1
|
Moreno-Lopez M, Louvet I, Delalleau N, Acosta-Montalvo A, Thevenet J, Pasquetti G, Gmyr V, Kerr-Conte J, Pattou F, Bonner C, Saponaro C. The role of the glucagon-FGF21 axis in improving beta cell function during glucose intolerance and SGLT2 inhibition. Diabetes Obes Metab 2025; 27:885-898. [PMID: 39618173 DOI: 10.1111/dom.16089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/30/2024] [Accepted: 11/09/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVE Although primarily secreted by the liver, Fibroblast Growth Factor 21 (FGF21) is also expressed in the pancreas, where its function remains unclear. This study aims to elucidate the role of the glucagon-FGF21 interaction in the metabolic benefits of SGLT2 inhibition (SGLT2i) and hypothesizes it is key to enhancing glucose and lipid metabolism in individuals with glucose intolerance or type 2 diabetes (T2D). METHODS FGF21, FGF1R, and β-klotho expression in human pancreas was analysed by RNAscope, qPCR and immunofluorescent techniques. Glucose-stimulated insulin secretion (GSIS) assay was used to investigate the effects of recombinant FGF21 (rFGF21) on islets from donors with glucose intolerance or T2D. To explore the role of the glucagon-FGF21 axis in the benefits of SGLT2i, we used WT and Sglt2 knockout (KO) mice fed a chow diet (CD) or a high-fat diet (HFD) and chronically treated with vehicle or dapagliflozin. RESULTS Chronic rFGF21 treatment enhanced GSIS in islets from donors with glucose intolerance, with increased FGFR1 expression, suggesting FGF21's greater efficacy in the early stages of disease. In diet-induced insulin-resistant mice, dapagliflozin reduced postprandial glycaemia and elevated plasma glucagon and FGF21 levels. Sglt2 KO mice on a CD showed increased fasting plasma glucagon without changes in FGF21. In diet-induced insulin-resistant Sglt2 KO mice, elevated glucagon and FGF21 levels paralleled chronic dapagliflozin treatment, indicating similar metabolic adaptations in both models. CONCLUSION Our findings indicate FGF21 as a crucial mediator in liver-pancreas crosstalk, improving lipid and glucose metabolism, enhancing pancreatic function, and potentiating the therapeutic efficacy of SGLT2i, thereby representing a target for prediabetes treatment.
Collapse
Affiliation(s)
- Maria Moreno-Lopez
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Isaline Louvet
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Nathalie Delalleau
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Ana Acosta-Montalvo
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Julien Thevenet
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Gianni Pasquetti
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Valery Gmyr
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Julie Kerr-Conte
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Francois Pattou
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Caroline Bonner
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| | - Chiara Saponaro
- Univ. Lille, CHU Lille, Inserm U1190, EGID, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
2
|
Wu Y, Chan AY, Hauke J, Htin Aung O, Foollee A, Cleofe MAS, Stölting H, Han ML, Jeppe KJ, Barlow CK, Okun JG, Rusu PM, Rose AJ. Variable glucagon metabolic actions in diverse mouse models of obesity and type 2 diabetes. Mol Metab 2024; 90:102064. [PMID: 39536823 PMCID: PMC11617456 DOI: 10.1016/j.molmet.2024.102064] [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: 10/03/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The study aimed to investigate the effects of glucagon on metabolic pathways in mouse models of obesity, fatty liver disease, and type 2 diabetes (T2D) to determine the extent and variability of hepatic glucagon resistance in these conditions. METHODS We investigated glucagon's effects in mouse models of fatty liver disease, obesity, and type 2 diabetes (T2D), including male BKS-db/db, high-fat diet-fed, and western diet-fed C57Bl/6 mice. Glucagon tolerance tests were performed using the selective glucagon receptor agonist acyl-glucagon (IUB288). Blood glucose, serum and liver metabolites include lipids and amino acids were measured. Additionally, liver protein expression related to glucagon signalling and a comprehensive liver metabolomics were performed. RESULTS Western diet-fed mice displayed impaired glucagon response, with reduced blood glucose and PKA activation. In contrast, high-fat diet-fed and db/db mice maintained normal glucagon sensitivity, showing significant elevations in blood glucose and phospho-PKA motif protein expression. Acyl-glucagon treatment also lowered liver alanine and histidine levels in high-fat diet-fed mice, but not in western diet-fed mice. Additionally, some amino acids, such as methionine, were increased by acyl-glucagon only in chow diet control mice. Despite normal glucagon sensitivity in PKA signalling, db/db mice had a distinct metabolomic response, with acyl-glucagon significantly altering 90 metabolites in db/+ mice but only 42 in db/db mice, and classic glucagon-regulated metabolites, such as cyclic adenosine monophosphate (cAMP), being less responsive in db/db mice. CONCLUSIONS The study reveals that hepatic glucagon resistance in obesity and T2D is complex and not uniform across metabolic pathways, underscoring the complexity of glucagon action in these conditions.
Collapse
Affiliation(s)
- Yuqin Wu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Andrea Y Chan
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Jana Hauke
- Division of Inherited Metabolic Diseases, University Children's Hospital, 69120 Heidelberg, Germany
| | - Okka Htin Aung
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Ashish Foollee
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Maria Almira S Cleofe
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Helen Stölting
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Mei-Ling Han
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - Katherine J Jeppe
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - Jürgen G Okun
- Division of Inherited Metabolic Diseases, University Children's Hospital, 69120 Heidelberg, Germany
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia
| | - Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria 3800, Australia.
| |
Collapse
|
3
|
Valdecantos MP, Ruiz L, Folgueira C, Rada P, Gomez-Santos B, Solas M, Hitos AB, Field J, Francisco V, Escalona-Garrido C, Zagmutt S, Calderon-Dominguez M, Mera P, Garcia-Martinez I, Maymó-Masip E, Grajales D, Alen R, Mora A, Sáinz N, Vides-Urrestarazu I, Vilarrasa N, Arbones-Mainar JM, Zaragoza C, Moreno-Aliaga MJ, Aspichueta P, Fernández-Veledo S, Vendrell J, Serra D, Herrero L, Schreiber R, Zechner R, Sabio G, Hornigold D, Rondinone CM, Jermutus L, Grimsby J, Valverde ÁM. The dual GLP-1/glucagon receptor agonist G49 mimics bariatric surgery effects by inducing metabolic rewiring and inter-organ crosstalk. Nat Commun 2024; 15:10342. [PMID: 39609390 PMCID: PMC11605122 DOI: 10.1038/s41467-024-54080-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] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 11/01/2024] [Indexed: 11/30/2024] Open
Abstract
Bariatric surgery is effective for the treatment and remission of obesity and type 2 diabetes, but pharmacological approaches which exert similar metabolic adaptations are needed to avoid post-surgical complications. Here we show how G49, an oxyntomodulin (OXM) analog and dual glucagon/glucagon-like peptide-1 receptor (GCGR/GLP-1R) agonist, triggers an inter-organ crosstalk between adipose tissue, pancreas, and liver which is initiated by a rapid release of free fatty acids (FFAs) by white adipose tissue (WAT) in a GCGR-dependent manner. This interactome leads to elevations in adiponectin and fibroblast growth factor 21 (FGF21), causing WAT beiging, brown adipose tissue (BAT) activation, increased energy expenditure (EE) and weight loss. Elevation of OXM, under basal and postprandial conditions, and similar metabolic adaptations after G49 treatment were found in plasma from patients with obesity early after metabolic bariatric surgery. These results identify G49 as a potential pharmacological alternative sharing with bariatric surgery hormonal and metabolic pathways.
Collapse
Affiliation(s)
- M Pilar Valdecantos
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain.
- Faculty of Experimental Science, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain.
| | - Laura Ruiz
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Instituto de Salud Carlos III, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Rada
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Gomez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
- BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Maite Solas
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, Pamplona, Spain
- IdISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Ana B Hitos
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Joss Field
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Vera Francisco
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Escalona-Garrido
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - María Calderon-Dominguez
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Irma Garcia-Martinez
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Elsa Maymó-Masip
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Diana Grajales
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Alen
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Instituto de Salud Carlos III, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Neira Sáinz
- University of Navarra, Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, 31008, Pamplona, Spain
| | - Irene Vides-Urrestarazu
- University of Navarra, Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, 31008, Pamplona, Spain
| | - Nuria Vilarrasa
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
- Obesity Unit and Endocrinology and Nutrition Departments, Hospital Universitari de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - José M Arbones-Mainar
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Adipocyte and Fat Biology Laboratory (AdipoFat), Unidad de Investigación Traslacional, Instituto Aragonés de Ciencias de la Salud (IACS), Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Carlos Zaragoza
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERcv), Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Investigación Cardiovascular, Universidad Francisco de Vitoria/Servicio de Cardiología, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, Spain
| | - María J Moreno-Aliaga
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdISNA, Navarra Institute for Health Research, Pamplona, Spain
- University of Navarra, Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, 31008, Pamplona, Spain
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
- BioBizkaia Health Research Institute, Barakaldo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Sonia Fernández-Veledo
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Rovira I Virgili University (URV), Tarragona, Spain
| | - Joan Vendrell
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Rovira I Virgili University (URV), Tarragona, Spain
| | - Dolors Serra
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Laura Herrero
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, 8010, Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010, Graz, Austria
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Instituto de Salud Carlos III, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - David Hornigold
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Cristina M Rondinone
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca Ltd, Gaithersburg, MD, USA
- Pep2Tango Therapeutics Inc., Potomac, MD, USA
| | - Lutz Jermutus
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Joseph Grimsby
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca Ltd, Gaithersburg, MD, USA
- Regeneron Pharmaceuticals, Inc., Internal Medicine, Tarrytown, NY, USA
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERdem), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Roell W, Coskun T, Kim T, O'Farrell L, Martin JA, Nason S, Hernandez-Alamillo J, Dhantu S, Drucker DJ, Sloop KW, Steele JP, Alsina-Fernandez J, Habegger KM. Characterization of LY3324954 a long-acting glucagon-receptor agonist. Mol Metab 2024; 91:102073. [PMID: 39603505 PMCID: PMC11696851 DOI: 10.1016/j.molmet.2024.102073] [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: 09/12/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE Glucagon is a crucial regulator of glucose and lipid metabolism as well as whole-body energy balance. Thus, modulation of glucagon receptor (GCGR) activity in the context of single-molecule multi-receptor co-agonists has become an emerging therapeutic target against obesity and obesity-associated metabolic dysfunction. To better elucidate the role of GCGR-signaling when paired with incretin receptor signaling or on its own, we developed, LY3324954, a GCGR agonist with improved potency and selectivity as compared to the native glucagon peptide. METHODS LY3324954 was administered to DIO mice, rats, dogs, and monkeys to evaluate pharmacokinetic (PK) profile. Biweekly treatments were conducted in lean and DIO mice to characterize LY3324954-effects on glucose homeostasis and energy balance. Single dose studies were also conducted in liver Gcgr-deficient mice to establish receptor specificity. RESULTS LY3324954 also exhibited extended PK profile in DIO mice, rats, dogs, and monkeys. When administered every 72 h, LY3324954 treatment stimulated transient glucose and insulin excursions in lean mice. In diet-induced obese mice, LY3324954 treatment stimulates energy expenditure, weight loss, and a reduction of adiposity in a dose-dependent manner. Benefit to whole-body lipid homeostasis was likewise observed in these mice. CONCLUSIONS Taken together, these studies characterize a long-acting and potent GCGR-agonist and its regulation of glucose and lipid metabolism as well as whole-body energy balance following both acute and chronic treatment in mice.
