1
|
Mott J, Celly C, Glock R, Gilor C. The glucagon-receptor antagonist MK-3577 reduces glucagon-stimulated plasma glucose and insulin concentrations in metabolically healthy overweight cats. Domest Anim Endocrinol 2024; 89:106874. [PMID: 39018655 DOI: 10.1016/j.domaniend.2024.106874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
The role of glucagon disturbances in diabetes mellitus is increasingly recognized and, hence, glucagon antagonism might aid in treatment of hyperglycemia and other metabolic disturbances. The aim of this study was to assess the pharmacokinetics of the glucagon receptor antagonist MK-3577 and its effect on plasma glucose, insulin, and glucagon concentrations in healthy cats. In a cross-over placebo-controlled study, 5 purpose-bred cats were treated with either Placebo, MK-3577 (1 mg/kg), or MK-3577 (3 mg/kg). Glucose, insulin and glucagon concentrations were measured at 0, 15, 225, 240 min post-treatment administration. Glucagon (20 mcg/kg, IM) was administered at 240 min and glucose and insulin were measured at 255, 265, 275, 285 and 300 min. Plasma MK-3577 concentrations peaked at 4.2 and 3.2 hours after 1 and 3 mg/kg dosing with a half-life of 14.8h and 15.5h respectively. Baseline glucose, insulin and glucagon concentrations did not differ significantly between treatment groups. At a dose of 3 mg/kg, MK-3577 blunted the glucagon-stimulated rise of glucose (p=0.0089) and insulin (p=0.02). Similar trends were observed with MK-3577 at the 1 mg/kg dose but the effect was smaller, and not significant. In conclusion, the GRA MK-3577 has a pharmacokinetic profile suitable for diminishing the glucagon-induced rise of glucose and insulin in healthy cats.
Collapse
Affiliation(s)
- J Mott
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 SW 16th Ave. Gainesville, FL 32608, USA
| | - C Celly
- Merck Animal Health, 126 East Lincoln Avenue, PO Box 2000, Rahway, NJ 07065, USA
| | - R Glock
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 601 Vernon Tharp St., Columbus, OH 43210, USA
| | - C Gilor
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 601 Vernon Tharp St., Columbus, OH 43210, USA; Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 SW 16th Ave. Gainesville, FL 32608, USA.
| |
Collapse
|
2
|
Winther JB, Holst JJ. Glucagon agonism in the treatment of metabolic diseases including type 2 diabetes mellitus and obesity. Diabetes Obes Metab 2024; 26:3501-3512. [PMID: 38853300 DOI: 10.1111/dom.15693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/18/2024] [Accepted: 05/18/2024] [Indexed: 06/11/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with obesity and, therefore, it is important to target both overweight and hyperglycaemia. Glucagon plays important roles in glucose, amino acid and fat metabolism and may also regulate appetite and energy expenditure. These physiological properties are currently being exploited therapeutically in several compounds, most often in combination with glucagon-like peptide-1 (GLP-1) agonism in the form of dual agonists. With this combination, increases in hepatic glucose production and hyperglycaemia, which would be counterproductive, are largely avoided. In multiple randomized trials, the co-agonists have been demonstrated to lead to significant weight loss and, in participants with T2DM, even improved glycated haemoglobin (HbA1c) levels. In addition, significant reductions in hepatic fat content have been observed. Here, we review and discuss the studies so far available. Twenty-six randomized trials of seven different GLP-1 receptor (GLP-1R)/glucagon receptor (GCGR) co-agonists were identified and reviewed. GLP-1R/GCGR co-agonists generally provided significant weight loss, reductions in hepatic fat content, improved lipid profiles, insulin secretion and sensitivity, and in some cases, improved HbA1c levels. A higher incidence of adverse effects was present with GLP-1R/GCGR co-agonist treatment than with GLP-1 agonist monotherapy or placebo. Possible additional risks associated with glucagon agonism are also discussed. A delicate balance between GLP-1 and glucagon agonism seems to be of particular importance. Further studies exploring the optimal ratio of GLP-1 and glucagon receptor activation and dosage and titration regimens are needed to ensure a sufficient safety profile while providing clinical benefits.
Collapse
Affiliation(s)
- Jonathan Brix Winther
- Department of Biomedical Sciences and the NovoNordisk Foundation Centre for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences and the NovoNordisk Foundation Centre for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
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
|
4
|
Lafferty R, Tanday N, Dubey V, Coulter-Parkhill A, Vishal K, Moffett RC, O'Harte F, Flatt PR, Irwin N. The glucagon receptor antagonist desHis 1Pro 4Glu 9-glucagon(Lys 12PAL) alters alpha-cell turnover and lineage in mice, but does not cause alpha-cell hyperplasia. Mol Cell Endocrinol 2023; 570:111932. [PMID: 37080378 DOI: 10.1016/j.mce.2023.111932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/24/2023] [Accepted: 04/16/2023] [Indexed: 04/22/2023]
Abstract
OBJECTIVE Glucagon receptor (GCGR) antagonism elicits antihyperglycemic effects in rodents and humans. The present study investigates whether the well characterised peptide-based GCGR antagonist, desHis1Pro4Glu9-glucagon (Lys12PAL), alters alpha-cell turnover or identity in mice. METHODS Multiple low-dose streptozotocin (STZ) treated (50 mg/kg bw, 5 days) transgenic GluCreERT2;ROSA26-eYFP mice were employed. STZ mice received twice daily administration of saline vehicle or desHis1Pro4Glu9-glucagon (Lys12PAL), at low- or high-dose (25 and 100 nmol/kg, respectively) for 11 days. RESULTS No GCGR antagonist induced changes in food or fluid intake, body weight or glucose homeostasis were observed. As expected, STZ dramatically reduced (P < 0.001) islet numbers and increased (P < 0.01) alpha-to beta-cell ratio, which was linked to elevated (P < 0.05) levels of beta-cell apoptosis. Whilst treatment with desHis1Pro4Glu9-glucagon (Lys12PAL) decreased (P < 0.05-P < 0.001) alpha- and beta-cell areas, it also helped restore the classic rodent islet alpha-cell mantle in STZ mice. Interestingly, low-dose desHis1Pro4Glu9-glucagon (Lys12PAL) increased (P < 0.05) alpha-cell apoptosis rates whilst high dose decreased (p < 0.05) this parameter. This difference reflects substantially increased (P < 0.001) alpha-to beta-cell transdifferentiation following high dose desHis1Pro4Glu9-glucagon (Lys12PAL) treatment, which was not fully manifest with low-dose therapy. CONCLUSIONS Taken together, the present study indicates that peptidic GCGR antagonists can positively influence alpha-cell turnover and lineage in identity in multiple low-dose STZ mice, but that such effects are dose-related.
Collapse
Affiliation(s)
- Ryan Lafferty
- Centre for Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Neil Tanday
- Centre for Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Vaibhav Dubey
- Centre for Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | | | - Karthick Vishal
- Centre for Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | | | - Finbarr O'Harte
- Centre for Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Centre for Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - Nigel Irwin
- Centre for Diabetes, Ulster University, Coleraine, Northern Ireland, UK.
| |
Collapse
|
5
|
Pixner T, Stummer N, Schneider AM, Lukas A, Gramlinger K, Julian V, Thivel D, Mörwald K, Mangge H, Dalus C, Aigner E, Furthner D, Weghuber D, Maruszczak K. The relationship between glucose and the liver-alpha cell axis - A systematic review. Front Endocrinol (Lausanne) 2023; 13:1061682. [PMID: 36686477 PMCID: PMC9849557 DOI: 10.3389/fendo.2022.1061682] [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: 10/04/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Until recently, glucagon was considered a mere antagonist to insulin, protecting the body from hypoglycemia. This notion changed with the discovery of the liver-alpha cell axis (LACA) as a feedback loop. The LACA describes how glucagon secretion and pancreatic alpha cell proliferation are stimulated by circulating amino acids. Glucagon in turn leads to an upregulation of amino acid metabolism and ureagenesis in the liver. Several increasingly common diseases (e.g., non-alcoholic fatty liver disease, type 2 diabetes, obesity) disrupt this feedback loop. It is important for clinicians and researchers alike to understand the liver-alpha cell axis and the metabolic sequelae of these diseases. While most of previous studies have focused on fasting concentrations of glucagon and amino acids, there is limited knowledge of their dynamics after glucose administration. The authors of this systematic review applied PRISMA guidelines and conducted PubMed searches to provide results of 8078 articles (screened and if relevant, studied in full). This systematic review aims to provide better insight into the LACA and its mediators (amino acids and glucagon), focusing on the relationship between glucose and the LACA in adult and pediatric subjects.
Collapse
Affiliation(s)
- Thomas Pixner
- Department of Pediatric and Adolescent Medicine, Salzkammergutklinikum Voecklabruck, Voecklabruck, Austria
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
| | - Nathalie Stummer
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Anna Maria Schneider
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Andreas Lukas
- Department of Pediatric and Adolescent Medicine, Salzkammergutklinikum Voecklabruck, Voecklabruck, Austria
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
| | - Karin Gramlinger
- Department of Pediatric and Adolescent Medicine, Salzkammergutklinikum Voecklabruck, Voecklabruck, Austria
| | - Valérie Julian
- Department of Sport Medicine and Functional Explorations, Diet and Musculoskeletal Health Team, Human Nutrition Research Center (CRNH), INRA, University Hospital of Clermont-Ferrand, University of Clermont Auvergne, Clermont-Ferrand, France
| | - David Thivel
- Laboratory of Metabolic Adaptations to Exercise under Physiological and Pathological Conditions (AME2P), University of Clermont Auvergne, Clermont-Ferrand, France
| | - Katharina Mörwald
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Christopher Dalus
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Elmar Aigner
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
- First Department of Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Dieter Furthner
- Department of Pediatric and Adolescent Medicine, Salzkammergutklinikum Voecklabruck, Voecklabruck, Austria
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
| | - Daniel Weghuber
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Maruszczak
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
6
|
Bossart M, Wagner M, Elvert R, Evers A, Hübschle T, Kloeckener T, Lorenz K, Moessinger C, Eriksson O, Velikyan I, Pierrou S, Johansson L, Dietert G, Dietz-Baum Y, Kissner T, Nowotny I, Einig C, Jan C, Rharbaoui F, Gassenhuber J, Prochnow HP, Agueusop I, Porksen N, Smith WB, Nitsche A, Konkar A. Effects on weight loss and glycemic control with SAR441255, a potent unimolecular peptide GLP-1/GIP/GCG receptor triagonist. Cell Metab 2022; 34:59-74.e10. [PMID: 34932984 DOI: 10.1016/j.cmet.2021.12.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/13/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022]
Abstract
Unimolecular triple incretins, combining the activity of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon (GCG), have demonstrated reduction in body weight and improved glucose control in rodent models. We developed SAR441255, a synthetic peptide agonist of the GLP-1, GCG, and GIP receptors, structurally based on the exendin-4 sequence. SAR441255 displays high potency with balanced activation of all three target receptors. In animal models, metabolic outcomes were superior to results with a dual GLP-1/GCG receptor agonist. Preclinical in vivo positron emission tomography imaging demonstrated SAR441255 binding to GLP-1 and GCG receptors. In healthy subjects, SAR441255 improved glycemic control during a mixed-meal tolerance test and impacted biomarkers for GCG and GIP receptor activation. Single doses of SAR441255 were well tolerated. The results demonstrate that integrating GIP activity into dual GLP-1 and GCG receptor agonism provides improved effects on weight loss and glycemic control while buffering the diabetogenic risk of chronic GCG receptor agonism.
