indexed: 06/22/2024]
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a class B G protein-coupled receptor involved in the regulation of blood glucose levels and food intake. Stabilized agonists targeting GLP-1R are used in the treatment of type 2 diabetes and have recently become a breakthrough obesity therapy. Here, we revisit a classic article in Diabetes by Thorens et al. that described the cloning, sequencing, and functional expression of the human GLP-1R. The article also demonstrated that exendin4(1-39) was a full agonist of the human GLP-1R whereas exendin4(9-39) was a full antagonist. We discuss how the knowledge imparted by these studies has gone on to inform multiple strands of GLP-1R biology over the past three decades, including pharmacology, signaling, human genetics, structural biology, and chemical biology.
Collapse
Affiliation(s)
- Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - David J. Hodson
- Oxford Centre for Diabetes, Endocrinology, and Metabolism (OCDEM), National Institute for Health and Care Research Oxford Biomedical Research Centre, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, U.K
| |
Collapse
|