Collapse
Affiliation(s)
| | - Tamer Coskun
- Lilly Research Laboratories, Indianapolis, IN, USA
| | - Teayoun Kim
- Comprehensive Diabetes Center and Department of Medicine - Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Shelly Nason
- Comprehensive Diabetes Center and Department of Medicine - Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jasmin Hernandez-Alamillo
- Comprehensive Diabetes Center and Department of Medicine - Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Saidharshana Dhantu
- Comprehensive Diabetes Center and Department of Medicine - Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, USA
| | - Kyle W Sloop
- Lilly Research Laboratories, Indianapolis, IN, USA
| | | | | | - Kirk M Habegger
- Comprehensive Diabetes Center and Department of Medicine - Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
5
|
Sangwung P, Ho JD, Siddall T, Lin J, Tomas A, Jones B, Sloop KW. Class B1 GPCRs: insights into multireceptor pharmacology for the treatment of metabolic disease. Am J Physiol Endocrinol Metab 2024; 327:E600-E615. [PMID: 38984948 PMCID: PMC11559640 DOI: 10.1152/ajpendo.00371.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/14/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
The secretin-like, class B1 subfamily of seven transmembrane-spanning G protein-coupled receptors (GPCRs) consists of 15 members that coordinate important physiological processes. These receptors bind peptide ligands and use a distinct mechanism of activation that is driven by evolutionarily conserved structural features. For the class B1 receptors, the C-terminus of the cognate ligand is initially recognized by the receptor via an N-terminal extracellular domain that forms a hydrophobic ligand-binding groove. This binding enables the N-terminus of the ligand to engage deep into a large volume, open transmembrane pocket of the receptor. Importantly, the phylogenetic basis of this ligand-receptor activation mechanism has provided opportunities to engineer analogs of several class B1 ligands for therapeutic use. Among the most accepted of these are drugs targeting the glucagon-like peptide-1 (GLP-1) receptor for the treatment of type 2 diabetes and obesity. Recently, multifunctional agonists possessing activity at the GLP-1 receptor and the glucose-dependent insulinotropic polypeptide (GIP) receptor, such as tirzepatide, and others that also contain glucagon receptor activity, have been developed. In this article, we review members of the class B1 GPCR family with focus on receptors for GLP-1, GIP, and glucagon, including their signal transduction and receptor trafficking characteristics. The metabolic importance of these receptors is also highlighted, along with the benefit of polypharmacologic ligands. Furthermore, key structural features and comparative analyses of high-resolution cryogenic electron microscopy structures for these receptors in active-state complexes with either native ligands or multifunctional agonists are provided, supporting the pharmacological basis of such therapeutic agents.
Collapse
Affiliation(s)
- Panjamaporn Sangwung
- Molecular Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States
| | - Joseph D Ho
- Department of Structural Biology, Lilly Biotechnology Center, San Diego, California, United States
| | - Tessa Siddall
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Jerry Lin
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Ben Jones
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Kyle W Sloop
- Diabetes, Obesity and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States
| |
Collapse
|
6
|
Wu Y, Foollee A, Chan AY, Hille S, Hauke J, Challis MP, Johnson JL, Yaron TM, Mynard V, Aung OH, Cleofe MAS, Huang C, Lim Kam Sian TCC, Rahbari M, Gallage S, Heikenwalder M, Cantley LC, Schittenhelm RB, Formosa LE, Smith GC, Okun JG, Müller OJ, Rusu PM, Rose AJ. Phosphoproteomics-directed manipulation reveals SEC22B as a hepatocellular signaling node governing metabolic actions of glucagon. Nat Commun 2024; 15:8390. [PMID: 39333498 PMCID: PMC11436942 DOI: 10.1038/s41467-024-52703-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] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
The peptide hormone glucagon is a fundamental metabolic regulator that is also being considered as a pharmacotherapeutic option for obesity and type 2 diabetes. Despite this, we know very little regarding how glucagon exerts its pleiotropic metabolic actions. Given that the liver is a chief site of action, we performed in situ time-resolved liver phosphoproteomics to reveal glucagon signaling nodes. Through pathway analysis of the thousands of phosphopeptides identified, we reveal "membrane trafficking" as a dominant signature with the vesicle trafficking protein SEC22 Homolog B (SEC22B) S137 phosphorylation being a top hit. Hepatocyte-specific loss- and gain-of-function experiments reveal that SEC22B was a key regulator of glycogen, lipid and amino acid metabolism, with SEC22B-S137 phosphorylation playing a major role in glucagon action. Mechanistically, we identify several protein binding partners of SEC22B affected by glucagon, some of which were differentially enriched with SEC22B-S137 phosphorylation. In summary, we demonstrate that phosphorylation of SEC22B is a hepatocellular signaling node mediating the metabolic actions of glucagon and provide a rich resource for future investigations on the biology of glucagon action.
Collapse
Affiliation(s)
- Yuqin Wu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Ashish Foollee
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Andrea Y Chan
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Susanne Hille
- Department of Internal Medicine V, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Jana Hauke
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Matthew P Challis
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Jared L Johnson
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Department of Cell Biology, Harvard Medical School, Boston, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Tomer M Yaron
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Victoria Mynard
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Okka H Aung
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Maria Almira S Cleofe
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Cheng Huang
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, Victoria, Australia
| | | | - Mohammad Rahbari
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Department of Surgery, Theodor-Kutzer-Ufer 1-3, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
| | - Suchira Gallage
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, Heidelberg, Germany
- University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard-Karls University, Tübingen, Germany
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, USA
- Department of Cell Biology, Harvard Medical School, Boston, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Ralf B Schittenhelm
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
- Monash Proteomics and Metabolomics Platform, Monash University, Victoria, Australia
| | - Luke E Formosa
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Greg C Smith
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Jürgen G Okun
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Oliver J Müller
- Department of Internal Medicine V, University Hospital of Schleswig-Holstein, Campus Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Patricia M Rusu
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia
| | - Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria, Australia.
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Victoria, Australia.
| |
Collapse
|
7
|
Liu J, Jiang J, Li Y, Chen Q, Yang T, Lei Y, He Z, Wang X, Na Q, Lao C, Luo X, Yang L, Yang Z. Effects of FGF21 overexpression in osteoporosis and bone mineral density: a two-sample, mediating Mendelian analysis. Front Endocrinol (Lausanne) 2024; 15:1439255. [PMID: 39296716 PMCID: PMC11409249 DOI: 10.3389/fendo.2024.1439255] [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: 05/27/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Objective Fibroblast growth factor 21 (FGF21) is a secreted protein that regulates body metabolism. In recent years, many observational studies have found that FGF21 is closely related to bone mineral density and osteoporosis, but the causal relationship between them is still unclear. Therefore, this study used two-sample, mediated Mendelian randomization (MR) analysis to explore the causal relationship between FGF21 and osteoporosis and bone mineral density. Methods We conducted a two-sample, mediator MR Analysis using genetic data from publicly available genome-wide association studies (GWAS) that included genetic variants in the inflammatory cytokine FGF21, and Total body bone mineral density, Heel bone mineral density, Forearm bone mineral density, Femoral neck bone mineral density, osteoporosis. The main analysis method used was inverse variance weighting (IVW) to investigate the causal relationship between exposure and outcome. In addition, weighted median, simple median method, weighted median method and MR-Egger regression were used to supplement the explanation, and sensitivity analysis was performed to evaluate the reliability of the results. Results MR Results showed that FGF21 overexpression reduced bone mineral density: Total body bone mineral density (OR=0.920, 95%CI: 0.876-0.966), P=0.001), Heel bone mineral density (OR=0.971, 95%CI (0.949-0.993); P=0.01), Forearm bone mineral density (OR=0.882, 95%CI(0.799-0.973); P=0.012), Femoral neck bone mineral density (OR=0.952, 95%CI(0.908-0.998), P=0.039); In addition, it also increased the risk of osteoporosis (OR=1.003, 95%CI (1.001-1.005), P=0.004). Sensitivity analysis supported the reliability of these results. The effect of FGF21 overexpression on osteoporosis may be mediated by type 2 diabetes mellitus and basal metabolic rate, with mediating effects of 14.96% and 12.21%, respectively. Conclusions Our study suggests that the overexpression of FGF21 may lead to a decrease in bone mineral density and increase the risk of osteoporosis, and the effect of FGF21 on osteoporosis may be mediated through type 2 diabetes and basal metabolic rate. This study can provide a reference for analyzing the potential mechanism of osteoporosis and is of great significance for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Jun Jiang
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Yunjia Li
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Qiaojun Chen
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Ting Yang
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Yanfa Lei
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Zewei He
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Xiaowei Wang
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Qiang Na
- Department of Spinal Surgery, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China
| | - Changtao Lao
- Department of Spinal Surgery, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China
| | - Xinlei Luo
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Lirong Yang
- Department of Oncology, Southern Central Hospital of Yunnan Province, Honghe, China
| | - Zhengchang Yang
- Department of Spinal Surgery, Southern Central Hospital of Yunnan Province, Honghe, China
| |
Collapse
|
8
|
Sidrak WR, Kalra S, Kalhan A. Approved and Emerging Hormone-Based Anti-Obesity Medications: A Review Article. Indian J Endocrinol Metab 2024; 28:445-460. [PMID: 39676791 PMCID: PMC11642516 DOI: 10.4103/ijem.ijem_442_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/22/2024] [Accepted: 03/18/2024] [Indexed: 12/17/2024] Open
Abstract
Obesity is a heterogeneous, complex, and chronic disease that has a detrimental impact on disability-adjusted life years across the globe. Recent advancements in our understanding of gut-brain communication at the molecular level have driven the development of next-generation anti-obesity medications (AOMs). Glucagon-like peptide-1 receptor agonists (GLP1RAs) remain the front-runners in this rapidly evolving landscape of hormone-based AOMs. Two GLP1RAs, namely Liraglutide and Semaglutide, have been approved by the Food and Drug Administration (FDA) and European Medicine Agency (EMA) for use in clinical practice for weight loss. Three oral GLP1RAs, namely Semaglutide, Danuglipron, and Orforglipron, are undergoing advanced clinical trials in individuals with obesity. Amylin receptor agonist (AMYRA) Cagrilintide, when used alone or in combination with Semaglutide, has demonstrated substantial weight reduction in clinical trials. Tirzepatide, a dual agonist for the glucose-dependent insulinotropic polypeptide (GIP) and GLP-1 receptors, has been observed to be associated with a significant placebo-subtracted weight reduction of 17.8% in a 72-week randomized controlled trial. Novel approaches targeting glucagon signalling have also yielded promising preliminary results. Three long-acting GLP1R/glucagon receptor (GCGR) dual agonists, namely Survodutide, Mazdutide, and Pemvidutide, exhibited significant weight loss in clinical trials. Retatrutide, a GLP1R/GCGR/GIPR tri-agonist, has been associated with a placebo-subtracted weight reduction of -22.1% in a 48-week phase-II trial. As a note of caution, long-term data on such medications' safety and cardiovascular benefits is yet to be ascertained. Our review provides a comprehensive overview of the approved and emerging hormone-based AOMs, highlighting the diversity of options that might become available in the near future.
Collapse
Affiliation(s)
- Wael R. Sidrak
- Department of Endocrinology and Diabetes, Abou-Seifein Diabetes and Endocrine Center, Cairo, Egypt
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, Haryana, India
| | - Atul Kalhan
- Department of Endocrinology and Diabetes, Royal Glamorgan Hospital, Llantrisant, UK
| |
Collapse
|
9
|
Kusminski CM, Perez-Tilve D, Müller TD, DiMarchi RD, Tschöp MH, Scherer PE. Transforming obesity: The advancement of multi-receptor drugs. Cell 2024; 187:3829-3853. [PMID: 39059360 PMCID: PMC11286204 DOI: 10.1016/j.cell.2024.06.003] [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: 11/09/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024]
Abstract
For more than a century, physicians have searched for ways to pharmacologically reduce excess body fat. The tide has finally turned with recent advances in biochemically engineered agonists for the receptor of glucagon-like peptide-1 (GLP-1) and their use in GLP-1-based polyagonists. These polyagonists reduce body weight through complementary pharmacology by incorporating the receptors for glucagon and/or the glucose-dependent insulinotropic polypeptide (GIP). In their most advanced forms, gut-hormone polyagonists achieve an unprecedented weight reduction of up to ∼20%-30%, offering a pharmacological alternative to bariatric surgery. Along with favorable effects on glycemia, fatty liver, and kidney disease, they also offer beneficial effects on the cardiovascular system and adipose tissue. These new interventions, therefore, hold great promise for the future of anti-obesity medications.
Collapse
Affiliation(s)
- Christine M Kusminski
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Munich, Germany; German Center for Diabetes Research (DZD) and Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | | | - Matthias H Tschöp
- Helmholtz Munich, Munich, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
10
|
Cui Y, Auclair H, He R, Zhang Q. GPCR-mediated regulation of beige adipocyte formation: Implications for obesity and metabolic health. Gene 2024; 915:148421. [PMID: 38561165 DOI: 10.1016/j.gene.2024.148421] [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: 01/11/2024] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Obesity and its associated complications pose a significant burden on health. The non-shivering thermogenesis (NST) and metabolic capacity properties of brown adipose tissue (BAT), which are distinct from those of white adipose tissue (WAT), in combating obesity and its related metabolic diseases has been well documented. However, beige adipose tissue, the third and relatively novel type of adipose tissue, which emerges in extensive presence of WAT and shares similar favorable metabolic properties with BAT, has garnered considerable attention in recent years. In this review, we focused on the role of G protein-coupled receptors (GPCRs), the largest receptor family and the most successful class of drug targets in humans, in the induction of beige adipocytes. More importantly, we highlight researchers' clinical treatment attempts to ameliorate obesity and other related metabolic diseases through the formation and activation of beige adipose tissue. In summary, this review provides valuable insights into the formation of beige adipose tissue and the involvement of GPCRs, based on the latest advancements in scientific research.