Collapse
Affiliation(s)
- Martin Bossart
- Synthetic Medicinal Modalities, Integrated Drug Discovery Germany, Sanofi, Frankfurt, Germany.
| | - Michael Wagner
- Synthetic Medicinal Modalities, Integrated Drug Discovery Germany, Sanofi, Frankfurt, Germany
| | | | - Andreas Evers
- Synthetic Medicinal Modalities, Integrated Drug Discovery Germany, Sanofi, Frankfurt, Germany
| | | | | | - Katrin Lorenz
- Synthetic Medicinal Modalities, Integrated Drug Discovery Germany, Sanofi, Frankfurt, Germany
| | | | - Olof Eriksson
- Antaros Medical AB, Mölndal, Sweden; Science For Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Irina Velikyan
- Science For Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden; PET Centre, Centre for Medical Imaging, Uppsala University Hospital, Uppsala, Sweden
| | | | | | | | | | | | - Irene Nowotny
- Translational Medicine & Early Development, Sanofi, Frankfurt, Germany
| | | | - Christelle Jan
- Clinical Sciences & Operations, Sanofi, Chilly-Mazarin, France
| | - Faiza Rharbaoui
- Translational Medicine & Early Development, Sanofi, Frankfurt, Germany
| | | | | | | | | | - William B Smith
- NOCCR Alliance for Multispecialty Research (AMR), Knoxville, TN, USA
| | | | | |
Collapse
|
7
|
Liu Q, Lin G, Chen Y, Feng W, Xu Y, Lyu J, Yang D, Wang MW. Deleterious mutation V369M in the mouse GCGR gene causes abnormal plasma amino acid levels indicative of a possible liver-α-cell axis. Biosci Rep 2021; 41:BSR20210758. [PMID: 34002801 PMCID: PMC8173527 DOI: 10.1042/bsr20210758] [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] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/30/2023] Open
Abstract
Glucagon plays an important role in glucose homeostasis and amino acid metabolism. It regulates plasma amino acid levels which in turn modulate glucagon secretion from the pancreatic α-cell, thereby establishing a liver-α-cell axis described recently. We reported previously that the knock-in mice bearing homozygous V369M substitution (equivalent to a naturally occurring mutation V368M in the human glucagon receptor, GCGR) led to hypoglycemia with improved glucose tolerance. They also exhibited hyperglucagonemia, pancreas enlargement and α-cell hyperplasia. Here, we investigated the effect of V369M/V368M mutation on glucagon-mediated amino acid metabolism. It was found that GcgrV369M+/+ mice displayed increased plasma amino acid levels in general, but significant accumulation of the ketogenic/glucogenic amino acids was observed in animals fed with a high-fat diet (HFD), resulting in deleterious metabolic consequence characteristic of α-cell proliferation and hyperglucagonemia.
Collapse
Affiliation(s)
- Qiaofeng Liu
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Guangyao Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yan Chen
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wenbo Feng
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yingna Xu
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianjun Lyu
- Department of Pathology, InnoStar BioTech Nantong Co., Ltd., Nantong 226133, China
| | - Dehua Yang
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
| | - Ming-Wei Wang
- School of Pharmacy, Fudan University, Shanghai 201203, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai 201203, China
- School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
8
|
Lafferty RA, O’Harte FPM, Irwin N, Gault VA, Flatt PR. Proglucagon-Derived Peptides as Therapeutics. Front Endocrinol (Lausanne) 2021; 12:689678. [PMID: 34093449 PMCID: PMC8171296 DOI: 10.3389/fendo.2021.689678] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Initially discovered as an impurity in insulin preparations, our understanding of the hyperglycaemic hormone glucagon has evolved markedly over subsequent decades. With description of the precursor proglucagon, we now appreciate that glucagon was just the first proglucagon-derived peptide (PGDP) to be characterised. Other bioactive members of the PGDP family include glucagon-like peptides -1 and -2 (GLP-1 and GLP-2), oxyntomodulin (OXM), glicentin and glicentin-related pancreatic peptide (GRPP), with these being produced via tissue-specific processing of proglucagon by the prohormone convertase (PC) enzymes, PC1/3 and PC2. PGDP peptides exert unique physiological effects that influence metabolism and energy regulation, which has witnessed several of them exploited in the form of long-acting, enzymatically resistant analogues for treatment of various pathologies. As such, intramuscular glucagon is well established in rescue of hypoglycaemia, while GLP-2 analogues are indicated in the management of short bowel syndrome. Furthermore, since approval of the first GLP-1 mimetic for the management of Type 2 diabetes mellitus (T2DM) in 2005, GLP-1 therapeutics have become a mainstay of T2DM management due to multifaceted and sustainable improvements in glycaemia, appetite control and weight loss. More recently, longer-acting PGDP therapeutics have been developed, while newfound benefits on cardioprotection, bone health, renal and liver function and cognition have been uncovered. In the present article, we discuss the physiology of PGDP peptides and their therapeutic applications, with a focus on successful design of analogues including dual and triple PGDP receptor agonists currently in clinical development.
Collapse
Affiliation(s)
| | | | | | - Victor A. Gault
- School of Biomedical Sciences, Ulster University, Coleraine, United Kingdom
| | | |
Collapse
|
9
|
Zeigerer A, Sekar R, Kleinert M, Nason S, Habegger KM, Müller TD. Glucagon's Metabolic Action in Health and Disease. Compr Physiol 2021; 11:1759-1783. [PMID: 33792899 PMCID: PMC8513137 DOI: 10.1002/cphy.c200013] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Discovered almost simultaneously with insulin, glucagon is a pleiotropic hormone with metabolic action that goes far beyond its classical role to increase blood glucose. Albeit best known for its ability to directly act on the liver to increase de novo glucose production and to inhibit glycogen breakdown, glucagon lowers body weight by decreasing food intake and by increasing metabolic rate. Glucagon further promotes lipolysis and lipid oxidation and has positive chronotropic and inotropic effects in the heart. Interestingly, recent decades have witnessed a remarkable renaissance of glucagon's biology with the acknowledgment that glucagon has pharmacological value beyond its classical use as rescue medication to treat severe hypoglycemia. In this article, we summarize the multifaceted nature of glucagon with a special focus on its hepatic action and discuss the pharmacological potential of either agonizing or antagonizing the glucagon receptor for health and disease. © 2021 American Physiological Society. Compr Physiol 11:1759-1783, 2021.
Collapse
Affiliation(s)
- Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Revathi Sekar
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Maximilian Kleinert
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Shelly Nason
- Comprehensive Diabetes Center, Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirk M. Habegger
- Comprehensive Diabetes Center, Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Timo D. Müller
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| |
Collapse
|
10
|
Galsgaard KD. The Vicious Circle of Hepatic Glucagon Resistance in Non-Alcoholic Fatty Liver Disease. J Clin Med 2020; 9:jcm9124049. [PMID: 33333850 PMCID: PMC7765287 DOI: 10.3390/jcm9124049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023] Open
Abstract
A key criterion for the most common chronic liver disease—non-alcoholic fatty liver disease (NAFLD)—is an intrahepatic fat content above 5% in individuals who are not using steatogenic agents or having significant alcohol intake. Subjects with NAFLD have increased plasma concentrations of glucagon, and emerging evidence indicates that subjects with NAFLD may show hepatic glucagon resistance. For many years, glucagon has been thought of as the counterregulatory hormone to insulin with a primary function of increasing blood glucose concentrations and protecting against hypoglycemia. However, in recent years, glucagon has re-emerged as an important regulator of other metabolic processes including lipid and amino acid/protein metabolism. This review discusses the evidence that in NAFLD, hepatic glucagon resistance may result in a dysregulated lipid and amino acid/protein metabolism, leading to excess accumulation of fat, hyperglucagonemia, and increased oxidative stress contributing to the worsening/progression of NAFLD.
Collapse
Affiliation(s)
- Katrine D. Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; ; Tel.: +45-6044-6145
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
11
|
Li W, Kirchner T, Ho G, Bonilla F, D'Aquino K, Littrell J, Zhang R, Jian W, Qiu X, Zheng S, Gao B, Wong P, Leonard JN, Camacho RC. Amino acids are sensitive glucagon receptor-specific biomarkers for glucagon-like peptide-1 receptor/glucagon receptor dual agonists. Diabetes Obes Metab 2020; 22:2437-2450. [PMID: 33463043 DOI: 10.1111/dom.14173] [Citation(s) in RCA: 12] [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: 05/20/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/31/2022]
Abstract
AIM The aim of this study was to evaluate amino acids as glucagon receptor (GCGR)-specific biomarkers in rodents and cynomolgus monkeys in the presence of agonism of both glucagon-like peptide-1 receptor (GLP1R) and GCGR with a variety of dual agonist compounds. MATERIALS AND METHODS Primary hepatocytes, rodents (normal, diet-induced obese and GLP1R knockout) and cynomolgus monkeys were treated with insulin (hepatocytes only), glucagon (hepatocytes and cynomolgus monkeys), the GLP1R agonist, dulaglutide, or a variety of dual agonists with varying GCGR potencies. RESULTS A long-acting dual agonist, Compound 2, significantly decreased amino acids in both wild-type and GLP1R knockout mice in the absence of changes in food intake, body weight, glucose or insulin, and increased expression of hepatic amino acid transporters. Dulaglutide, or a variant of Compound 2 lacking GCGR agonism, had no effect on amino acids. A third variant with ~31-fold less GCGR potency than Compound 2 significantly decreased amino acids, albeit to a significantly lesser extent than Compound 2. Dulaglutide (with saline infusion) had no effect on amino acids, but an infusion of glucagon dose-dependently decreased amino acids on the background of GLP1R engagement (dulaglutide) in cynomolgus monkeys, as did Compound 2. CONCLUSIONS These results show that amino acids are sensitive and translatable GCGR-specific biomarkers.
Collapse
Affiliation(s)
- Wenyu Li
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - Thomas Kirchner
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - George Ho
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - Fany Bonilla
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - Katharine D'Aquino
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - James Littrell
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - Rui Zhang
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - Wenying Jian
- Pharmacokinetics, Dynamics, and Metabolism, Janssen R&D, Spring House, Pennsylvania, USA
| | - Xi Qiu
- Pharmacokinetics, Dynamics, and Metabolism, Janssen R&D, Spring House, Pennsylvania, USA
| | - Songmao Zheng
- Janssen Biotherapeutics, Janssen R&D, Spring House, Pennsylvania, USA
| | - Bin Gao
- Translational Medicine and Early Development Statistics, Janssen R&D, Spring House, Pennsylvania, USA
| | - Peggy Wong
- Quantitative Sciences, Janssen R&D, Raritan, New Jersey, USA
| | - James N Leonard
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| | - Raul C Camacho
- Cardiovascular Metabolism Discovery, Janssen R&D, Spring House, Pennsylvania, USA
| |
Collapse
|
12
|
Dutta K, Das R, Ling J, Monibas RM, Carballo-Jane E, Kekec A, Feng DD, Lin S, Mu J, Saklatvala R, Thayumanavan S, Liang Y. In Situ Forming Injectable Thermoresponsive Hydrogels for Controlled Delivery of Biomacromolecules. ACS OMEGA 2020; 5:17531-17542. [PMID: 32715238 PMCID: PMC7379096 DOI: 10.1021/acsomega.0c02009] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/25/2020] [Indexed: 05/23/2023]
Abstract
Due to their relatively large molecular sizes and delicate nature, biologic drugs such as peptides, proteins, and antibodies often require high and repeated dosing, which can cause undesired side effects and physical discomfort in patients and render many therapies inordinately expensive. To enhance the efficacy of biologic drugs, they could be encapsulated into polymeric hydrogel formulations to preserve their stability and help tune their release in the body to their most favorable profile of action for a given therapy. In this study, a series of injectable, thermoresponsive hydrogel formulations were evaluated as controlled delivery systems for various peptides and proteins, including insulin, Merck proprietary peptides (glucagon-like peptide analogue and modified insulin analogue), bovine serum albumin, and immunoglobulin G. These hydrogels were prepared using concentrated solutions of poly(lactide-co-glycolide)-block-poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PLGA-PEG-PLGA), which can undergo temperature-induced sol-gel transitions and spontaneously solidify into hydrogels near the body temperature, serving as an in situ depot for sustained drug release. The thermoresponsiveness and gelation properties of these triblock copolymers were characterized by dynamic light scattering (DLS) and oscillatory rheology, respectively. The impact of different hydrogel-forming polymers on release kinetics was systematically investigated based on their hydrophobicity (LA/GA ratios), polymer concentrations (20, 25, and 30%), and phase stability. These hydrogels were able to release active peptides and proteins in a controlled manner from 4 to 35 days, depending on the polymer concentration, solubility nature, and molecular sizes of the cargoes. Biophysical studies via size exclusion chromatography (SEC) and circular dichroism (CD) indicated that the encapsulation and release did not adversely affect the protein conformation and stability. Finally, a selected PLGA-PEG-PLGA hydrogel system was further investigated by the encapsulation of a therapeutic glucagon-like peptide analogue and a modified insulin peptide analogue in diabetic mouse and minipig models for studies of glucose-lowering efficacy and pharmacokinetics, where superior sustained peptide release profiles and long-lasting glucose-lowering effects were observed in vivo without any significant tolerability issues compared to peptide solution controls. These results suggest the promise of developing injectable thermoresponsive hydrogel formulations for the tunable release of protein therapeutics to improve patient's comfort, convenience, and compliance.
Collapse
Affiliation(s)
- Kingshuk Dutta
- Discovery
Pharmaceutical Sciences, Merck & Co.,
Inc., West Point, Pennsylvania 19486, United States
- Department
of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ritam Das
- Department
of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jing Ling
- Discovery
Pharmaceutical Sciences, Merck & Co.,
Inc., South San Francisco, California 94080, United States
| | - Rafael Mayoral Monibas
- Discovery
Biology, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Ester Carballo-Jane
- External
In Vivo Pharmacology, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Ahmet Kekec
- Chemistry
Capabilities Accelerating Therapeutics, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Danqing Dennis Feng
- Chemistry
Capabilities Accelerating Therapeutics, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Songnian Lin
- Chemistry
Capabilities Accelerating Therapeutics, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - James Mu
- Discovery
Biology, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Robert Saklatvala
- Discovery
Pharmaceutical Sciences, Merck & Co.,
Inc., Boston, Massachusetts 02115, United States
| | - S. Thayumanavan
- Department
of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Yingkai Liang
- Discovery
Pharmaceutical Sciences, Merck & Co.,
Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
13
|
Global Transcriptomic Analysis of Zebrafish Glucagon Receptor Mutant Reveals Its Regulated Metabolic Network. Int J Mol Sci 2020; 21:ijms21030724. [PMID: 31979106 PMCID: PMC7037442 DOI: 10.3390/ijms21030724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 12/18/2022] Open
Abstract
The glucagon receptor (GCGR) is a G-protein-coupled receptor (GPCR) that mediates the activity of glucagon. Disruption of GCGR results in many metabolic alterations, including increased glucose tolerance, decreased adiposity, hypoglycemia, and pancreatic α-cell hyperplasia. To better understand the global transcriptomic changes resulting from GCGR deficiency, we performed whole-organism RNA sequencing analysis in wild type and gcgr-deficient zebrafish. We found that the expression of 1645 genes changes more than two-fold among mutants. Most of these genes are related to metabolism of carbohydrates, lipids, and amino acids. Genes related to fatty acid β-oxidation, amino acid catabolism, and ureagenesis are often downregulated. Among gcrgr-deficient zebrafish, we experimentally confirmed increases in lipid accumulation in the liver and whole-body glucose uptake, as well as a modest decrease in total amino acid content. These results provide new information about the global metabolic network that GCGR signaling regulates in addition to a better understanding of the receptor’s physiological functions.
Collapse
|
14
|
Wewer Albrechtsen NJ, Pedersen J, Galsgaard KD, Winther-Sørensen M, Suppli MP, Janah L, Gromada J, Vilstrup H, Knop FK, Holst JJ. The Liver-α-Cell Axis and Type 2 Diabetes. Endocr Rev 2019; 40:1353-1366. [PMID: 30920583 DOI: 10.1210/er.2018-00251] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/19/2019] [Indexed: 02/08/2023]
Abstract
Both type 2 diabetes (T2D) and nonalcoholic fatty liver disease (NAFLD) strongly associate with increasing body mass index, and together these metabolic diseases affect millions of individuals. In patients with T2D, increased secretion of glucagon (hyperglucagonemia) contributes to diabetic hyperglycemia as proven by the significant lowering of fasting plasma glucose levels following glucagon receptor antagonist administration. Emerging data now indicate that the elevated plasma concentrations of glucagon may also be associated with hepatic steatosis and not necessarily with the presence or absence of T2D. Thus, fatty liver disease, most often secondary to overeating, may result in impaired amino acid turnover, leading to increased plasma concentrations of certain glucagonotropic amino acids (e.g., alanine). This, in turn, causes increased glucagon secretion that may help to restore amino acid turnover and ureagenesis, but it may eventually also lead to increased hepatic glucose production, a hallmark of T2D. Early experimental findings support the hypothesis that hepatic steatosis impairs glucagon's actions on amino acid turnover and ureagenesis. Hepatic steatosis also impairs hepatic insulin sensitivity and clearance that, together with hyperglycemia and hyperaminoacidemia, lead to peripheral hyperinsulinemia; systemic hyperinsulinemia may itself contribute to worsen peripheral insulin resistance. Additionally, obesity is accompanied by an impaired incretin effect, causing meal-related glucose intolerance. Lipid-induced impairment of hepatic sensitivity, not only to insulin but potentially also to glucagon, resulting in both hyperinsulinemia and hyperglucagonemia, may therefore contribute to the development of T2D at least in a subset of individuals with NAFLD.
Collapse
Affiliation(s)
- Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiology, Nephrology and Endocrinology, Nordsjællands Hospital Hillerød, University of Copenhagen, Hillerød, Denmark
| | - Katrine D Galsgaard
- 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
| | - Marie Winther-Sørensen
- 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
| | - Malte P Suppli
- Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Lina Janah
- 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
| | | | - Hendrik Vilstrup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Filip K Knop
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Department of Clinical Medicine, 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
| |
Collapse
|
15
|
Gao C, Ren SV, Yu J, Baal U, Thai D, Lu J, Zeng C, Yan H, Wang Y. Glucagon Receptor Antagonism Ameliorates Progression of Heart Failure. JACC Basic Transl Sci 2019; 4:161-172. [PMID: 31061918 PMCID: PMC6488764 DOI: 10.1016/j.jacbts.2018.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 02/08/2023]
Abstract
Mice were treated with a fully human monoclonal glucagon receptor antagonistic antibody REMD2.59 following myocardial infarction or pressure overload. REMD2.59 treatment blunted cardiac hypertrophy and fibrotic remodeling, and attenuated contractile dysfunction at 4 weeks after myocardial infarction. In addition, REMD2.59 treatment at the onset of pressure overload significantly suppressed cardiac hypertrophy and chamber dilation with marked preservation of cardiac systolic and diastolic function. Initiation of REMD2.59 treatment 2 weeks after pressure overload significantly blunted the progression of cardiac pathology. These results provide the first in vivo proof-of-concept evidence that glucagon receptor antagonism is a potentially efficacious therapy to ameliorate both onset and progression of heart failure.