Collapse
Affiliation(s)
- Yuanxu Cui
- Animal Zoology Department, Kunming Medical University, Kunming, China; Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
| | - Hugo Auclair
- Faculty of Medicine, François-Rabelais University, Tours, France
| | - Rong He
- Animal Zoology Department, Kunming Medical University, Kunming, China
| | - Qiang Zhang
- Animal Zoology Department, Kunming Medical University, Kunming, China.
| |
Collapse
|
11
|
Cuboni D, Caputo M, Ghigo E, Aimaretti G, Gasco V. Once upon a time: the glucagon stimulation test in diagnosing adult GH deficiency. J Endocrinol Invest 2024; 47:1621-1631. [PMID: 38461479 PMCID: PMC11196325 DOI: 10.1007/s40618-024-02322-5] [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: 12/07/2023] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
PURPOSE The clinical features of adult GH deficiency (GHD) are nonspecific, and its diagnosis is established through GH stimulation testing, which is often complex, expensive, time-consuming and may be associated with adverse side effects. Moreover, diagnosing adult GHD can be challenging due to the influence of age, gender, and body mass index on GH peak at each test. The insulin tolerance test (ITT), GHRH + arginine test, glucagon stimulation test (GST), and, more recently, testing with macimorelin are all recognized as useful in diagnosing adult GHD. To date GST is still little used, but due to the unavailability of the GHRH all over the world and the high cost of macimorelin, in the next future it will probably become the most widely used test when ITT is contraindicated. The aim of the present review is to describe the current knowledge on GST. METHODS Narrative review. RESULTS In the last years several studies have suggested some changes in the original GST protocol and have questioned its diagnostic accuracy when the classic GH cut-point of 3 μg/L is used, suggesting to use a lower GH cut-point to improve its sensitivity and specificity in overweight/obese patients and in those with lower pretest GHD probability. CONCLUSION This document provides an update on the utility of GST, summarizes how to perform the test, shows which cut-points should be used in interpreting the results, and discusses its drawbacks and caveats referring to the most recent studies.
Collapse
Affiliation(s)
- D Cuboni
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Science, ASOU "Città della Salute e Della Scienza" di Torino, University of Turin, C.So Dogliotti 14, 10126, Turin, Italy
| | - M Caputo
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - E Ghigo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Science, ASOU "Città della Salute e Della Scienza" di Torino, University of Turin, C.So Dogliotti 14, 10126, Turin, Italy
| | - G Aimaretti
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - V Gasco
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Science, ASOU "Città della Salute e Della Scienza" di Torino, University of Turin, C.So Dogliotti 14, 10126, Turin, Italy.
| |
Collapse
|
12
|
Sanyal AJ, Kaplan LM, Frias JP, Brouwers B, Wu Q, Thomas MK, Harris C, Schloot NC, Du Y, Mather KJ, Haupt A, Hartman ML. Triple hormone receptor agonist retatrutide for metabolic dysfunction-associated steatotic liver disease: a randomized phase 2a trial. Nat Med 2024; 30:2037-2048. [PMID: 38858523 PMCID: PMC11271400 DOI: 10.1038/s41591-024-03018-2] [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: 09/14/2023] [Accepted: 04/24/2024] [Indexed: 06/12/2024]
Abstract
Retatrutide is a novel triple agonist of the glucose-dependent insulinotropic polypeptide, glucagon-like peptide 1 and glucagon receptors. A 48-week phase 2 obesity study demonstrated weight reductions of 22.8% and 24.2% with retatrutide 8 and 12 mg, respectively. The primary objective of this substudy was to assess mean relative change from baseline in liver fat (LF) at 24 weeks in participants from that study with metabolic dysfunction-associated steatotic liver disease and ≥10% of LF. Here, in this randomized, double-blind, placebo-controlled trial, participants (n = 98) were randomly assigned to 48 weeks of once-weekly subcutaneous retatrutide (1, 4, 8 or 12 mg dose) or placebo. The mean relative change from baseline in LF at 24 weeks was -42.9% (1 mg), -57.0% (4 mg), -81.4% (8 mg), -82.4% (12 mg) and +0.3% (placebo) (all P < 0.001 versus placebo). At 24 weeks, normal LF (<5%) was achieved by 27% (1 mg), 52% (4 mg), 79% (8 mg), 86% (12 mg) and 0% (placebo) of participants. LF reductions were significantly related to changes in body weight, abdominal fat and metabolic measures associated with improved insulin sensitivity and lipid metabolism. The ClinicalTrials.gov registration is NCT04881760 .
Collapse
Affiliation(s)
- Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health and Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Lee M Kaplan
- Section of Obesity Medicine and Weight and Wellness Center, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Juan P Frias
- Velocity Clinical Research, Los Angeles, CA, USA
| | | | - Qiwei Wu
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | | | - Yu Du
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Axel Haupt
- Eli Lilly and Company, Indianapolis, IN, USA
| | | |
Collapse
|
13
|
Kajani S, Laker RC, Ratkova E, Will S, Rhodes CJ. Hepatic glucagon action: beyond glucose mobilization. Physiol Rev 2024; 104:1021-1060. [PMID: 38300523 DOI: 10.1152/physrev.00028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Glucagon's ability to promote hepatic glucose production has been known for over a century, with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism [when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions] is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a "catabolic hormone." Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive "omics" technologies, has implicated glucagon as more than just a "glucose liberator." Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that "catabolic hormone."
Collapse
Affiliation(s)
- Sarina Kajani
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Rhianna C Laker
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Ekaterina Ratkova
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sarah Will
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Christopher J Rhodes
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| |
Collapse
|
14
|
Richter MM, Kemp IM, Heebøll S, Winther-Sørensen M, Kjeldsen SAS, Jensen NJ, Nybing JD, Linden FH, Høgh-Schmidt E, Boesen MP, Madsbad S, Schiødt FV, Nørgaard K, Schmidt S, Gluud LL, Haugaard SB, Holst JJ, Nielsen S, Rungby J, Wewer Albrechtsen NJ. Glucagon augments the secretion of FGF21 and GDF15 in MASLD by indirect mechanisms. Metabolism 2024; 156:155915. [PMID: 38631460 DOI: 10.1016/j.metabol.2024.155915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Glucagon receptor agonism is currently explored for the treatment of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). The metabolic effects of glucagon receptor agonism may in part be mediated by increases in circulating levels of Fibroblast Growth Factor 21 (FGF21) and Growth Differentiation Factor 15 (GDF15). The effect of glucagon agonism on FGF21 and GDF15 levels remains uncertain, especially in the context of elevated insulin levels commonly observed in metabolic diseases. METHODS We investigated the effect of a single bolus of glucagon and a continuous infusion of glucagon on plasma concentrations of FGF21 and GDF15 in conditions of endogenous low or high insulin levels. The studies included individuals with overweight with and without MASLD, healthy controls (CON) and individuals with type 1 diabetes (T1D). The direct effect of glucagon on FGF21 and GDF15 was evaluated using our in-house developed isolated perfused mouse liver model. RESULTS FGF21 and GDF15 correlated with plasma levels of insulin, but not glucagon, and their secretion was highly increased in MASLD compared with CON and T1D. Furthermore, FGF21 levels in individuals with overweight with or without MASLD did not increase after glucagon stimulation when insulin levels were kept constant. FGF21 and GDF15 levels were unaffected by direct stimulation with glucagon in the isolated perfused mouse liver. CONCLUSION The glucagon-induced secretion of FGF21 and GDF15 is augmented in MASLD and may depend on insulin. Thus, glucagon receptor agonism may augment its metabolic benefits in patients with MASLD through enhanced secretion of FGF21 and GDF15.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ida M Kemp
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sara Heebøll
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Marie Winther-Sørensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nicole J Jensen
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Janus D Nybing
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Frederik H Linden
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Erik Høgh-Schmidt
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Mikael P Boesen
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Frank Vinholt Schiødt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kirsten Nørgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Signe Schmidt
- Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Lise Lotte Gluud
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Gastro Unit, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
15
|
McGlone ER, Bloom SR, Tan TMM. Glucagon resistance and metabolic-associated steatotic liver disease: a review of the evidence. J Endocrinol 2024; 261:e230365. [PMID: 38579751 PMCID: PMC11067060 DOI: 10.1530/joe-23-0365] [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: 11/21/2023] [Accepted: 04/03/2024] [Indexed: 04/07/2024]
Abstract
Metabolic-associated steatotic liver disease (MASLD) is closely associated with obesity. MASLD affects over 1 billion adults globally but there are few treatment options available. Glucagon is a key metabolic regulator, and its actions include the reduction of liver fat through direct and indirect means. Chronic glucagon signalling deficiency is associated with hyperaminoacidaemia, hyperglucagonaemia and increased circulating levels of glucagon-like peptide 1 (GLP-1) and fibroblast growth factor 21 (FGF-21). Reduction in glucagon activity decreases hepatic amino acid and triglyceride catabolism; metabolic effects include improved glucose tolerance, increased plasma cholesterol and increased liver fat. Conversely, glucagon infusion in healthy volunteers leads to increased hepatic glucose output, decreased levels of plasma amino acids and increased urea production, decreased plasma cholesterol and increased energy expenditure. Patients with MASLD share many hormonal and metabolic characteristics with models of glucagon signalling deficiency, suggesting that they could be resistant to glucagon. Although there are few studies of the effects of glucagon infusion in patients with obesity and/or MASLD, there is some evidence that the expected effect of glucagon on amino acid catabolism may be attenuated. Taken together, this evidence supports the notion that glucagon resistance exists in patients with MASLD and may contribute to the pathogenesis of MASLD. Further studies are warranted to investigate the direct effects of glucagon on metabolism in patients with MASLD.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Stephen R Bloom
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M-M Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
16
|
Kistkins S, Moser O, Ankudovičs V, Blizņuks D, Mihailovs T, Lobanovs S, Sourij H, Pfeiffer AFH, Pīrāgs V. From classical dualistic antagonism to hormone synergy: potential of overlapping action of glucagon, insulin and GLP-1 for the treatment of diabesity. Endocr Connect 2024; 13:e230529. [PMID: 38579770 PMCID: PMC11046332 DOI: 10.1530/ec-23-0529] [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: 12/18/2023] [Accepted: 04/03/2024] [Indexed: 04/07/2024]
Abstract
The increasing prevalence of 'diabesity', a combination of type 2 diabetes and obesity, poses a significant global health challenge. Unhealthy lifestyle factors, including poor diet, sedentary behaviour, and high stress levels, combined with genetic and epigenetic factors, contribute to the diabesity epidemic. Diabesity leads to various significant complications such as cardiovascular diseases, stroke, and certain cancers. Incretin-based therapies, such as GLP-1 receptor agonists and dual hormone therapies, have shown promising results in improving glycaemic control and inducing weight loss. However, these therapies also come with certain disadvantages, including potential withdrawal effects. This review aims to provide insights into the cross-interactions of insulin, glucagon, and GLP-1, revealing the complex hormonal dynamics during fasting and postprandial states, impacting glucose homeostasis, energy expenditure, and other metabolic functions. Understanding these hormonal interactions may offer novel hypotheses in the development of 'anti-diabesity' treatment strategies. The article also explores the question of the antagonism of insulin and glucagon, providing insights into the potential synergy and hormonal overlaps between these hormones.
Collapse
Affiliation(s)
| | - Othmar Moser
- Division of Exercise Physiology and Metabolism, Institute of Sport Science, University of Bayreuth, Bayreuth, Germany
| | | | - Dmitrijs Blizņuks
- Institute of Smart Computing Technologies, Riga Technical University, Riga, Latvia
| | - Timurs Mihailovs
- Institute of Smart Computing Technologies, Riga Technical University, Riga, Latvia
| | | | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetolgoy, Medical University of Graz, Graz, Austria
| | - Andreas F H Pfeiffer
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm, Berlin, Germany
| | - Valdis Pīrāgs
- Pauls Stradiņš Clinical University Hospital, Riga, Latvia
- Faculty of Medicine, University of Latvia, Riga, Latvia
| |
Collapse
|
17
|
Liu S, Ezran C, Wang MFZ, Li Z, Awayan K, Long JZ, De Vlaminck I, Wang S, Epelbaum J, Kuo CS, Terrien J, Krasnow MA, Ferrell JE. An organism-wide atlas of hormonal signaling based on the mouse lemur single-cell transcriptome. Nat Commun 2024; 15:2188. [PMID: 38467625 PMCID: PMC10928088 DOI: 10.1038/s41467-024-46070-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/07/2024] [Indexed: 03/13/2024] Open
Abstract
Hormones mediate long-range cell communication and play vital roles in physiology, metabolism, and health. Traditionally, endocrinologists have focused on one hormone or organ system at a time. Yet, hormone signaling by its very nature connects cells of different organs and involves crosstalk of different hormones. Here, we leverage the organism-wide single cell transcriptional atlas of a non-human primate, the mouse lemur (Microcebus murinus), to systematically map source and target cells for 84 classes of hormones. This work uncovers previously-uncharacterized sites of hormone regulation, and shows that the hormonal signaling network is densely connected, decentralized, and rich in feedback loops. Evolutionary comparisons of hormonal genes and their expression patterns show that mouse lemur better models human hormonal signaling than mouse, at both the genomic and transcriptomic levels, and reveal primate-specific rewiring of hormone-producing/target cells. This work complements the scale and resolution of classical endocrine studies and sheds light on primate hormone regulation.