Collapse
Affiliation(s)
- Chen Gao
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Address for correspondence: Dr. Yibin Wang or Dr. Chen Gao, Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, 650 Charles E. Young Drive, Room CHS 37-200J, Los Angeles, California 90095.
| | - Shuxun Vincent Ren
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Junyi Yu
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Ulysis Baal
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Dung Thai
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - John Lu
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Hai Yan
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Address for correspondence: Dr. Yibin Wang or Dr. Chen Gao, Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, 650 Charles E. Young Drive, Room CHS 37-200J, Los Angeles, California 90095.
| |
Collapse
|
16
|
Design, synthesis, and effects of novel phenylpyrimidines as glucagon receptor antagonists. Bioorg Med Chem 2018; 26:5701-5710. [PMID: 30366787 DOI: 10.1016/j.bmc.2018.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/16/2018] [Accepted: 10/18/2018] [Indexed: 11/20/2022]
Abstract
The hormone glucagon increases blood glucose levels through increasing hepatic glucose output. In diabetic patients, dysregulation of glucagon secretion contributes to hyperglycemia. Thus, the inhibition of glucagon receptor is one target for the treatment of hyperglycemia in type 2 diabetes. Here we designed and synthesized a series of small molecules based on phenylpyrimidine. Of these, the compound (R)-7a most significantly decreased the glucagon-induced cAMP production and glucagon-induced glucose production during in vitro and in vivo assays. In addition, (R)-7a showed good efficacy in glucagon challenge tests and lowered blood glucose levels in diabetic db/db mice. Our results suggest that the compound (R)-7a could be a potential glucose-lowering agent for treating type 2 diabetes.
Collapse
|
17
|
Abstract
Globally, 13% of the world's adult population is obese, and more than 400 million people suffer from diabetes. These conditions are both associated with significant morbidity, mortality and financial cost. Therefore, finding new pharmacological treatments is an imperative. Relative hyperglucagonaemia is seen in all types of diabetes, and has been implicated in its pathogenesis. Consequently, clinical trials are underway using drugs which block glucagon activity to treat type 2 diabetes. Conversely, exogenous glucagon can increase energy expenditure. Therefore, researchers are designing peptides that combine activation of the glucagon receptor with further incretin properties, which will treat obesity while mitigating the hyperglycaemic effects of glucagon. This review will discuss these conflicting physiological properties of glucagon, and the attempts to harness these effects pharmacologically.
Collapse
Affiliation(s)
- R V Scott
- Imperial College London, 6th Floor, Commonwealth Building, Hammersmith Hospital, London, W12 0NN, United Kingdom.
| | - S R Bloom
- Imperial College London, 6th Floor, Commonwealth Building, Hammersmith Hospital, London, W12 0NN, United Kingdom.
| |
Collapse
|
18
|
Hayashi Y, Seino Y. Regulation of amino acid metabolism and α-cell proliferation by glucagon. J Diabetes Investig 2018; 9:464-472. [PMID: 29314731 PMCID: PMC5934249 DOI: 10.1111/jdi.12797] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 12/21/2017] [Indexed: 12/25/2022] Open
Abstract
Both glucagon and glucagon-like peptide-1 (GLP-1) are produced from proglucagon through proteolytic cleavage. Blocking glucagon action increases the circulating levels of glucagon and GLP-1, reduces the blood glucose level, and induces the proliferation of islet α-cells. Glucagon blockade also suppresses hepatic amino acid catabolism and increases the serum amino acid level. In animal models defective in both glucagon and GLP-1, the blood glucose level is not reduced, indicating that GLP-1 is required for glucagon blockade to reduce the blood glucose level. In contrast, hyperplasia of α-cells and hyperaminoacidemia are observed in such animal models, indicating that GLP-1 is not required for the regulation of α-cell proliferation or amino acid metabolism. These findings suggest that the regulation of amino acid metabolism is a more important specific physiological role of glucagon than the regulation of glucose metabolism. Although the effects of glucagon deficiency on glucose metabolism are compensated by the suppression of insulin secretion, the effects on amino acid metabolism are not. Recently, data showing a feedback regulatory mechanism between the liver and islet α-cells, which is mediated by glucagon and amino acids, are accumulating. However, a number of questions on the mechanism of this regulation remain to be addressed. The profile of glucagon as a regulator of amino acid metabolism must be carefully considered for glucagon blockade to be applied therapeutically in the treatment of patients with diabetes.
Collapse
Affiliation(s)
- Yoshitaka Hayashi
- Division of Stress Adaptation and ProtectionResearch Institute of Environmental MedicineNagoyaJapan
| | - Yusuke Seino
- Department of Endocrinology and DiabetesNagoya University Graduate School of MedicineNagoya UniversityNagoyaJapan
| |
Collapse
|
19
|
Kim J, Okamoto H, Huang Z, Anguiano G, Chen S, Liu Q, Cavino K, Xin Y, Na E, Hamid R, Lee J, Zambrowicz B, Unger R, Murphy AJ, Xu Y, Yancopoulos GD, Li WH, Gromada J. Amino Acid Transporter Slc38a5 Controls Glucagon Receptor Inhibition-Induced Pancreatic α Cell Hyperplasia in Mice. Cell Metab 2017; 25:1348-1361.e8. [PMID: 28591637 PMCID: PMC8206958 DOI: 10.1016/j.cmet.2017.05.006] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 03/09/2017] [Accepted: 05/21/2017] [Indexed: 12/22/2022]
Abstract
Glucagon supports glucose homeostasis by stimulating hepatic gluconeogenesis, in part by promoting the uptake and conversion of amino acids into gluconeogenic precursors. Genetic disruption or pharmacologic inhibition of glucagon signaling results in elevated plasma amino acids and compensatory glucagon hypersecretion involving expansion of pancreatic α cell mass. Recent findings indicate that hyperaminoacidemia triggers pancreatic α cell proliferation via an mTOR-dependent pathway. We confirm and extend these findings by demonstrating that glucagon pathway blockade selectively increases expression of the sodium-coupled neutral amino acid transporter Slc38a5 in a subset of highly proliferative α cells and that Slc38a5 controls the pancreatic response to glucagon pathway blockade; most notably, mice deficient in Slc38a5 exhibit markedly decreased α cell hyperplasia to glucagon pathway blockade-induced hyperaminoacidemia. These results show that Slc38a5 is a key component of the feedback circuit between glucagon receptor signaling in the liver and amino-acid-dependent regulation of pancreatic α cell mass in mice.
Collapse
Affiliation(s)
- Jinrang Kim
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Haruka Okamoto
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - ZhiJiang Huang
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Guillermo Anguiano
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Shiuhwei Chen
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Qing Liu
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Katie Cavino
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Yurong Xin
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Erqian Na
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Rachid Hamid
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Joseph Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Roger Unger
- Touchstone Center for Diabetes Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | | | - Yan Xu
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | | | - Wen-Hong Li
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| | - Jesper Gromada
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA.
| |
Collapse
|
20
|
Müller TD, Finan B, Clemmensen C, DiMarchi RD, Tschöp MH. The New Biology and Pharmacology of Glucagon. Physiol Rev 2017; 97:721-766. [PMID: 28275047 DOI: 10.1152/physrev.00025.2016] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last two decades we have witnessed sizable progress in defining the role of gastrointestinal signals in the control of glucose and energy homeostasis. Specifically, the molecular basis of the huge metabolic benefits in bariatric surgery is emerging while novel incretin-based medicines based on endogenous hormones such as glucagon-like peptide 1 and pancreas-derived amylin are improving diabetes management. These and related developments have fostered the discovery of novel insights into endocrine control of systemic metabolism, and in particular a deeper understanding of the importance of communication across vital organs, and specifically the gut-brain-pancreas-liver network. Paradoxically, the pancreatic peptide glucagon has reemerged in this period among a plethora of newly identified metabolic macromolecules, and new data complement and challenge its historical position as a gut hormone involved in metabolic control. The synthesis of glucagon analogs that are biophysically stable and soluble in aqueous solutions has promoted biological study that has enriched our understanding of glucagon biology and ironically recruited glucagon agonism as a central element to lower body weight in the treatment of metabolic disease. This review summarizes the extensive historical record and the more recent provocative direction that integrates the prominent role of glucagon in glucose elevation with its under-acknowledged effects on lipids, body weight, and vascular health that have implications for the pathophysiology of metabolic diseases, and the emergence of precision medicines to treat metabolic diseases.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - B Finan
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - C Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - R D DiMarchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| |
Collapse
|
21
|
Holst JJ, Wewer Albrechtsen NJ, Pedersen J, Knop FK. Glucagon and Amino Acids Are Linked in a Mutual Feedback Cycle: The Liver-α-Cell Axis. Diabetes 2017; 66:235-240. [PMID: 28108603 DOI: 10.2337/db16-0994] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/31/2016] [Indexed: 11/13/2022]
Abstract
Glucagon is usually viewed as an important counterregulatory hormone in glucose metabolism, with actions opposing those of insulin. Evidence exists that shows glucagon is important for minute-to-minute regulation of postprandial hepatic glucose production, although conditions of glucagon excess or deficiency do not cause changes compatible with this view. In patients with glucagon-producing tumors (glucagonomas), the most conspicuous signs are skin lesions (necrolytic migratory erythema), while in subjects with inactivating mutations of the glucagon receptor, pancreatic swelling may be the first sign; neither condition is necessarily associated with disturbed glucose metabolism. In glucagonoma patients, amino acid turnover and ureagenesis are greatly accelerated, and low plasma amino acid levels are probably at least partly responsible for the necrolytic migratory erythema, which resolves after amino acid administration. In patients with receptor mutations (and in knockout mice), pancreatic swelling is due to α-cell hyperplasia with gross hypersecretion of glucagon, which according to recent groundbreaking research may result from elevated amino acid levels. Additionally, solid evidence indicates that ureagenesis, and thereby amino acid levels, is critically controlled by glucagon. Together, this constitutes a complete endocrine system; feedback regulation involving amino acids regulates α-cell function and secretion, while glucagon, in turn, regulates amino acid turnover.