Collapse
Affiliation(s)
- Shixuan Liu
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Camille Ezran
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Michael F Z Wang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Zhengda Li
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kyle Awayan
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford, CA, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Sheng Wang
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Jacques Epelbaum
- Adaptive Mechanisms and Evolution (MECADEV), UMR 7179, National Center for Scientific Research, National Museum of Natural History, Brunoy, France
| | - Christin S Kuo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jérémy Terrien
- Adaptive Mechanisms and Evolution (MECADEV), UMR 7179, National Center for Scientific Research, National Museum of Natural History, Brunoy, France
| | - Mark A Krasnow
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford, CA, USA.
| | - James E Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
18
|
Allard C, Cota D, Quarta C. Poly-Agonist Pharmacotherapies for Metabolic Diseases: Hopes and New Challenges. Drugs 2024; 84:127-148. [PMID: 38127286 DOI: 10.1007/s40265-023-01982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
The use of glucagon-like peptide-1 (GLP-1) receptor-based multi-agonists in the treatment of type 2 diabetes and obesity holds great promise for improving glycaemic control and weight management. Unimolecular dual and triple agonists targeting multiple gut hormone-related pathways are currently in clinical trials, with recent evidence supporting their efficacy. However, significant knowledge gaps remain regarding the biological mechanisms and potential adverse effects associated with these multi-target agents. The mechanisms underlying the therapeutic efficacy of GLP-1 receptor-based multi-agonists remain somewhat mysterious, and hidden threats may be associated with the use of gut hormone-based polyagonists. In this review, we provide a critical analysis of the benefits and risks associated with the use of these new drugs in the management of obesity and diabetes, while also exploring new potential applications of GLP-1-based pharmacology beyond the field of metabolic disease.
Collapse
Affiliation(s)
- Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France.
| |
Collapse
|
19
|
Melander SA, Kayed A, Andreassen KV, Karsdal MA, Henriksen K. OXM-104, a potential candidate for the treatment of obesity, NASH and type 2 diabetes. Eur J Pharmacol 2024; 962:176215. [PMID: 38056618 DOI: 10.1016/j.ejphar.2023.176215] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023]
Abstract
OBJECTIVE Dual glucagon-like peptide-1 (GLP-1) and glucagon receptor agonists are therapeutic agents with an interesting liver-specific mode of action suitable for metabolic complications. In this study, dual GLP-1 and glucagon receptor agonist OXM-104 is compared head-to-head with the once-daily dual GLP-1 and glucagon receptor agonist cotadutide and GLP-1 receptor agonist semaglutide to explore the metabolic efficacy of OXM-104. METHODS The in vitro potencies of OXM-104, cotadutide and semaglutide were assessed using reporter assays. In addition, in vivo efficacy was investigated using mouse models of diet-induced obesity (DIO mice), diabetes (db/db mice) and diet-induced NASH mice (MS-NASH). RESULTS OXM-104 was found to only activate the GLP-1 and glucagon with no cross-reactivity at the (GIP) receptor. Cotadutide was also found to activate the GLP-1 and glucagon receptors, whereas semaglutide only showed activity at the GLP-1 receptor. OXM-104, cotadutide, and semaglutide elicited marked reductions in body weight and improved glucose control. In contrast, hepatoprotective effects, i.e., reductions in steatosis and fibrosis, as well as liver fibrotic biomarkers, were more prominent with OXM-104 and cotadutide than those seen with semaglutide, demonstrated by an improved NAFLD activity score (NAS) by OXM-104 and cotadutide, underlining the importance of the glucagon receptor. CONCLUSION These results show that dual GLP-1 and glucagon receptor agonism is superior to GLP-1 alone. OXM-104 was found to be a promising therapeutic candidate for the treatment of metabolic complications such as obesity, type 2 diabetes and NASH.
Collapse
Affiliation(s)
| | | | | | | | - Kim Henriksen
- Nordic Bioscience, 2730 Herlev, Denmark; KeyBioscience AG, Stans, Switzerland
| |
Collapse
|
20
|
Carbonetti MP, Almeida-Oliveira F, Majerowicz D. Use of FGF21 analogs for the treatment of metabolic disorders: a systematic review and meta-analysis. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 68:e220493. [PMID: 37948566 PMCID: PMC10916804 DOI: 10.20945/2359-4292-2022-0493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/23/2023] [Indexed: 11/12/2023]
Abstract
FGF21 is a hormone produced primarily by the liver with several metabolic functions, such as induction of heat production, control of glucose homeostasis, and regulation of blood lipid levels. Due to these actions, several laboratories have developed FGF21 analogs to treat patients with metabolic disorders such as obesity and diabetes. Here, we performed a systematic review and meta-analysis of randomized controlled trials that used FGF21 analogs and analyzed metabolic outcomes. Our search yielded 236 articles, and we included eight randomized clinical trials in the meta-analysis. The use of FGF21 analogs exhibited no effect on fasting blood glucose, glycated hemoglobin, HOMA index, blood free fatty acids or systolic blood pressure. However, the treatment significantly reduced fasting insulinemia, body weight and total cholesterolemia. None of the included studies were at high risk of bias. The quality of the evidence ranged from moderate to very low, especially due to imprecision and indirection issues. These results indicate that FGF21 analogs can potentially treat metabolic syndrome. However, more clinical trials are needed to increase the quality of evidence and confirm the effects seen thus far.
Collapse
Affiliation(s)
- Maria Paula Carbonetti
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Fernanda Almeida-Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - David Majerowicz
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
- Programa de Pós-graduação em Biociências, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brasil,
| |
Collapse
|
21
|
Ramne S, Duizer L, Nielsen MS, Jørgensen NR, Svenningsen JS, Grarup N, Sjödin A, Raben A, Gillum MP. Meal sugar-protein balance determines postprandial FGF21 response in humans. Am J Physiol Endocrinol Metab 2023; 325:E491-E499. [PMID: 37729024 PMCID: PMC10874651 DOI: 10.1152/ajpendo.00241.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Biological mechanisms to promote dietary balance remain unclear. Fibroblast growth factor 21 (FGF21) has been suggested to contribute to such potential regulation considering that FGF21 1) is genetically associated with carbohydrate/sugar and protein intake in opposite directions, 2) is secreted after sugar ingestion and protein restriction, and 3) pharmacologically reduces sugar and increases protein intake in rodents. To gain insight of the nature of this potential regulation, we aimed to study macronutrient interactions in the secretory regulation of FGF21 in healthy humans. We conducted a randomized, double-blinded, crossover meal study (NCT05061485), wherein healthy volunteers consumed a sucrose drink, a sucrose + protein drink, and a sucrose + fat drink (matched sucrose content), and compared postprandial FGF21 responses between the three macronutrient combinations. Protein suppressed the sucrose-induced FGF21 secretion [incremental area under the curve (iAUC) for sucrose 484 ± 127 vs. sucrose + protein -35 ± 49 pg/mL × h, P < 0.001]. The same could not be demonstrated for fat (iAUC 319 ± 102 pg/mL × h, P = 203 for sucrose + fat vs. sucrose). We found no indications that regulators of glycemic homeostasis could explain this effect. This indicates that FGF21 responds to disproportionate intake of sucrose relative to protein acutely within a meal, and that protein outweighs sucrose in FGF21 regulation. Together with previous findings, our results suggests that FGF21 might act to promote macronutrient balance and sufficient protein intake.NEW & NOTEWORTHY Here we test the interactions between sugar, protein, and fat in human FGF21 regulation and demonstrate that protein, but not fat, suppresses sugar-induced FGF21 secretion. This indicates that protein outweighs the effects of sugar in the secretory regulation of FGF21, and could suggest that the nutrient-specific appetite-regulatory actions of FGF21 might prioritize ensuring sufficient protein intake over limiting sugar intake.
Collapse
Affiliation(s)
- Stina Ramne
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisanne Duizer
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Mette S Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niklas Rye Jørgensen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jens S Svenningsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Sjödin
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Anne Raben
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
- Department of Clinical and Translational Research, Copenhagen University Hospital-Diabetes Center Copenhagen, Herlev, Denmark
| | - Matthew P Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Parente M, Tonini C, Segatto M, Pallottini V. Regulation of cholesterol metabolism: New players for an old physiological process. J Cell Biochem 2023; 124:1449-1465. [PMID: 37796135 DOI: 10.1002/jcb.30477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
Identified more than two centuries ago, cholesterol plays a pivotal role in human physiology. Since cholesterol metabolism is a physiologically significant process, it is not surprising that its alterations are associated with several pathologies. The discovery of new molecular targets or compounds able to modulate this sophisticated metabolism has been capturing the attention of research groups worldwide since many years. Endogenous and exogenous compounds are known to regulate cellular cholesterol synthesis and uptake, or reduce cholesterol absorption at the intestinal level, thereby regulating cholesterol homeostasis. However, there is a great need of new modulators and diverse new pathways have been uncovered. Here, after illustrating cholesterol metabolism and its well-known regulators, some new players of this important physiological process are also described.
Collapse
Affiliation(s)
| | | | - Marco Segatto
- Department of Bioscience and Territory, University of Molise, Pesche, Italy
| | - Valentina Pallottini
- Department of Science, University Roma Tre, Rome, Italy
- Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Via del Fosso Fiorano, Rome, Italy
| |
Collapse
|
23
|
Tschöp M, Nogueiras R, Ahrén B. Gut hormone-based pharmacology: novel formulations and future possibilities for metabolic disease therapy. Diabetologia 2023; 66:1796-1808. [PMID: 37209227 PMCID: PMC10474213 DOI: 10.1007/s00125-023-05929-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/27/2023] [Indexed: 05/22/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) receptor agonists are established pharmaceutical therapies for the treatment of type 2 diabetes and obesity. They mimic the action of GLP-1 to reduce glucose levels through stimulation of insulin secretion and inhibition of glucagon secretion. They also reduce body weight by inducing satiety through central actions. The GLP-1 receptor agonists used clinically are based on exendin-4 and native GLP-1 and are available as formulations for daily or weekly s.c. or oral administration. GLP-1 receptor agonism is also achieved by inhibitors of dipeptidyl peptidase-4 (DPP-4), which prevent the inactivation of GLP-1 and glucose-dependent insulinotropic polypeptide (GIP), thereby prolonging their raised levels after meal ingestion. Other developments in GLP-1 receptor agonism include the formation of small orally available agonists and compounds with the potential to pharmaceutically stimulate GLP-1 secretion from the gut. In addition, GLP-1/glucagon and GLP-1/GIP dual receptor agonists and GLP-1/GIP/glucagon triple receptor agonists have shown the potential to reduce blood glucose levels and body weight through their effects on islets and peripheral tissues, improving beta cell function and stimulating energy expenditure. This review summarises developments in gut hormone-based therapies and presents the future outlook for their use in type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum, München, Germany
| | - Ruben Nogueiras
- Department of Physiology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Bo Ahrén
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
24
|
Wewer Albrechtsen NJ, Holst JJ, Cherrington AD, Finan B, Gluud LL, Dean ED, Campbell JE, Bloom SR, Tan TMM, Knop FK, Müller TD. 100 years of glucagon and 100 more. Diabetologia 2023; 66:1378-1394. [PMID: 37367959 DOI: 10.1007/s00125-023-05947-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/18/2023] [Indexed: 06/28/2023]
Abstract
The peptide hormone glucagon, discovered in late 1922, is secreted from pancreatic alpha cells and is an essential regulator of metabolic homeostasis. This review summarises experiences since the discovery of glucagon regarding basic and clinical aspects of this hormone and speculations on the future directions for glucagon biology and glucagon-based therapies. The review was based on the international glucagon conference, entitled 'A hundred years with glucagon and a hundred more', held in Copenhagen, Denmark, in November 2022. The scientific and therapeutic focus of glucagon biology has mainly been related to its role in diabetes. In type 1 diabetes, the glucose-raising properties of glucagon have been leveraged to therapeutically restore hypoglycaemia. The hyperglucagonaemia evident in type 2 diabetes has been proposed to contribute to hyperglycaemia, raising questions regarding underlying mechanism and the importance of this in the pathogenesis of diabetes. Mimicry experiments of glucagon signalling have fuelled the development of several pharmacological compounds including glucagon receptor (GCGR) antagonists, GCGR agonists and, more recently, dual and triple receptor agonists combining glucagon and incretin hormone receptor agonism. From these studies and from earlier observations in extreme cases of either glucagon deficiency or excess secretion, the physiological role of glucagon has expanded to also involve hepatic protein and lipid metabolism. The interplay between the pancreas and the liver, known as the liver-alpha cell axis, reflects the importance of glucagon for glucose, amino acid and lipid metabolism. In individuals with diabetes and fatty liver diseases, glucagon's hepatic actions may be partly impaired resulting in elevated levels of glucagonotropic amino acids, dyslipidaemia and hyperglucagonaemia, reflecting a new, so far largely unexplored pathophysiological phenomenon termed 'glucagon resistance'. Importantly, the hyperglucagonaemia as part of glucagon resistance may result in increased hepatic glucose production and hyperglycaemia. Emerging glucagon-based therapies show a beneficial impact on weight loss and fatty liver diseases and this has sparked a renewed interest in glucagon biology to enable further pharmacological pursuits.
Collapse
Affiliation(s)
- Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Lise Lotte Gluud
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - E Danielle Dean
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Endocrinology Division, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Stephen R Bloom
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M-M Tan
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Filip K Knop
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München Neuherberg, Germany
| |
Collapse
|
25
|
Novikoff A, Müller TD. The molecular pharmacology of glucagon agonists in diabetes and obesity. Peptides 2023; 165:171003. [PMID: 36997003 PMCID: PMC10265134 DOI: 10.1016/j.peptides.2023.171003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Within recent decades glucagon receptor (GcgR) agonism has drawn attention as a therapeutic tool for the treatment of type 2 diabetes and obesity. In both mice and humans, glucagon administration enhances energy expenditure and suppresses food intake suggesting a promising metabolic utility. Therefore synthetic optimization of glucagon-based pharmacology to further resolve the physiological and cellular underpinnings mediating these effects has advanced. Chemical modifications to the glucagon sequence have allowed for greater peptide solubility, stability, circulating half-life, and understanding of the structure-function potential behind partial and "super"-agonists. The knowledge gained from such modifications has provided a basis for the development of long-acting glucagon analogues, chimeric unimolecular dual- and tri-agonists, and novel strategies for nuclear hormone targeting into glucagon receptor-expressing tissues. In this review, we summarize the developments leading toward the current advanced state of glucagon-based pharmacology, while highlighting the associated biological and therapeutic effects in the context of diabetes and obesity.
Collapse
Affiliation(s)
- Aaron Novikoff
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
26
|
Ye X, Chen Y, Qi J, Zhu S, Wu Y, Xiong J, Hu F, Guo Z, Liang X. Design and pharmaceutical evaluation of bifunctional fusion protein of FGF21 and GLP-1 in the treatment of nonalcoholic steatohepatitis. Eur J Pharmacol 2023:175811. [PMID: 37245859 DOI: 10.1016/j.ejphar.2023.175811] [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: 02/13/2023] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 05/30/2023]
Abstract
Fibroblast growth factor 21 (FGF21) and glucagon-like peptide-1 (GLP-1) may be useful for the treatment of type 2 diabetes, obesity, and non-alcoholic fatty liver disease (NAFLD). Previous studies have shown that GLP-1 may synergize with FGF21 in the regulation of glucose and lipid metabolism. Currently, no approved drug therapy is available for non-alcoholic steatohepatitis (NASH). Here, we constructed and screened dual-targeting fusion proteins of GLP-1 and FGF21, connected by elastin-like polypeptides (ELPs), to investigate whether a combination of these two hormones would have therapeutic effects in models of NASH. The temperature phase transition and release of the hormones under physiological conditions were studied to identify a bifunctional fusion protein of FGF21 and GLP-1 (GEF) that was highly stable and showed sustained release. We further evaluated the quality and therapeutic efficacy of GEF in three mouse models of NASH. We successfully synthesized a novel recombinant bifunctional fusion protein with high stability and low immunogenicity. The GEF protein synthesized ameliorated hepatic lipid accumulation, hepatocyte damage, and inflammation; prevented the progression of NASH in the three models; reduced glycemia; and caused weight loss. This novel GEF molecule may be suitable for clinical use for the treatment of NAFLD/NASH and related metabolic diseases.
Collapse
Affiliation(s)
- Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China.
| | - Yingli Chen
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Jianying Qi
- National Research Centre for Carbohydrate Synthesis, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang, 330022, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuanyuan Wu
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Jingjing Xiong
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Fei Hu
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China
| | - Zhimou Guo
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian, 116023, China.
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian, 116023, China.
| |
Collapse
|
27
|
Garbuzova Striukova EV, Shramko VS, Kashtanova EV, Polonskaya YV, Stakhneva EM, Kurguzov AV, Murashov IS, Chernyavsky AM, Ragino YI. Adipokine-Cytokine Profile in Patients with Unstable Atherosclerotic Plaques and Abdominal Obesity. Int J Mol Sci 2023; 24:ijms24108937. [PMID: 37240282 DOI: 10.3390/ijms24108937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
The goal of the research was to study the levels of adipokines and their associations with unstable atherosclerotic plaques in patients with coronary atherosclerosis and abdominal obesity (AO). METHODS The study included 145 men aged 38-79 with atherosclerosis of the coronary arteries (CA) and stable angina pectoris II-III FC who were hospitalized for coronary bypass surgery (2011-2022). The final analysis included 116 patients. Notably, 70 men had stable plaques in the CA (of which 44.3% had AO), and 46 men had unstable plaques in the CA (of which 43.5% had AO). Adipocytokine levels were determined using multiplex analysis (Human Metabolic Hormone V3 panel). RESULTS In the subgroup of patients with unstable plaques, patients with AO had a GLP-1 level that was 1.5 times higher and a lipocalin-2 level that was 2.1 times lower, respectively. GLP-1 is direct, and lipocalin-2 is inversely associated with AO in patients with unstable plaques. Among patients with AO, the level of lipocalin-2 in patients with unstable plaques was 2.2 times lower than in patients with stable plaques in the CA. The level of lipocalin-2 was inversely associated with the presence of unstable atherosclerotic plaques in the CA. CONCLUSION GLP-1 is directly associated with AO in patients with unstable atherosclerotic plaques. Lipocalin-2 is inversely associated with unstable atherosclerotic plaques in patients with AO.
Collapse
Affiliation(s)
- Evgeniia V Garbuzova Striukova
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Victoriya S Shramko
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Elena V Kashtanova
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Yana V Polonskaya
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Ekaterina M Stakhneva
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Alexey V Kurguzov
- Federal State Budgetary Institution "National Medical Research Center named after Academician E.N. Meshalkin" Ministry of Health of the Russian Federation, Rechkunovskaya Str., 15, 630055 Novosibirsk, Russia
| | - Ivan S Murashov
- Federal State Budgetary Institution "National Medical Research Center named after Academician E.N. Meshalkin" Ministry of Health of the Russian Federation, Rechkunovskaya Str., 15, 630055 Novosibirsk, Russia
| | - Alexander M Chernyavsky
- Federal State Budgetary Institution "National Medical Research Center named after Academician E.N. Meshalkin" Ministry of Health of the Russian Federation, Rechkunovskaya Str., 15, 630055 Novosibirsk, Russia
| | - Yuliya I Ragino
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| |
Collapse
|
28
|
Pokhrel S, Dilts M, Stahl Z, Boehme S, Frame G, Chiang JY, Ferrell JM. Tgr5-/- mice are protected from ethanol-induced metabolic alterations through enhanced leptin and Fgf21 signaling. Hepatol Commun 2023; 7:e0138. [PMID: 37185802 PMCID: PMC10145946 DOI: 10.1097/hc9.0000000000000138] [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: 10/20/2022] [Accepted: 02/23/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Alcohol-associated liver disease (ALD) is caused by chronic use of alcohol and ranges from hepatic steatosis to fibrosis and cirrhosis. Bile acids are physiological detergents that also regulate hepatic glucose and lipid homeostasis by binding to several receptors. One such receptor, Takeda G protein-coupled receptor 5 (TGR5), may represent a therapeutic target for ALD. Here, we used a chronic 10-day + binge ethanol-feeding model in mice to study the role of TGR5 in alcohol-induced liver injury. METHODS Female C57BL/6J wild-type mice and Tgr5-/- mice were pair-fed Lieber-DeCarli liquid diet with ethanol (5% v/v) or isocaloric control diet for 10 days followed by a gavage of 5% ethanol or isocaloric maltose control, respectively, to represent a binge-drinking episode. Tissues were harvested 9 hours following the binge, and metabolic phenotypes were characterized through examination of liver, adipose, and brain mechanistic pathways. RESULTS Tgr5-/- mice were protected from alcohol-induced accumulation of hepatic triglycerides. Interestingly, liver and serum levels of Fgf21 were significantly increased during ethanol feeding in Tgr5-/- mice, as was phosphorylation of Stat3. Parallel to Fgf21 levels, increased leptin gene expression in white adipose tissue and increased leptin receptor in liver were detected in Tgr5-/- mice fed ethanol diet. Adipocyte lipase gene expression was significantly increased in Tgr5-/- mice regardless of diet, whereas adipose browning markers were also increased in ethanol-fed Tgr5-/- mice, indicating potential for enhanced white adipose metabolism. Lastly, hypothalamic mRNA targets of leptin, involved in the regulation of food intake, were significantly increased in Tgr5-/- mice fed ethanol diet. CONCLUSIONS Tgr5-/- mice are protected from ethanol-induced liver damage and lipid accumulation. Alterations in lipid uptake and Fgf21 signaling, and enhanced metabolic activity of white adipose tissue, may mediate these effects.
Collapse
Affiliation(s)
- Sabita Pokhrel
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Matthew Dilts
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Zachary Stahl
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Shannon Boehme
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Gabrielle Frame
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - John Y.L. Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Jessica M. Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| |
Collapse
|
29
|
Hope DCD, Tan TMM. Glucagon and energy expenditure; Revisiting amino acid metabolism and implications for weight loss therapy. Peptides 2023; 162:170962. [PMID: 36736539 DOI: 10.1016/j.peptides.2023.170962] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Glucagon receptor (GCGR)-targeted multi-agonists are being developed for the treatment of obesity and metabolic disease. GCGR activity is utilised for its favourable weight loss and metabolic properties, including increased energy expenditure (EE) and hepatic lipid metabolism. GLP1R and GIPR activities are increasingly present in a multi-agonist strategy. Due to the compound effect of increased satiety, reduced food intake and increased energy expenditure, the striking weight loss effects of these multi-agonists has been demonstrated in pre-clinical models of obesity. The precise contribution and mechanism of GCGR activity to enhanced energy expenditure and weight loss in both rodents and humans is not fully understood. In this review, our understanding of glucagon-mediated EE is explored, and an amino acid-centric paradigm contributing to this phenomenon is presented. The current progress of GCGR-targeted multi-agonists in development is also highlighted with a focus on the implications of glucagon-stimulated hypoaminoacidemia.