Collapse
Affiliation(s)
- 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
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- 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
| | - Filip K Knop
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| |
Collapse
|
22
|
Angptl4 does not control hyperglucagonemia or α-cell hyperplasia following glucagon receptor inhibition. Proc Natl Acad Sci U S A 2017; 114:2747-2752. [PMID: 28143927 DOI: 10.1073/pnas.1620989114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Genetic disruption or pharmacologic inhibition of glucagon signaling effectively lowers blood glucose but results in compensatory glucagon hypersecretion involving expansion of pancreatic α-cell mass. Ben-Zvi et al. recently reported that angiopoietin-like protein 4 (Angptl4) links glucagon receptor inhibition to hyperglucagonemia and α-cell proliferation [Ben-Zvi et al. (2015) Proc Natl Acad Sci USA 112:15498-15503]. Angptl4 is a secreted protein and inhibitor of lipoprotein lipase-mediated plasma triglyceride clearance. We report that Angptl4-/- mice treated with an anti-glucagon receptor monoclonal antibody undergo elevation of plasma glucagon levels and α-cell expansion similar to wild-type mice. Overexpression of Angptl4 in liver of mice caused a 8.6-fold elevation in plasma triglyceride levels, but did not alter plasma glucagon levels or α-cell mass. Furthermore, administration of glucagon receptor-blocking antibody to healthy individuals increased plasma glucagon and amino acid levels, but did not change circulating Angptl4 concentration. These data show that Angptl4 does not link glucagon receptor inhibition to compensatory hyperglucagonemia or expansion of α-cell mass, and that it cannot be given to induce such secretion and growth. The reduction of plasma triglyceride levels in Angptl4-/- mice and increase following Angptl4 overexpression suggest that changes in plasma triglyceride metabolism do not regulate α-cells in the pancreas. Our findings corroborate recent data showing that increased plasma amino acids and their transport into α-cells link glucagon receptor blockage to α-cell hyperplasia.
Collapse
|
23
|
Glucagon receptor inhibition normalizes blood glucose in severe insulin-resistant mice. Proc Natl Acad Sci U S A 2017; 114:2753-2758. [PMID: 28115707 DOI: 10.1073/pnas.1621069114] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inactivating mutations in the insulin receptor results in extreme insulin resistance. The resulting hyperglycemia is very difficult to treat, and patients are at risk for early morbidity and mortality from complications of diabetes. We used the insulin receptor antagonist S961 to induce severe insulin resistance, hyperglycemia, and ketonemia in mice. Using this model, we show that glucagon receptor (GCGR) inhibition with a monoclonal antibody normalized blood glucose and β-hydroxybutyrate levels. Insulin receptor antagonism increased pancreatic β-cell mass threefold. Normalization of blood glucose levels with GCGR-blocking antibody unexpectedly doubled β-cell mass relative to that observed with S961 alone and 5.8-fold over control. GCGR antibody blockage expanded α-cell mass 5.7-fold, and S961 had no additional effects. Collectively, these data show that GCGR antibody inhibition represents a potential therapeutic option for treatment of patients with extreme insulin-resistance syndromes.
Collapse
|
24
|
Bankir L, Bouby N, Blondeau B, Crambert G. Glucagon actions on the kidney revisited: possible role in potassium homeostasis. Am J Physiol Renal Physiol 2016; 311:F469-86. [DOI: 10.1152/ajprenal.00560.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/31/2016] [Indexed: 12/25/2022] Open
Abstract
It is now recognized that the metabolic disorders observed in diabetes are not, or not only due to the lack of insulin or insulin resistance, but also to elevated glucagon secretion. Accordingly, selective glucagon receptor antagonists are now proposed as a novel strategy for the treatment of diabetes. However, besides its metabolic actions, glucagon also influences kidney function. The glucagon receptor is expressed in the thick ascending limb, distal tubule, and collecting duct, and glucagon regulates the transepithelial transport of several solutes in these nephron segments. Moreover, it also influences solute transport in the proximal tubule, possibly by an indirect mechanism. This review summarizes the knowledge accumulated over the last 30 years about the influence of glucagon on the renal handling of electrolytes and urea. It also describes a possible novel role of glucagon in the short-term regulation of potassium homeostasis. Several original findings suggest that pancreatic α-cells may express a “potassium sensor” sensitive to changes in plasma K concentration and could respond by adapting glucagon secretion that, in turn, would regulate urinary K excretion. By their combined actions, glucagon and insulin, working in a combinatory mode, could ensure an independent regulation of both plasma glucose and plasma K concentrations. The results and hypotheses reviewed here suggest that the use of glucagon receptor antagonists for the treatment of diabetes should take into account their potential consequences on electrolyte handling by the kidney.
Collapse
Affiliation(s)
- Lise Bankir
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie, Paris, France; and
| | - Nadine Bouby
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie, Paris, France; and
- Université Paris-Descartes, Paris, France
| | - Bertrand Blondeau
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie, Paris, France; and
| | - Gilles Crambert
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Pierre et Marie Curie, Paris, France; and
| |
Collapse
|
25
|
Kazierad DJ, Bergman A, Tan B, Erion DM, Somayaji V, Lee DS, Rolph T. Effects of multiple ascending doses of the glucagon receptor antagonist PF-06291874 in patients with type 2 diabetes mellitus. Diabetes Obes Metab 2016; 18:795-802. [PMID: 27059951 DOI: 10.1111/dom.12672] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/21/2023]
Abstract
AIMS To assess the pharmacokinetics, pharmacodynamics, safety and tolerability of multiple ascending doses of the glucagon receptor antagonist PF-06291874 in patients with type 2 diabetes mellitus (T2DM). METHODS Patients were randomized to oral PF-06291874 or placebo on a background of either metformin (Part A, Cohorts 1-5: 5-150 mg once daily), or metformin and sulphonylurea (Part B, Cohorts 1-2: 15 or 30 mg once daily) for 14-28 days. A mixed-meal tolerance test (MMTT) was administered on days -1 (baseline), 14 and 28. Assessments were conducted with regard to pharmacokinetics, various pharmacodynamic variables, safety and tolerability. Circulating amino acid concentrations were also measured. RESULTS PF-06291874 exposure was approximately dose-proportional with a half-life of ∼19.7-22.7 h. Day 14 fasting plasma glucose and mean daily glucose values were reduced from baseline in a dose-dependent manner, with placebo-corrected decreases of 34.3 and 42.4 mg/dl, respectively, at the 150 mg dose. After the MMTT, dose-dependent increases in glucagon and total glucagon-like peptide-1 (GLP-1) were observed, although no meaningful changes were noted in insulin, C-peptide or active GLP-1 levels. Small dose-dependent increases in LDL cholesterol were observed, along with reversible increases in serum aminotransferases that were largely within the laboratory reference range. An increase in circulating gluconeogenic amino acids was also observed on days 2 and 14. All dose levels of PF-06291874 were well tolerated. CONCLUSION PF-06291874 was well tolerated, has a pharmacokinetic profile suitable for once-daily dosing, and results in reductions in glucose with minimal risk of hypoglycaemia.
Collapse
Affiliation(s)
| | | | - B Tan
- Pfizer, Cambridge, MA, USA
| | | | | | | | | |
Collapse
|
26
|
Abstract
Type 2 diabetes mellitus is the result of impaired systemic control of glucose homeostasis, in part through the dysregulation of the hormone glucagon. Glucagon acts on the liver to increase glucose production through alterations in hepatic metabolism, and reducing the elevated glucagon signalling in diabetic patients is an attractive strategy for the treatment of hyperglycaemia. Here we review the actions of the hormone in the liver, focusing on the acute alterations of metabolic pathways. This review summarises a presentation given at the 'Novel data on glucagon' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Mona Abraham and Tony Lam, DOI: 10.1007/s00125-016-3950-3 , and by Young Lee and colleagues, DOI: 10.1007/s00125-016-3965-9 ) and an overview by the Session Chair, Isabel Valverde (DOI: 10.1007/s00125-016-3946-z ).
Collapse
Affiliation(s)
- Russell A Miller
- Pfizer Inc. CVMET RU, 610 Main Street, Cambridge, MA, 02139, USA
| | | |
Collapse
|
27
|
Gaitonde P, Garhyan P, Link C, Chien JY, Trame MN, Schmidt S. A Comprehensive Review of Novel Drug–Disease Models in Diabetes Drug Development. Clin Pharmacokinet 2016; 55:769-788. [DOI: 10.1007/s40262-015-0359-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
28
|
Angptl4 links α-cell proliferation following glucagon receptor inhibition with adipose tissue triglyceride metabolism. Proc Natl Acad Sci U S A 2015; 112:15498-503. [PMID: 26621734 DOI: 10.1073/pnas.1513872112] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Type 2 diabetes is characterized by a reduction in insulin function and an increase in glucagon activity that together result in hyperglycemia. Glucagon receptor antagonists have been developed as drugs for diabetes; however, they often increase glucagon plasma levels and induce the proliferation of glucagon-secreting α-cells. We find that the secreted protein Angiopoietin-like 4 (Angptl4) is up-regulated via Pparγ activation in white adipose tissue and plasma following an acute treatment with a glucagon receptor antagonist. Induction of adipose angptl4 and Angptl4 supplementation promote α-cell proliferation specifically. Finally, glucagon receptor antagonist improves glycemia in diet-induced obese angptl4 knockout mice without increasing glucagon levels or α-cell proliferation, underscoring the importance of this protein. Overall, we demonstrate that triglyceride metabolism in adipose tissue regulates α-cells in the endocrine pancreas.