Collapse
Affiliation(s)
- D C D Hope
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - T M-M Tan
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
30
|
Roh E, Choi KM. Hormonal Gut-Brain Signaling for the Treatment of Obesity. Int J Mol Sci 2023; 24:ijms24043384. [PMID: 36834794 PMCID: PMC9959457 DOI: 10.3390/ijms24043384] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
The brain, particularly the hypothalamus and brainstem, monitors and integrates circulating metabolic signals, including gut hormones. Gut-brain communication is also mediated by the vagus nerve, which transmits various gut-derived signals. Recent advances in our understanding of molecular gut-brain communication promote the development of next-generation anti-obesity medications that can safely achieve substantial and lasting weight loss comparable to metabolic surgery. Herein, we comprehensively review the current knowledge about the central regulation of energy homeostasis, gut hormones involved in the regulation of food intake, and clinical data on how these hormones have been applied to the development of anti-obesity drugs. Insight into and understanding of the gut-brain axis may provide new therapeutic perspectives for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Eun Roh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang 14068, Republic of Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Correspondence: or
| |
Collapse
|
31
|
Elmelund E, Galsgaard KD, Johansen CD, Trammell SA, Bomholt AB, Winther-Sørensen M, Hunt JE, Sørensen CM, Kruse T, Lau JF, Grevengoed TJ, Holst JJ, Wewer Albrechtsen NJ. Opposing effects of chronic glucagon receptor agonism and antagonism on amino acids, hepatic gene expression, and alpha cells. iScience 2022; 25:105296. [PMID: 36325048 PMCID: PMC9618771 DOI: 10.1016/j.isci.2022.105296] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/29/2022] [Accepted: 09/30/2022] [Indexed: 01/09/2023] Open
Abstract
The pancreatic hormone, glucagon, is known to regulate hepatic glucose production, but recent studies suggest that its regulation of hepatic amino metabolism is equally important. Here, we show that chronic glucagon receptor activation with a long-acting glucagon analog increases amino acid catabolism and ureagenesis and causes alpha cell hypoplasia in female mice. Conversely, chronic glucagon receptor inhibition with a glucagon receptor antibody decreases amino acid catabolism and ureagenesis and causes alpha cell hyperplasia and beta cell loss. These effects were associated with the transcriptional regulation of hepatic genes related to amino acid uptake and catabolism and by the non-transcriptional modulation of the rate-limiting ureagenesis enzyme, carbamoyl phosphate synthetase-1. Our results support the importance of glucagon receptor signaling for amino acid homeostasis and pancreatic islet integrity in mice and provide knowledge regarding the long-term consequences of chronic glucagon receptor agonism and antagonism. Glucagon receptor agonism increases amino acid catabolism and hepatic CPS-1 activity Glucagon receptor signaling regulates the number of pancreatic alpha cells Glucagon regulates the hepatic transcription of genes involved in amino acid metabolism
Collapse
Affiliation(s)
- Emilie Elmelund
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Katrine D. Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christian D. Johansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Samuel A.J. Trammell
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Anna B. Bomholt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Marie Winther-Sørensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jenna E. Hunt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Charlotte M. Sørensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thomas Kruse
- Novo Nordisk A/S, Research Chemistry, Novo Nordisk Park, 2760 Måløv, Denmark
| | - Jesper F. Lau
- Novo Nordisk A/S, Research Chemistry, Novo Nordisk Park, 2760 Måløv, Denmark
| | - Trisha J. Grevengoed
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens J. Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nicolai J. Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg & Frederiksberg Hospitals, University of Copenhagen, 2400 Bispebjerg, Denmark
- Corresponding author
| |
Collapse
|
32
|
Regulation of feeding and therapeutic application of bioactive peptides. Pharmacol Ther 2022; 239:108187. [DOI: 10.1016/j.pharmthera.2022.108187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
33
|
Kim T, Nason S, Antipenko J, Finan B, Shalev A, DiMarchi R, Habegger KM. Hepatic mTORC2 Signaling Facilitates Acute Glucagon Receptor Enhancement of Insulin-Stimulated Glucose Homeostasis in Mice. Diabetes 2022; 71:2123-2135. [PMID: 35877180 PMCID: PMC9501720 DOI: 10.2337/db21-1018] [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: 11/11/2021] [Accepted: 07/21/2022] [Indexed: 11/13/2022]
Abstract
Long-term glucagon receptor (GCGR) agonism is associated with hyperglycemia and glucose intolerance, while acute GCGR agonism enhances whole-body insulin sensitivity and hepatic AKTSer473 phosphorylation. These divergent effects establish a critical gap in knowledge surrounding GCGR action. mTOR complex 2 (mTORC2) is composed of seven proteins, including RICTOR, which dictates substrate binding and allows for targeting of AKTSer473. We used a liver-specific Rictor knockout mouse (RictorΔLiver) to investigate whether mTORC2 is necessary for insulin receptor (INSR) and GCGR cross talk. RictorΔLiver mice were characterized by impaired AKT signaling and glucose intolerance. Intriguingly, RictorΔLiver mice were also resistant to GCGR-stimulated hyperglycemia. Consistent with our prior report, GCGR agonism increased glucose infusion rate and suppressed hepatic glucose production during hyperinsulinemic-euglycemic clamp of control animals. However, these benefits to insulin sensitivity were ablated in RictorΔLiver mice. We observed diminished AKTSer473 and GSK3α/βSer21/9 phosphorylation in RictorΔLiver mice, whereas phosphorylation of AKTThr308 was unaltered in livers from clamped mice. These signaling effects were replicated in primary hepatocytes isolated from RictorΔLiver and littermate control mice, confirming cell-autonomous cross talk between GCGR and INSR pathways. In summary, our study reveals the necessity of RICTOR, and thus mTORC2, in GCGR-mediated enhancement of liver and whole-body insulin action.
Collapse
Affiliation(s)
- Teayoun Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Shelly Nason
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Jessica Antipenko
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN
| | - Anath Shalev
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Kirk M. Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
34
|
Frampton J, Izzi-Engbeaya C, Salem V, Murphy KG, Tan TM, Chambers ES. The acute effect of glucagon on components of energy balance and glucose homoeostasis in adults without diabetes: a systematic review and meta-analysis. Int J Obes (Lond) 2022; 46:1948-1959. [PMID: 36123404 PMCID: PMC9584822 DOI: 10.1038/s41366-022-01223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 12/05/2022]
Abstract
Objective Using a systematic review and meta-analysis, we aimed to estimate the mean effect of acute glucagon administration on components of energy balance and glucose homoeostasis in adults without diabetes. Methods CENTRAL, CINAHL, Embase, MEDLINE, PubMed, and Scopus databases were searched from inception to May 2021. To be included, papers had to be a randomised, crossover, single- or double-blind study, measuring ad libitum meal energy intake, energy expenditure, subjective appetite, glucose, and/or insulin following acute administration of glucagon and an appropriate comparator in adults without diabetes. Risk of bias was assessed using the Revised Cochrane Risk of Bias Tool for Randomized trials with additional considerations for cross-over trials. Certainty of evidence was assessed using the GRADE approach. Random-effect meta-analyses were performed for outcomes with at least five studies. This study is registered on PROSPERO (CRD42021269623). Results In total, 13 papers (15 studies) were considered eligible: energy intake (5 studies, 77 participants); energy expenditure (5 studies, 59 participants); subjective appetite (3 studies, 39 participants); glucose (13 studies, 159 participants); insulin (12 studies, 147 participants). All studies had some concerns with regards to risk of bias. Mean intervention effect of acute glucagon administration on energy intake was small (standardised mean difference [SMD]: –0.19; 95% CI, –0.59 to 0.21; P = 0.345). Mean intervention effect of acute glucagon administration on energy expenditure (SMD: 0.72; 95% CI, 0.37–1.08; P < 0.001), glucose (SMD: 1.11; 95% CI, 0.60–1.62; P < 0.001), and insulin (SMD: 1.33; 95% CI, 0.88–1.77; P < 0.001) was moderate to large. Conclusions Acute glucagon administration produces substantial increases in energy expenditure, and in circulating insulin and glucose concentrations. However, the effect of acute glucagon administration on energy intake is unclear. Insufficient evidence was available to evaluate the acute effect of glucagon on subjective appetite.
Collapse
Affiliation(s)
- James Frampton
- Section for Nutrition Research, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W12 0NN, UK. .,Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
| | - Chioma Izzi-Engbeaya
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Victoria Salem
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London, SW7 2BX, UK
| | - Kevin G Murphy
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Tricia M Tan
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Edward S Chambers
- Section for Nutrition Research, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
35
|
Habegger KM. Cross Talk Between Insulin and Glucagon Receptor Signaling in the Hepatocyte. Diabetes 2022; 71:1842-1851. [PMID: 35657690 PMCID: PMC9450567 DOI: 10.2337/dbi22-0002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022]
Abstract
While the consumption of external energy (i.e., feeding) is essential to life, this action induces a temporary disturbance of homeostasis in an animal. A primary example of this effect is found in the regulation of glycemia. In the fasted state, stored energy is released to maintain physiological glycemic levels. Liver glycogen is liberated to glucose, glycerol and (glucogenic) amino acids are used to build new glucose molecules (i.e., gluconeogenesis), and fatty acids are oxidized to fuel long-term energetic demands. This regulation is driven primarily by the counterregulatory hormones epinephrine, growth hormone, cortisol, and glucagon. Conversely, feeding induces a rapid influx of diverse nutrients, including glucose, that disrupt homeostasis. Consistently, a host of hormonal and neural systems under the coordination of insulin are engaged in the transition from fasting to prandial states to reduce this disruption. The ultimate action of these systems is to appropriately store the newly acquired energy and to return to the homeostatic norm. Thus, at first glance it is tempting to assume that glucagon is solely antagonistic regarding the anabolic effects of insulin. We have been intrigued by the role of glucagon in the prandial transition and have attempted to delineate its role as beneficial or inhibitory to glycemic control. The following review highlights this long-known yet poorly understood hormone.
Collapse
Affiliation(s)
- Kirk M. Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
36
|
Huang C, Palani A, Yang Z, Deng Q, Reddy V, Nargund RP, Lin S, Altezza S, Bianchi E, Orvieto F, Carrington P. Discovery of Insulin/GLP-1/Glucagon Triagonists for the Treatment of Diabetes and Obesity. ACS Med Chem Lett 2022; 13:1255-1261. [PMID: 35978702 PMCID: PMC9377023 DOI: 10.1021/acsmedchemlett.2c00218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/15/2022] [Indexed: 11/29/2022] Open
Abstract
The combination of insulin and incretin-based therapies has emerged as a potential promising tactic for the treatment of diabetes. Here we report the first example of a unimolecular triagonist to simultaneously target insulin, GLP-1, and glucagon receptors, aiming for better glycemic control and superior weight loss. The strategy for constructing such a unimolecular triagonist is the conjugation of the insulin moiety and GLP-1R/GCGR coagonist peptide via alkyne-azide click chemistry. Two tractable series differentiated by insulin conjugation sites, B1F and B29K, were identified. Triagonist 13 prepared through the conjugation at insulin B1F and position 24 of GLP-1R/GCGR coagonist exhibited insulin activity comparable to that of insulin degludec and potent and balanced GLP-1R and GCGR activities. Pharmacokinetic profiles of 13 in both rat and minipig were also discussed.
Collapse
Affiliation(s)
- Chunhui Huang
- Merck
& Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Anandan Palani
- Merck
& Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Zhiqiang Yang
- Merck
& Co., Inc., 2015
Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qiaolin Deng
- Merck
& Co., Inc., 2015
Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Vijay Reddy
- Merck
& Co., Inc., 2015
Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ravi P. Nargund
- Merck
& Co., Inc., 2015
Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Songnian Lin
- Merck
& Co., Inc., 2015
Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Simona Altezza
- Peptide
Chemistry Unit, Peptides and Small Molecules R&D, IRBM S.p.A., Via Pontina km 30600, 00071 Pomezia (RM), Italy
| | - Elisabetta Bianchi
- Peptide
Chemistry Unit, Peptides and Small Molecules R&D, IRBM S.p.A., Via Pontina km 30600, 00071 Pomezia (RM), Italy
| | - Federica Orvieto
- Peptide
Chemistry Unit, Peptides and Small Molecules R&D, IRBM S.p.A., Via Pontina km 30600, 00071 Pomezia (RM), Italy
| | - Paul Carrington
- Merck
& Co., Inc., 2015
Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
37
|
Ali MM, Hafez A, Abdelgalil MS, Hasan MT, El-Ghannam MM, Ghogar OM, Elrashedy AA, Abd-ElGawad M. Impact of Cotadutide drug on patients with type 2 diabetes mellitus: a systematic review and meta-analysis. BMC Endocr Disord 2022; 22:113. [PMID: 35488292 PMCID: PMC9055739 DOI: 10.1186/s12902-022-01031-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/06/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The food and drug administration approved many drugs to treat diabetes mellitus, but those drugs do not have a noticeable effect on weight management. Recently, glucagon-like peptide 1 agonist known as Cotadutide serve as a potent drug in treating type 2 diabetes by reducing blood glucose levels and body weight indices. This study aimed to explore the safety and efficacy of Cotadutide as a treatment for type 2 diabetes individuals. METHODS A comprehensive literature search was done on different databases, including PubMed, Scopus, Web of Science, and Cochrane Library to capture all relevant articles using an established search strategy. The inclusion criteria were randomized controlled trials that assessed the safety and efficacy of Cotadutide versus placebo or any anti-diabetes drugs in patients with type 2 diabetes mellitus and a BMI between 22 kg/m2 and 40 kg/m2. We conducted the analysis using Revman software version 5.4. RESULTS We found 663 relevant articles. From which nine studies were included and subjected to qualitative analysis and eight for quantitative analysis. The pooled effect showed that Cotadutide was better than placebo in reducing body weight (kg) (Mean difference (MD) = 3.31, p < 0.00001), glycated hemoglobin (HbA1c) (MD = 0.68, p > 0.00001), glucose area under the plasma concentration curve (AUC [0-4 h]) (MD = 30.15, p < 0.00001), and fasting plasma glucose over time (mg/dl) (MD = 31.31, p < 0.00001). CONCLUSION Cotadutide is safe and effective in reducing plasma glucose levels, HbA1c and body weight in individuals with type 2 diabetes. TRIAL REGISTRATION The study protocol was registered on PROSPERO (CRD: CRD42021257670 ).
Collapse
Affiliation(s)
| | - Ahmed Hafez
- Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | | | | | | | | | | | | |
Collapse
|
38
|
Zhao S, Yan Z, Du Y, Li Z, Tang C, Jing L, Sun L, Yang Q, Tang X, Yuan Y, Han J, Jiang N. A GLP-1/glucagon/CCK-2 receptors tri-agonist provides new therapy for obesity and diabetes. Br J Pharmacol 2022; 179:4360-4377. [PMID: 35484823 DOI: 10.1111/bph.15860] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 03/22/2022] [Accepted: 04/05/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Glucagon-like peptide-1 receptor (GLP-1R) and glucagon receptor (GCGR) dual agonists have exerted promising therapeutic effects for the treatment of obesity and diabetes in clinical development. Moreover, GLP-1R and cholecystokinin 2 receptor (CCK-2R) dual agonists have been shown to restore pancreas function and improve glycemic control in many preclinical studies. In the present study, we describe for the first time that the beneficial effects of GLP-1R/GCGR and GLP-1R/CCK-2R dual agonists can be integrated into one peptide, resulting in significant anti-diabetes and anti-obesity effectiveness. EXPERIMENTAL APPROACH The in vitro potency of a novel GLP-1R/GCGR/CCK-2R tri-agonist (xGLP/GCG/gastrin) against GLP-1R, GCGR, CCK-1R and CCK-2R was determined on cells expressing the corresponding receptors by cAMP accumulation or ERK1/2 phosphorylation assays. The in vivo anti-diabetes and anti-obesity effects of xGLP/GCG/gastrin were studied in both db/db and diet induced obesity (DIO) mice. KEY RESULTS xGLP/GCG/gastrin was a potent and selective GLP-1R, GCGR, and CCK-2R tri-agonist. In DIO mice, the metabolic benefits of xGLP-1/GCG/gastrin such as reduction of body weight and hepatic lipid contents were significantly better than those of ZP3022 (GLP-1R/CCK-2R dual agonist) and liraglutide. In the short term study in db/db mice, xGLP/GCG/gastrin treatment exerted considerable effects on increasing islet numbers, islet areas, and insulin content. In the long-term treatment study in db/db mice, xGLP-1/GCG/gastrin displayed a significantly sustained improvement in glucose tolerance and glucose control compared with those of liraglutide, ZP3022, cotadutide (GLP-1R/GCGR dual agonist), and xGLP/GCG-15. CONCLUSIONS AND IMPLICATIONS These results demonstrate the therapeutic promise of xGLP-1/GCG/gastrin for obesity and diabetes.
Collapse
Affiliation(s)
- Songfeng Zhao
- Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhiming Yan
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Yue Du
- Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zeyun Li
- Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Chunli Tang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Lin Jing
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| | - Lidan Sun
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, People's Republic of China
| | - Qimeng Yang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, People's Republic of China
| | - Xueling Tang
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, People's Republic of China
| | - Yongliang Yuan
- Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jing Han
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, People's Republic of China
| | - Neng Jiang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
39
|
Intermittent protein restriction protects islet β cells and improves glucose homeostasis in diabetic mice. Sci Bull (Beijing) 2022; 67:733-747. [PMID: 36546138 DOI: 10.1016/j.scib.2021.12.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/15/2021] [Accepted: 12/09/2021] [Indexed: 01/06/2023]
Abstract
Diabetes is caused by the interplay between genetics and environmental factors, tightly linked to lifestyle and dietary patterns. In this study, we explored the effectiveness of intermittent protein restriction (IPR) in diabetes control. IPR drastically reduced hyperglycemia in both streptozotocin-treated and leptin receptor-deficient db/db mouse models. IPR improved the number, proliferation, and function of β cells in pancreatic islets. IPR reduced glucose production in the liver and elevated insulin signaling in the skeletal muscle. IPR elevated serum level of FGF21, and deletion of the Fgf21 gene in the liver abrogated the hypoglycemic effect of IPR without affecting β cells. IPR caused less lipid accumulation and damage in the liver than that caused by continuous protein restriction in streptozotocin-treated mice. Single-cell RNA sequencing using mouse islets revealed that IPR reversed diabetes-associated β cell reduction and immune cell accumulation. As IPR is not based on calorie restriction and is highly effective in glycemic control and β cell protection, it has promising translational potential in the future.
Collapse
|
40
|
Population Pharmacokinetics of Cotadutide in Subjects with Type 2 Diabetes. Clin Pharmacokinet 2022; 61:833-845. [PMID: 35235191 DOI: 10.1007/s40262-021-01094-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND OBJECTIVES Cotadutide is a balanced dual glucagon-like peptide-1/glucagon receptor agonist under development for the treatment of nonalcoholic steatohepatitis and chronic kidney disease with type 2 diabetes. The objectives of the analysis were to characterize the population pharmacokinetics of cotadutide following daily subcutaneous injection in subjects with type 2 diabetes and to evaluate the effect of demographic and clinical variables of interest on cotadutide pharmacokinetics. METHODS This study analyzed 8834 plasma concentrations of cotadutide from 759 subjects with type 2 diabetes who received daily subcutaneous doses from 20 to 600 μg from six clinical studies. The impact of covariates on cotadutide pharmacokinetics was quantified, and body weight effect on cotadutide exposure was further evaluated using a simulation approach. The model performance was evaluated through prediction-corrected visual predictive checks. RESULTS A one-compartment model with first-order absorption and elimination described cotadutide pharmacokinetic data well. The mean values for cotadutide apparent clearance, apparent distribution volume, absorption rate constant, and half-life were 1.04 L/h (interindividual variability [IIV]: 26.5%), 18.7 L (IIV: 28.7%), 0.343 h-1 (IIV: 38.6%), and 12.9 h, respectively. Higher body weight, lower albumin, and higher alanine aminotransferase were associated with an increase in cotadutide clearance, while an increase in anti-drug antibody titers was associated with a decrease in cotadutide clearance. These statistically significant effects were not considered clinically significant and did not warrant dose adjustment. Effects of other tested baseline covariates (age, sex, body mass index, hemoglobin A1c, renal function, duration of diabetes) were not found to statistically significantly affect cotadutide pharmacokinetics. CONCLUSIONS Cotadutide pharmacokinetics was adequately described by a one-compartment linear model with first-order absorption and elimination. Body weight-based dosing is not necessary for cotadutide based on the simulation using the final population pharmacokinetic modeling. This model will be used to evaluate exposure-response relationships for efficacy and safety in different indications that are being studied for cotadutide.
Collapse
|
41
|
High Protein Diet Feeding Aggravates Hyperaminoacidemia in Mice Deficient in Proglucagon-Derived Peptides. Nutrients 2022; 14:nu14050975. [PMID: 35267952 PMCID: PMC8912298 DOI: 10.3390/nu14050975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Protein stimulates the secretion of glucagon (GCG), which can affect glucose metabolism. This study aimed to analyze the metabolic effect of a high-protein diet (HPD) in the presence or absence of proglucagon-derived peptides, including GCG and GLP-1. (2) Methods: The response to HPD feeding for 7 days was analyzed in mice deficient in proglucagon-derived peptides (GCGKO). (3) Results: In both control and GCGKO mice, food intake and body weight decreased with HPD and intestinal expression of Pepck increased. HPD also decreased plasma FGF21 levels, regardless of the presence of proglucagon-derived peptides. In control mice, HPD increased the hepatic expression of enzymes involved in amino acid metabolism without the elevation of plasma amino acid levels, except branched-chain amino acids. On the other hand, HPD-induced changes in the hepatic gene expression were attenuated in GCGKO mice, resulting in marked hyperaminoacidemia with lower blood glucose levels; the plasma concentration of glutamine exceeded that of glucose in HPD-fed GCGKO mice. (4) Conclusions: Increased plasma amino acid levels are a common feature in animal models with blocked GCG activity, and our results underscore that GCG plays essential roles in the homeostasis of amino acid metabolism in response to altered protein intake.
Collapse
|
42
|
Fournier C, Karagounis LG, Sacco SM, Horcajada MN, Decaens T, Offord EA, Bouzakri K, Ammann P. Impact of moderate dietary protein restriction on glucose homeostasis in a model of oestrogen deficiency. J Nutr Biochem 2022; 102:108952. [PMID: 35122999 DOI: 10.1016/j.jnutbio.2022.108952] [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: 12/01/2020] [Revised: 09/27/2021] [Accepted: 01/04/2022] [Indexed: 11/26/2022]
Abstract
The need to consume adequate dietary protein to preserve physical function during ageing is well recognized. However, the effect of protein intakes on glucose metabolism is still intensively debated. During age-related oestrogen withdrawal at the time of the menopause, it is known that glucose homeostasis may be impaired but the influence of dietary protein levels in this context is unknown. The aim of the present study is to elucidate the individual and interactive effects of oestrogen deficiency and suboptimal protein intake on glucose homeostasis in a preclinical model involving ovariectomy (OVX) and a 13-week period of a moderately reduced protein intake in 7-month-old ageing rats. To investigate mechanisms of action acting via the pancreas-liver-muscle axis, fasting circulating levels of insulin, glucagon, IGF-1, FGF21 and glycemia were measured. The hepatic lipid infiltration and the protein expression of GLUT4 in the gastrocnemius were analyzed. The gene expression of some hepatokines, myokines and lipid storage/oxidation related transcription factors were quantified in the liver and the gastrocnemius. We show that, regardless of the oestrogen status, moderate dietary protein restriction increases fasting glycaemia without modifying insulinemia, body weight gain and composition. This fasting hyperglycaemia is associated with oestrogen status-specific metabolic alterations in the muscle and liver. In oestrogen-replete (SHAM) rats, GLUT4 was down-regulated in skeletal muscle while in oestrogen-deficient (OVX) rats, hepatic stress-associated hyperglucagonaemia and high serum FGF21 were observed. These findings highlight the importance of meeting dietary protein needs to avoid disturbances in glucose homeostasis in ageing female rats with or without oestrogen withdrawal.
Collapse
Affiliation(s)
- Carole Fournier
- Service of Bone Diseases, Department of Rehabilitation and Geriatrics, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Institute for Advanced Biosciences, Research Center UGA/Inserm U 1209/CNRS 5309, La Tronche, France.
| | - Leonidas G Karagounis
- Nestlé Health Science, Translation Research, Epalinges, Switzerland; Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Sandra M Sacco
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., 1015 Lausanne, Switzerland
| | - Marie-Noelle Horcajada
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., 1015 Lausanne, Switzerland
| | - Thomas Decaens
- Institute for Advanced Biosciences, Research Center UGA/Inserm U 1209/CNRS 5309, La Tronche, France; Université Grenoble Alpes, 38000 Grenoble, France; Service d'hépato-gastroentérologie, Pôle Digidune, CHU Grenoble Alpes, 38700 La Tronche, France
| | - Elizabeth A Offord
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., 1015 Lausanne, Switzerland
| | - Karim Bouzakri
- Department of Genetic Medicine and Development, University of Geneva Medical Center, Geneva, Switzerland; UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Patrick Ammann
- Service of Bone Diseases, Department of Rehabilitation and Geriatrics, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
43
|
Kepple JD, Barra JM, Young ME, Hunter CS, Tse HM. Islet transplantation into brown adipose tissue can delay immune rejection. JCI Insight 2022; 7:152800. [PMID: 35015736 PMCID: PMC8876467 DOI: 10.1172/jci.insight.152800] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease characterized by insulin-producing β cell destruction. Although islet transplantation restores euglycemia and improves patient outcomes, an ideal transplant site remains elusive. Brown adipose tissue (BAT) has a highly vascularized and antiinflammatory microenvironment. Because these tissue features can promote islet graft survival, we hypothesized that islets transplanted into BAT will maintain islet graft and BAT function while delaying immune-mediated rejection. We transplanted syngeneic and allogeneic islets into BAT or under the kidney capsule of streptozotocin-induced diabetic NOD.Rag and NOD mice to investigate islet graft function, BAT function, metabolism, and immune-mediated rejection. Islet grafts within BAT restored euglycemia similarly to kidney capsule controls. Islets transplanted in BAT maintained expression of islet hormones and transcription factors and were vascularized. Compared with those in kidney capsule and euglycemic mock-surgery controls, no differences in glucose or insulin tolerance, thermogenic regulation, or energy expenditure were observed with islet grafts in BAT. Immune profiling of BAT revealed enriched antiinflammatory macrophages and T cells. Compared with the kidney capsule control, there were significant delays in autoimmune and allograft rejection of islets transplanted in BAT, possibly due to increased antiinflammatory immune populations. Our data support BAT as an alternative islet transplant site that may improve graft survival.