Collapse
|
29
|
Ozcan L, Xu X, Deng SX, Ghorpade DS, Thomas T, Cremers S, Hubbard B, Serrano-Wu MH, Gaestel M, Landry DW, Tabas I. Treatment of Obese Insulin-Resistant Mice With an Allosteric MAPKAPK2/3 Inhibitor Lowers Blood Glucose and Improves Insulin Sensitivity. Diabetes 2015; 64:3396-405. [PMID: 26068544 PMCID: PMC4587644 DOI: 10.2337/db14-1945] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 06/04/2015] [Indexed: 12/31/2022]
Abstract
The prevalence of obesity-induced type 2 diabetes (T2D) is increasing worldwide, and new treatment strategies are needed. We recently discovered that obesity activates a previously unknown pathway that promotes both excessive hepatic glucose production (HGP) and defective insulin signaling in hepatocytes, leading to exacerbation of hyperglycemia and insulin resistance in obesity. At the hub of this new pathway is a kinase cascade involving calcium/calmodulin-dependent protein kinase II (CaMKII), p38α mitogen-activated protein kinase (MAPK), and MAPKAPK2/3 (MK2/3). Genetic-based inhibition of these kinases improves metabolism in obese mice. Here, we report that treatment of obese insulin-resistant mice with an allosteric MK2/3 inhibitor, compound (cmpd) 28, ameliorates glucose homeostasis by suppressing excessive HGP and enhancing insulin signaling. The metabolic improvement seen with cmpd 28 is additive with the leading T2D drug, metformin, but it is not additive with dominant-negative MK2, suggesting an on-target mechanism of action. Allosteric MK2/3 inhibitors represent a potentially new approach to T2D that is highly mechanism based, has links to human T2D, and is predicted to avoid certain adverse effects seen with current T2D drugs.
Collapse
Affiliation(s)
- Lale Ozcan
- Department of Medicine, Columbia University, New York, NY
| | - Xiaoming Xu
- Department of Medicine, Columbia University, New York, NY
| | - Shi-Xian Deng
- Department of Medicine, Columbia University, New York, NY
| | | | - Tiffany Thomas
- Department of Pathology and Cell Biology, Columbia University, New York, NY Irving Institute for Clinical and Translational Research, Columbia University, New York, NY
| | - Serge Cremers
- Department of Medicine, Columbia University, New York, NY Department of Pathology and Cell Biology, Columbia University, New York, NY Irving Institute for Clinical and Translational Research, Columbia University, New York, NY
| | | | | | - Matthias Gaestel
- Department of Biochemistry, Hannover Medical School, Hannover, Germany
| | | | - Ira Tabas
- Department of Medicine, Columbia University, New York, NY Department of Pathology and Cell Biology, Columbia University, New York, NY Department of Physiology and Cellular Biophysics, Columbia University, New York, NY
| |
Collapse
|
30
|
Guan HP, Yang X, Lu K, Wang SP, Castro-Perez JM, Previs S, Wright M, Shah V, Herath K, Xie D, Szeto D, Forrest G, Xiao JC, Palyha O, Sun LP, Andryuk PJ, Engel SS, Xiong Y, Lin S, Kelley DE, Erion MD, Davis HR, Wang L. Glucagon receptor antagonism induces increased cholesterol absorption. J Lipid Res 2015; 56:2183-95. [PMID: 26373568 DOI: 10.1194/jlr.m060897] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Indexed: 12/26/2022] Open
Abstract
Glucagon and insulin have opposing action in governing glucose homeostasis. In type 2 diabetes mellitus (T2DM), plasma glucagon is characteristically elevated, contributing to increased gluconeogenesis and hyperglycemia. Therefore, glucagon receptor (GCGR) antagonism has been proposed as a pharmacologic approach to treat T2DM. In support of this concept, a potent small-molecule GCGR antagonist (GRA), MK-0893, demonstrated dose-dependent efficacy to reduce hyperglycemia, with an HbA1c reduction of 1.5% at the 80 mg dose for 12 weeks in T2DM. However, GRA treatment was associated with dose-dependent elevation of plasma LDL-cholesterol (LDL-c). The current studies investigated the cause for increased LDL-c. We report findings that link MK-0893 with increased glucagon-like peptide 2 and cholesterol absorption. There was not, however, a GRA-related modulation of cholesterol synthesis. These findings were replicated using structurally diverse GRAs. To examine potential pharmacologic mitigation, coadministration of ezetimibe (a potent inhibitor of cholesterol absorption) in mice abrogated the GRA-associated increase of LDL-c. Although the molecular mechanism is unknown, our results provide a novel finding by which glucagon and, hence, GCGR antagonism govern cholesterol metabolism.
Collapse
Affiliation(s)
- Hong-Ping Guan
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Xiaodong Yang
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Ku Lu
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Sheng-Ping Wang
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Jose M Castro-Perez
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Stephen Previs
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Michael Wright
- Late Stage In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Vinit Shah
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Kithsiri Herath
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Dan Xie
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Daphne Szeto
- Late Stage In Vivo Pharmacology, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Gail Forrest
- Late Stage In Vivo Pharmacology, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Jing Chen Xiao
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Oksana Palyha
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Li-Ping Sun
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Paula J Andryuk
- Clinical Research Department, Merck Research Laboratories, Rahway, NJ 07065
| | - Samuel S Engel
- Clinical Research Department, Merck Research Laboratories, Rahway, NJ 07065
| | - Yusheng Xiong
- Discovery Chemistry, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Songnian Lin
- Discovery Chemistry, Merck Research Laboratories, Kenilworth, NJ 07033
| | - David E Kelley
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Mark D Erion
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Harry R Davis
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| | - Liangsu Wang
- Departments of Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ 07033
| |
Collapse
|
31
|
Sammons MF, Lee ECY. Recent progress in the development of small-molecule glucagon receptor antagonists. Bioorg Med Chem Lett 2015; 25:4057-64. [PMID: 26271588 DOI: 10.1016/j.bmcl.2015.07.092] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/24/2015] [Accepted: 07/28/2015] [Indexed: 01/05/2023]
Abstract
The endocrine hormone glucagon stimulates hepatic glucose output via its action at the glucagon receptor (GCGr) in the liver. In the diabetic state, dysregulation of glucagon secretion contributes to abnormally elevated hepatic glucose output. The inhibition of glucagon-induced hepatic glucose output via antagonism of the GCGr using small-molecule ligands is a promising mechanism for improving glycemic control in the diabetic state. Clinical data evaluating the therapeutic potential of small-molecule GCGr antagonists is currently emerging. Recently disclosed clinical data demonstrates the potential efficacy and possible therapeutic limitations of small-molecule GCGr antagonists. Recent pre-clinical work on the development of GCGr antagonists is also summarized.
Collapse
Affiliation(s)
- Matthew F Sammons
- Cardiovascular, Metabolic and Endocrine Diseases Chemistry, Pfizer Worldwide Research and Development, 610 Main St, Cambridge, MA 02139, United States
| | - Esther C Y Lee
- Cardiovascular, Metabolic and Endocrine Diseases Chemistry, Pfizer Worldwide Research and Development, 610 Main St, Cambridge, MA 02139, United States
| |
Collapse
|
32
|
Bernardo B, Lu M, Bandyopadhyay G, Li P, Zhou Y, Huang J, Levin N, Tomas EM, Calle RA, Erion DM, Rolph TP, Brenner M, Talukdar S. FGF21 does not require interscapular brown adipose tissue and improves liver metabolic profile in animal models of obesity and insulin-resistance. Sci Rep 2015; 5:11382. [PMID: 26153793 PMCID: PMC4495598 DOI: 10.1038/srep11382] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/22/2015] [Indexed: 12/22/2022] Open
Abstract
FGF21 is a key metabolic regulator modulating physiological processes and its pharmacological administration improves metabolic profile in preclinical species and humans. We used native-FGF21 and a long-acting FGF21 (PF-05231023), to determine the contribution of liver and brown adipose tissue (BAT) towards metabolic improvements in Zucker rats and DIO mice (DIOs). FGF21 improved glucose tolerance and liver insulin sensitivity in Zuckers without affecting BW and improved liver function by decreased lipogenesis, increased fatty acid oxidation and improved insulin signaling. Through detailed lipidomic analyses of liver metabolites in DIOs, we demonstrate that FGF21 favorably alters liver metabolism. We observed a dose-dependent increase of [(18)F]-FDG-glucose uptake in interscapular BAT (iBAT) of DIOs upon FGF21 administration. Upon excision of iBAT (X-BAT) and administration of FGF21 to mice housed at 80 °F or 72 °F, the favorable effects of FGF21 on BW and glucose excursion were fully retained in both sham and X-BAT animals. Taken together, we demonstrate the liver as an organ that integrates the actions of FGF21 and provide metabolic benefits of FGF21 in Zucker rats and DIOs. Finally, our data demonstrates iBAT does not play a role in mediating favorable metabolic effects of FGF21 administration in DIOs housed at 80 °F or 72 °F.
Collapse
Affiliation(s)
- Barbara Bernardo
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| | - Min Lu
- 1] Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA [2] CovX Research, Pfizer WRD, USA
| | - Gautam Bandyopadhyay
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Pingping Li
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yingjiang Zhou
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| | | | | | - Eva M Tomas
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| | - Roberto A Calle
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| | - Derek M Erion
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| | - Timothy P Rolph
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| | - Martin Brenner
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| | - Saswata Talukdar
- Cardiovascular Metabolic and Endocrine Diseases (CVMED) Pfizer, Inc. 610 Main Street, Cambridge, MA 02139, USA
| |
Collapse
|
33
|
Interaction of Glucagon G-Protein Coupled Receptor with Known Natural Antidiabetic Compounds: Multiscoring In Silico Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:497253. [PMID: 26236379 PMCID: PMC4508340 DOI: 10.1155/2015/497253] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/15/2015] [Indexed: 01/18/2023]
Abstract
Glucagon receptor (GCGR) is a secretin-like (class B) family of G-protein coupled receptors (GPCRs) in humans that plays an important role in elevating the glucose concentration in blood and has thus become one of the promising therapeutic targets for treatment of type 2 diabetes mellitus. GCGR based inhibitors for the treatment of type 2 diabetes are either glucagon neutralizers or small molecular antagonists. Management of diabetes without any side effects is still a challenge to the medical system, and the search for a new and effective natural GCGR antagonist is an important area for the treatment of type 2 diabetes. In the present study, a number of natural compounds containing antidiabetic properties were selected from the literature and their binding potential against GCGR was determined using molecular docking and other in silico approaches. Among all selected natural compounds, curcumin was found to be the most effective compound against GCGR followed by amorfrutin 1 and 4-hydroxyderricin. These compounds were rescored to confirm the accuracy of binding using another scoring function (x-score). The final conclusions were drawn based on the results obtained from the GOLD and x-score. Further experiments were conducted to identify the atomic level interactions of selected compounds with GCGR.
Collapse
|
34
|
Bankir L, Roussel R, Bouby N. Protein- and diabetes-induced glomerular hyperfiltration: role of glucagon, vasopressin, and urea. Am J Physiol Renal Physiol 2015; 309:F2-23. [DOI: 10.1152/ajprenal.00614.2014] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/13/2015] [Indexed: 12/21/2022] Open
Abstract
A single protein-rich meal (or an infusion of amino acids) is known to increase the glomerular filtration rate (GFR) for a few hours, a phenomenon known as “hyperfiltration.” It is important to understand the factors that initiate this upregulation because it becomes maladaptive in the long term. Several mediators and paracrine factors have been shown to participate in this upregulation, but they are not directly triggered by protein intake. Here, we explain how a rise in glucagon and in vasopressin secretion, directly induced by protein ingestion, might be the initial factors triggering the hepatic and renal events leading to an increase in the GFR. Their effects include metabolic actions in the liver and stimulation of sodium chloride reabsorption in the thick ascending limb. Glucagon is not only a glucoregulatory hormone. It is also important for the excretion of nitrogen end products by stimulating both urea synthesis in the liver (along with gluconeogenesis from amino acids) and urea excretion by the kidney. Vasopressin allows the concentration of nitrogenous end products (urea, ammonia, etc.) and other protein-associated wastes in a hyperosmotic urine, thus allowing a very significant water economy characteristic of all terrestrial mammals. No hyperfiltration occurs in the absence of one or the other hormone. Experimental results suggest that the combined actions of these two hormones, along with the complex intrarenal handling of urea, lead to alter the composition of the tubular fluid at the macula densa and to reduce the intensity of the signal activating the tubuloglomerular feedback control of GFR, thus allowing GFR to raise. Altogether, glucagon, vasopressin, and urea contribute to set up the best compromise between efficient urea excretion and water economy.
Collapse
Affiliation(s)
- Lise Bankir
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
| | - Ronan Roussel
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
- Diabétologie Endocrinologie Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France
| | - Nadine Bouby
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
| |
Collapse
|
35
|
Han J, Zhang M, Froese S, Dai FF, Robitaille M, Bhattacharjee A, Huang X, Jia W, Angers S, Wheeler MB, Wei L. The Identification of Novel Protein-Protein Interactions in Liver that Affect Glucagon Receptor Activity. PLoS One 2015; 10:e0129226. [PMID: 26075596 PMCID: PMC4468146 DOI: 10.1371/journal.pone.0129226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/06/2015] [Indexed: 11/18/2022] Open
Abstract
Glucagon regulates glucose homeostasis by controlling glycogenolysis and gluconeogenesis in the liver. Exaggerated and dysregulated glucagon secretion can exacerbate hyperglycemia contributing to type 2 diabetes (T2D). Thus, it is important to understand how glucagon receptor (GCGR) activity and signaling is controlled in hepatocytes. To better understand this, we sought to identify proteins that interact with the GCGR to affect ligand-dependent receptor activation. A Flag-tagged human GCGR was recombinantly expressed in Chinese hamster ovary (CHO) cells, and GCGR complexes were isolated by affinity purification (AP). Complexes were then analyzed by mass spectrometry (MS), and protein-GCGR interactions were validated by co-immunoprecipitation (Co-IP) and Western blot. This was followed by studies in primary hepatocytes to assess the effects of each interactor on glucagon-dependent glucose production and intracellular cAMP accumulation, and then in immortalized CHO and liver cell lines to further examine cell signaling. Thirty-three unique interactors were identified from the AP-MS screening of GCGR expressing CHO cells in both glucagon liganded and unliganded states. These studies revealed a particularly robust interaction between GCGR and 5 proteins, further validated by Co-IP, Western blot and qPCR. Overexpression of selected interactors in mouse hepatocytes indicated that two interactors, LDLR and TMED2, significantly enhanced glucagon-stimulated glucose production, while YWHAB inhibited glucose production. This was mirrored with glucagon-stimulated cAMP production, with LDLR and TMED2 enhancing and YWHAB inhibiting cAMP accumulation. To further link these interactors to glucose production, key gluconeogenic genes were assessed. Both LDLR and TMED2 stimulated while YWHAB inhibited PEPCK and G6Pase gene expression. In the present study, we have probed the GCGR interactome and found three novel GCGR interactors that control glucagon-stimulated glucose production by modulating cAMP accumulation and genes that control gluconeogenesis. These interactors may be useful targets to control glucose homeostasis in T2D.
Collapse
Affiliation(s)
- Junfeng Han
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ming Zhang
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sean Froese
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Feihan F. Dai
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mélanie Robitaille
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S1A8, Canada
| | - Alpana Bhattacharjee
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xinyi Huang
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Stéphane Angers
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S1A8, Canada
| | - Michael B. Wheeler
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (MW); (LW)
| | - Li Wei
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- * E-mail: (MW); (LW)
| |
Collapse
|
36
|
Luo J, Liang M, Mitch WE, Danesh FR, Yu M, Cheng J. FSP-1 Impairs the Function of Endothelium Leading to Failure of Arteriovenous Grafts in Diabetic Mice. Endocrinology 2015; 156:2200-10. [PMID: 25774552 PMCID: PMC4430603 DOI: 10.1210/en.2014-1841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To understand how endothelial cell (EC) dysfunction contributes to the failure of arteriovenous graft (AVG), we investigated the role of fibroblast-specific protein 1 (FSP-1) in cultured ECs and a mouse AVG model. In vitro, we uncovered a new FSP-1-dependent pathway that activates rho-associated, coiled-coil-containing protein kinase 1 (ROCK1) in ECs, leading to phosphorylation of myosin light chain 2 resulting in EC dysfunction. In cultured ECs, high glucose stimulated FSP-1 expression and increased permeability of an EC monolayer. The increase in permeability by the high glucose concentration was mediated by FSP-1 expression. Treatment of cultured ECs with FSP-1 caused leakage of the endothelial barrier plus increased expression of adhesion molecules and decreased expression of junction molecules. These responses were initiated by binding of FSP-1 to receptor for advanced glycation end products, which resulted in ROCK1 activation. In vivo, diabetes increased infiltration of inflammatory cells into AVGs and stimulated neointima formation. Increased FSP-1 expression and ROCK1 activation were found in AVGs of diabetic mice. Blocking FSP-1 suppressed diabetes-induced ROCK1 activation in AVGs. In mice with FSP-1 knockout or with ROCK1 knockout, accumulation of inflammatory cells and neointima formation in AVG were attenuated despite diabetes. Thus, mechanisms of inhibiting FSP-1 in ECs could improve AVG function.
Collapse
Affiliation(s)
- Jinlong Luo
- Nephrology Division (J.L., M.L., W.E.M., M.Y., J.C.), Baylor College of Medicine, Houston, Texas 77030; Emergency Medicine (F.R.D.), University of Texas MD Anderson Cancer Center, Houston, Texas 77030; and Department of Emergency (J.L.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China 430074
| | | | | | | | | | | |
Collapse
|
37
|
Kelly RP, Garhyan P, Raddad E, Fu H, Lim CN, Prince MJ, Pinaire JA, Loh MT, Deeg MA. Short-term administration of the glucagon receptor antagonist LY2409021 lowers blood glucose in healthy people and in those with type 2 diabetes. Diabetes Obes Metab 2015; 17:414-22. [PMID: 25656305 DOI: 10.1111/dom.12446] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/29/2015] [Accepted: 01/31/2015] [Indexed: 11/30/2022]
Abstract
AIM To describe the clinical effects of single and multiple doses of a potent, selective, orally administered, small-molecule antagonist of the human glucagon receptor, LY2409021, in healthy subjects and in patients with type 2 diabetes. METHODS LY2409021 was administered in dose-escalation studies to healthy subjects (n = 23) and patients with type 2 diabetes (n = 9) as single doses (Study 1) and daily to patients with type 2 diabetes (n = 47) for 28 days (Study 2). Safety, tolerability, pharmacokinetic (PK) and pharmacodynamic (PD) assessments were made after single doses and in patients receiving once-daily doses of LY2409021 (5, 30, 60 or 90 mg) for 28 days. RESULTS LY2409021 was well tolerated at all dose levels in both studies. Fasting and postprandial glucose were reduced and glucagon levels increased after single and multiple dosing, with reductions in fasting serum glucose of up to ∼1.25 mmol/l on day 28. Serum aminotransferases increased in a dose-dependent manner with multiple dosing and reversed after cessation of dosing. Significant glucose-lowering was observed with LY2409021 at dose levels associated with only minor aminotransferase increases. CONCLUSION Blockade of glucagon signalling in patients with type 2 diabetes is well tolerated and results in substantial reduction of fasting and postprandial glucose with minimal hypoglycaemia, but with reversible increases in aminotransferases. Inhibition of glucagon signalling by LY2409021 is a promising potential treatment for patients with type 2 diabetes and should be evaluated in longer clinical trials to better evaluate benefits and risks.