Collapse
Affiliation(s)
- Jessica D Kepple
- Department of Medicine, University of Alabama at Birmingham, Birmingham, United States of America
| | - Jessie M Barra
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, United States of America
| | - Martin E Young
- Department of Medicine, University of Alabama at Birmingham, Birmingham, United States of America
| | - Chad S Hunter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, United States of America
| | - Hubert M Tse
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, United States of America
| |
Collapse
|
44
|
Miedzybrodzka EL, Gribble FM, Reimann F. Targeting the Enteroendocrine System for Treatment of Obesity. Handb Exp Pharmacol 2022; 274:487-513. [PMID: 35419620 DOI: 10.1007/164_2022_583] [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: 11/27/2022]
Abstract
Mimetics of the anorexigenic gut hormone glucagon-like peptide 1 (GLP-1) were originally developed as insulinotropic anti-diabetic drugs but also evoke significant weight loss, leading to their recent approval as obesity therapeutics. Co-activation of receptors for GLP-1 and other gut hormones which reduce food intake - peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP) - is now being explored clinically to enhance efficacy. An alternative approach involves pharmacologically stimulating endogenous secretion of these hormones from enteroendocrine cells (EECs) to recapitulate the metabolic consequences of bariatric surgery, where highly elevated postprandial levels of GLP-1 and PYY3-36 are thought to contribute to improved glycaemia and weight loss.
Collapse
Affiliation(s)
- Emily L Miedzybrodzka
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
45
|
Conceição-Furber E, Coskun T, Sloop KW, Samms RJ. Is Glucagon Receptor Activation the Thermogenic Solution for Treating Obesity? Front Endocrinol (Lausanne) 2022; 13:868037. [PMID: 35547006 PMCID: PMC9081793 DOI: 10.3389/fendo.2022.868037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 12/19/2022] Open
Abstract
A major challenge of obesity therapy is to sustain clinically relevant weight loss over time. Achieving this goal likely requires both reducing daily caloric intake and increasing caloric expenditure. Over the past decade, advances in pharmaceutical engineering of ligands targeting G protein-coupled receptors have led to the development of highly effective anorectic agents. These include mono-agonists of the GLP-1R and dual GIPR/GLP-1R co-agonists that have demonstrated substantial weight loss in experimental models and in humans. By contrast, currently, there are no medicines available that effectively augment metabolic rate to promote weight loss. Here, we present evidence indicating that activation of the GCGR may provide a solution to this unmet therapeutic need. In adult humans, GCGR agonism increases energy expenditure to a magnitude sufficient for inducing a negative energy balance. In preclinical studies, the glucagon-GCGR system affects key metabolically relevant organs (including the liver and white and brown adipose tissue) to boost whole-body thermogenic capacity and protect from obesity. Further, activation of the GCGR has been shown to augment both the magnitude and duration of weight loss that is achieved by either selective GLP-1R or dual GIPR/GLP-1R agonism in rodents. Based on the accumulation of such findings, we propose that the thermogenic activity of GCGR agonism will also complement other anti-obesity agents that lower body weight by suppressing appetite.
Collapse
|
46
|
Akkar I, Karaca Z, Taheri S, Unluhizarci K, Hacioglu A, Kelestimur F. The stimulatory effects of glucagon on cortisol and GH secretion occur independently from FGF-21. Endocrine 2022; 75:211-218. [PMID: 34562190 DOI: 10.1007/s12020-021-02829-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Glucagon stimulation test (GST) is used to assess the hypothalamo-pituitary-adrenal (HPA) and growth hormone (GH) axes with an incompletely defined mechanism. We aimed to assess if glucagon acted through fibroblast growth factor-21 (FGF-21) to stimulate cortisol and GH secretion. The secondary outcome was to determine the relationship of FGF-21 with variable GH responses to GST in obesity. METHODS A total of 26 healthy participants; 11 obese (body mass index (BMI) > 30 kg/m2) and 15 leans (BMI < 25 kg/m2) were included. Basal pituitary and target hormone levels were measured and GST was performed. During GST, glucose, insulin, cortisol, GH, and FGF-21 responses were measured. RESULTS The mean age of the participants was 26.3±3.6 years. Glucagon resulted in significant increases in FGF-21, glucose, insulin, cortisol, and GH levels. The levels of basal cortisol, GH, FGF-21, and IGF-1 were similar in the two groups. The peak GH and area under the curve (AUC)(GH) responses to GST in the obese group were lower than those of the normal-weight group with a different pattern of response. There were no differences between the groups in terms of peak cortisol, AUC(cortisol), peak insulin, AUC(insulin), peak FGF-21, and AUC(FGF21). Obesity was associated with significantly increased glucose and insulin responses and slightly decreased FGF-21 response to glucagon. CONCLUSION Obesity was associated with blunted and delayed GH, but preserved cortisol responses to GST. This is the first study showing that glucagon stimulates the HPA and GH axis independently from FGF-21. The delayed GH response to GST in obesity does not seem to be related to FGF-21.
Collapse
Affiliation(s)
- Ilyas Akkar
- Erciyes University Medical School Department of Endocrinology, Kayseri, Turkey
| | - Zuleyha Karaca
- Erciyes University Medical School Department of Endocrinology, Kayseri, Turkey.
| | - Serpil Taheri
- Erciyes University Medical School Department of Medical Biology, Kayseri, Turkey
| | - Kursad Unluhizarci
- Erciyes University Medical School Department of Endocrinology, Kayseri, Turkey
| | - Aysa Hacioglu
- Erciyes University Medical School Department of Endocrinology, Kayseri, Turkey
| | - Fahrettin Kelestimur
- Yeditepe University Medical School Department of Endocrinology, Istanbul, Turkey
| |
Collapse
|
47
|
Rhyu J, Yu R. Newly discovered endocrine functions of the liver. World J Hepatol 2021; 13:1611-1628. [PMID: 34904032 PMCID: PMC8637678 DOI: 10.4254/wjh.v13.i11.1611] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/05/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
The liver, the largest solid visceral organ of the body, has numerous endocrine functions, such as direct hormone and hepatokine production, hormone metabolism, synthesis of binding proteins, and processing and redistribution of metabolic fuels. In the last 10 years, many new endocrine functions of the liver have been discovered. Advances in the classical endocrine functions include delineation of mechanisms of liver production of endocrine hormones [including 25-hydroxyvitamin D, insulin-like growth factor 1 (IGF-1), and angiotensinogen], hepatic metabolism of hormones (including thyroid hormones, glucagon-like peptide-1, and steroid hormones), and actions of specific binding proteins to glucocorticoids, sex steroids, and thyroid hormones. These studies have furthered insight into cirrhosis-associated endocrinopathies, such as hypogonadism, osteoporosis, IGF-1 deficiency, vitamin D deficiency, alterations in glucose and lipid homeostasis, and controversially relative adrenal insufficiency. Several novel endocrine functions of the liver have also been unraveled, elucidating the liver’s key negative feedback regulatory role in the pancreatic α cell-liver axis, which regulates pancreatic α cell mass, glucagon secretion, and circulating amino acid levels. Betatrophin and other hepatokines, such as fetuin-A and fibroblast growth factor 21, have also been discovered to play important endocrine roles in modulating insulin sensitivity, lipid metabolism, and body weight. It is expected that more endocrine functions of the liver will be revealed in the near future.
Collapse
Affiliation(s)
- Jane Rhyu
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, United States
| | - Run Yu
- Division of Endocrinology, Diabetes, and Metabolism, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, United States
| |
Collapse
|
48
|
Hinds CE, Owen BM, Hope DCD, Pickford P, Jones B, Tan TM, Minnion JS, Bloom SR. A glucagon analogue decreases body weight in mice via signalling in the liver. Sci Rep 2021; 11:22577. [PMID: 34799628 PMCID: PMC8604983 DOI: 10.1038/s41598-021-01912-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/26/2021] [Indexed: 12/30/2022] Open
Abstract
Glucagon receptor agonists show promise as components of next generation metabolic syndrome pharmacotherapies. However, the biology of glucagon action is complex, controversial, and likely context dependent. As such, a better understanding of chronic glucagon receptor (GCGR) agonism is essential to identify and mitigate potential clinical side-effects. Herein we present a novel, long-acting glucagon analogue (GCG104) with high receptor-specificity and potent in vivo action. It has allowed us to make two important observations about the biology of sustained GCGR agonism. First, it causes weight loss in mice by direct receptor signalling at the level of the liver. Second, subtle changes in GCG104-sensitivity, possibly due to interindividual variation, may be sufficient to alter its effects on metabolic parameters. Together, these findings confirm the liver as a principal target for glucagon-mediated weight loss and provide new insights into the biology of glucagon analogues.
Collapse
Affiliation(s)
- Charlotte E Hinds
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Bryn M Owen
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK
| | - David C D Hope
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Philip Pickford
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Ben Jones
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Tricia M Tan
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK
| | - James S Minnion
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Stephen R Bloom
- Section of Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
49
|
Bethea M, Bozadjieva-Kramer N, Sandoval DA. Preproglucagon Products and Their Respective Roles Regulating Insulin Secretion. Endocrinology 2021; 162:6329397. [PMID: 34318874 PMCID: PMC8375443 DOI: 10.1210/endocr/bqab150] [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: 05/24/2021] [Indexed: 11/19/2022]
Abstract
Historically, intracellular function and metabolic adaptation within the α-cell has been understudied, with most of the attention being placed on the insulin-producing β-cells due to their role in the pathophysiology of type 2 diabetes mellitus. However, there is a growing interest in understanding the function of other endocrine cell types within the islet and their paracrine role in regulating insulin secretion. For example, there is greater appreciation for α-cell products and their contributions to overall glucose homeostasis. Several recent studies have addressed a paracrine role for α-cell-derived glucagon-like peptide-1 (GLP-1) in regulating glucose homeostasis and responses to metabolic stress. Further, other studies have demonstrated the ability of glucagon to impact insulin secretion by acting through the GLP-1 receptor. These studies challenge the central dogma surrounding α-cell biology describing glucagon's primary role in glucose counterregulation to one where glucagon is critical in regulating both hyper- and hypoglycemic responses. Herein, this review will update the current understanding of the role of glucagon and α-cell-derived GLP-1, placing emphasis on their roles in regulating glucose homeostasis, insulin secretion, and β-cell mass.
Collapse
Affiliation(s)
- Maigen Bethea
- Department of Pediatrics, Nutrition Section, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Darleen A Sandoval
- Department of Pediatrics, Nutrition Section, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Correspondence: Darleen A. Sandoval, PhD, University of Colorado Anschut, Division of Endocrinology, Metabolism, and Diabetes,12801 E 17th Ave. Research Complex 1 South 7th Floor, Aurora, CO 80045, USA. E-mail:
| |
Collapse
|
50
|
Hope DCD, Vincent ML, Tan TMM. Striking the Balance: GLP-1/Glucagon Co-Agonism as a Treatment Strategy for Obesity. Front Endocrinol (Lausanne) 2021; 12:735019. [PMID: 34566894 PMCID: PMC8457634 DOI: 10.3389/fendo.2021.735019] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/18/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity and Type 2 diabetes represent global health challenges, and there is an unmet need for long-lasting and effective pharmacotherapies. Although long-acting glucagon-like peptide-1 (GLP-1) analogues are now in routine use for diabetes and are now being utilised for obesity per se, the need for ever better treatments has driven the development of co-agonists, with the theoretical advantages of improved efficacy by targeting multiple pathways and reduced adverse effects. In this review, we highlight the past and present progress in our understanding and development of treatments based on GLP-1/glucagon co-agonism. We also reflect on the divergent effects of varying the GLP-1:glucagon activity and ratio in the context of pre-clinical and human clinical trial findings. In particular, the multiple metabolic actions of glucagon highlight the importance of understanding the contributions of individual hormone action to inform the safe, effective and tailored use of GLP-1/glucagon co-agonists to target weight loss and metabolic disease in the future.
Collapse
Affiliation(s)
| | | | - Tricia M. M. Tan
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|