Collapse
Affiliation(s)
- R P Kelly
- Lilly-NUS, Clinical Research Centre, National University of Singapore, 10 Medical Drive, 117597, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Filipski KJ. Small molecule glucagon receptor antagonists: a patent review (2011 – 2014). Expert Opin Ther Pat 2015; 25:819-30. [DOI: 10.1517/13543776.2015.1032250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
39
|
Glucagon receptor antibody completely suppresses type 1 diabetes phenotype without insulin by disrupting a novel diabetogenic pathway. Proc Natl Acad Sci U S A 2015; 112:2503-8. [PMID: 25675519 DOI: 10.1073/pnas.1424934112] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Insulin monotherapy can neither maintain normoglycemia in type 1 diabetes (T1D) nor prevent the long-term damage indicated by elevated glycation products in blood, such as glycated hemoglobin (HbA1c). Here we find that hyperglycemia, when unaccompanied by an acute increase in insulin, enhances itself by paradoxically stimulating hyperglucagonemia. Raising glucose from 5 to 25 mM without insulin enhanced glucagon secretion ∼two- to fivefold in InR1-G9 α cells and ∼18-fold in perfused pancreata from insulin-deficient rats with T1D. Mice with T1D receiving insulin treatment paradoxically exhibited threefold higher plasma glucagon during hyperglycemic surges than during normoglycemic intervals. Blockade of glucagon action with mAb Ac, a glucagon receptor (GCGR) antagonizing antibody, maintained glucose below 100 mg/dL and HbA1c levels below 4% in insulin-deficient mice with T1D. In rodents with T1D, hyperglycemia stimulates glucagon secretion, up-regulating phosphoenolpyruvate carboxykinase and enhancing hyperglycemia. GCGR antagonism in mice with T1D normalizes glucose and HbA1c, even without insulin.
Collapse
|
40
|
Grover S, Dhanjal JK, Goyal S, Grover A, Sundar D. Computational identification of novel natural inhibitors of glucagon receptor for checking type II diabetes mellitus. BMC Bioinformatics 2014; 15 Suppl 16:S13. [PMID: 25521597 PMCID: PMC4290642 DOI: 10.1186/1471-2105-15-s16-s13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Interaction of the small peptide hormone glucagon with glucagon receptor (GCGR) stimulates the release of glucose from the hepatic cells during fasting; hence GCGR performs a significant function in glucose homeostasis. Inhibiting the interaction between glucagon and its receptor has been reported to control hepatic glucose overproduction and thus GCGR has evolved as an attractive therapeutic target for the treatment of type II diabetes mellitus. RESULTS In the present study, a large library of natural compounds was screened against 7 transmembrane domain of GCGR to identify novel therapeutic molecules that can inhibit the binding of glucagon with GCGR. Molecular dynamics simulations were performed to study the dynamic behaviour of the docked complexes and the molecular interactions between the screened compounds and the ligand binding residues of GCGR were analysed in detail. The top scoring compounds were also compared with already documented GCGR inhibitors- MK-0893 and LY2409021 for their binding affinity and other ADME properties. Finally, we have reported two natural drug like compounds PIB and CAA which showed good binding affinity for GCGR and are potent inhibitor of its functional activity. CONCLUSION This study contributes evidence for application of these compounds as prospective small ligand molecules against type II diabetes. Novel natural drug like inhibitors against the 7 transmembrane domain of GCGR have been identified which showed high binding affinity and potent inhibition of GCGR.
Collapse
|
41
|
Abstract
Diabetes is affecting more than 25.8 million people in the United States, causing huge burden on the health care system and economy. Insulin injection, which is the predominant treatment for diabetes, is incapable of replenishing the lost insulin-producing beta cell in patients. Restoring beta cell mass through replacement therapy such as islet transplantation or beta cell regeneration through in vitro and in vivo strategies has attracted particular attentions in the field due to its potential to cure diabetes. In the aspect of islet transplantation, gene therapy, stem cell therapy, and more biocompatible immunosuppressive drugs have been tested in various preclinical animal models to improve the longevity and function of human islets against the posttransplantation challenges. In the islet regeneration aspect, insulin-producing cells have been generated through in vitro transdifferentiation of stem cells and other types of cells and demonstrated to be capable of glycemic control. Moreover, several biomarkers including cell-surface receptors, soluble factors, and transcriptional factors have been identified or rediscovered in mediating the process of beta cell proliferation in rodents. This review summarizes the current progress and hurdles in the preclinical efforts in resurrecting beta cells. It may provide some useful insights into the future drug discovery for antidiabetic purposes.
Collapse
Affiliation(s)
| | - Hao Wu
- NGM Biopharmaceuticals, Inc, South San Francisco, CA, USA
| |
Collapse
|
42
|
Franklin ZJ, O’Harte FP, Irwin N. Effects of short-term chemical ablation of glucagon signalling by peptide-based glucagon receptor antagonists on insulin secretion and glucose homeostasis in mice. Biol Chem 2014; 395:433-42. [DOI: 10.1515/hsz-2013-0224] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 11/04/2013] [Indexed: 11/15/2022]
Abstract
Abstract
Glucagon is a hormone with important effects on blood glucose regulation. This study has utilized the stable glucagon receptor antagonists, desHis1Pro4Glu9-glucagon and desHis1Pro4Glu9(Lys12PAL)-glucagon, to evaluate the effects of sustained inhibition of glucagon receptor signalling in normal mice. Twice-daily injection of either analogue for 10 days had no effect on food intake, body weight and non-fasting plasma glucose concentrations. However, insulin levels were significantly raised (p<0.05 to p<0.01) from day 3 onwards in desHis1Pro4Glu9-glucagon mice. After 10 days, glucose tolerance was improved (p<0.05) in desHis1Pro4Glu9-glucagon treated mice. Glucose-mediated insulin secretion and circulating cholesterol levels were significantly (p<0.05 to p<0.01) decreased in both treatment groups. Importantly, the effects of glucagon to increase blood glucose and insulin concentrations were still annulled on day 10. Insulin sensitivity was almost identical in all groups of mice at the end of the study. In addition, no changes in pancreatic insulin and glucagon content or islet morphology were observed in either treatment group. Finally, acute injection of desHis1Pro4Glu9-glucagon followed by a 24-h fast in treatment naïve mice was not associated with any hypoglycaemic episodes. These data indicate that peptide-based glucagon receptor antagonists represent safe and effective treatment options for type 2 diabetes.
Collapse
|
43
|
O'Harte FPM, Franklin ZJ, Rafferty EP, Irwin N. Characterisation of structurally modified analogues of glucagon as potential glucagon receptor antagonists. Mol Cell Endocrinol 2013; 381:26-34. [PMID: 23891841 DOI: 10.1016/j.mce.2013.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 06/12/2013] [Accepted: 07/16/2013] [Indexed: 01/16/2023]
Abstract
Acute in vitro and in vivo biological activities of four novel structural analogues of glucagon were tested. desHis(1)Pro(4)-glucagon, desHis(1)Pro(4)Glu(9)-glucagon, desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon and desHis(1)Pro(4)Glu(9)Lys(30)FA-glucagon were stable to DPP-4 degradation and dose-dependently inhibited glucagon-mediated cAMP production (p<0.05 to p<0.001). None stimulated insulin secretion in vitro above basal levels, but all inhibited glucagon-induced insulin secretion (p<0.01 to p<0.001). In normal mice all analogues antagonised acute glucagon-mediated elevations of blood glucose (p<0.05 to p<0.001) and blocked corresponding insulinotropic responses. In high-fat fed mice, glucagon-induced increases in plasma insulin (p<0.05 to p<0.001) and glucagon-induced hyperglycaemia were blocked (p<0.05 to p<0.01) by three analogues. In obese diabetic (ob/ob) mice only desHis(1)Pro(4)Glu(9)-glucagon effectively (p<0.05 to p<0.01) inhibited both glucagon-mediated glycaemic and insulinotropic responses. desHis(1)Pro(4)-glucagon and desHis(1)Pro(4)Glu(9)-glucagon were biologically ineffective when administered 8h prior to glucagon, whereas desHis(1)Pro(4)Glu(9)Lys(12)FA-glucagon retained efficacy (p<0.01) for up to 24h. Such peptide-derived glucagon receptor antagonists have potential for type 2 diabetes therapy.
Collapse
Affiliation(s)
- F P M O'Harte
- The Saad Centre for Pharmacy & Diabetes, School of Biomedical Sciences, University of Ulster, Coleraine, Co. Londonderry BT52 1SA, Northern Ireland, United Kingdom.
| | | | | | | |
Collapse
|
44
|
Erion DM, Kotas ME, McGlashon J, Yonemitsu S, Hsiao JJ, Nagai Y, Iwasaki T, Murray SF, Bhanot S, Cline GW, Samuel VT, Shulman GI, Gillum MP. cAMP-responsive element-binding protein (CREB)-regulated transcription coactivator 2 (CRTC2) promotes glucagon clearance and hepatic amino acid catabolism to regulate glucose homeostasis. J Biol Chem 2013; 288:16167-76. [PMID: 23595987 DOI: 10.1074/jbc.m113.460246] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
cAMP-responsive element-binding protein (CREB)-regulated transcription coactivator 2 (CRTC2) regulates transcription of gluconeogenic genes by specifying targets for the transcription factor CREB in response to glucagon. We used an antisense oligonucleotide directed against CRTC2 in both normal rodents and in rodent models of increased gluconeogenesis to better understand the role of CRTC2 in metabolic disease. In the context of severe hyperglycemia and elevated hepatic glucose production, CTRC2 knockdown (KD) improved glucose homeostasis by reducing endogenous glucose production. Interestingly, despite the known role of CRTC2 in coordinating gluconeogenic gene expression, CRTC2 KD in a rodent model of type 2 diabetes resulted in surprisingly little alteration of glucose production. However, CRTC2 KD animals had elevated circulating concentrations of glucagon and a ∼80% reduction in glucagon clearance. When this phenomenon was prevented with somatostatin or a glucagon-neutralizing antibody, endogenous glucose production was reduced by CRTC2 KD. Additionally, CRTC2 inhibition resulted in reduced expression of several glucagon-induced pyridoxal 5'-phosphate-dependent enzymes that convert amino acids to gluconeogenic intermediates, suggesting that it may control substrate availability as well as gluconeogenic gene expression. CRTC2 is an important regulator of gluconeogenesis with tremendous impact in models of elevated hepatic glucose production. Surprisingly, it is also part of a previously unidentified negative feedback loop that degrades glucagon and regulates amino acid metabolism to coordinately control glucose homeostasis in vivo.
Collapse
Affiliation(s)
- Derek M Erion
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06536, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|