151
|
Ojha A, Ojha U, Mohammed R, Chandrashekar A, Ojha H. Current perspective on the role of insulin and glucagon in the pathogenesis and treatment of type 2 diabetes mellitus. Clin Pharmacol 2019; 11:57-65. [PMID: 31191043 PMCID: PMC6515536 DOI: 10.2147/cpaa.s202614] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/28/2019] [Indexed: 01/17/2023] Open
Abstract
According to the World Health Organization, 422 million adults worldwide live with diabetes mellitus (DM), a significant portion of whom have type 2 diabetes. The discovery of insulin as a key regulator of glucose metabolism has revolutionized our understanding of DM and provided several therapeutic avenues. Most studies have so far predominantly focused on the role of insulin in type 2 diabetes. However, the balance between insulin and glucagon is essential in ensuring glucose homeostasis. In this review, we begin by evaluating the principal differences between insulin and glucagon with regard to their mechanism and control of their secretion. Next, we discuss their mode of action and effects on metabolism. We further explore how the two hormones impact the natural history of type 2 diabetes. Finally, we outline how current and emerging pharmacological agents attempt to exploit the properties of insulin and glucagon to benefit patients with type 2 diabetes.
Collapse
Affiliation(s)
- Ashutosh Ojha
- Shobhaben Pratapbhai Patel School Of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Mumbai, India
| | - Utkarsh Ojha
- Faculty of Medicine, Imperial College London, London, UK
| | - Raihan Mohammed
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Harsh Ojha
- Department of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
152
|
Alexiadou K, Anyiam O, Tan T. Cracking the combination: Gut hormones for the treatment of obesity and diabetes. J Neuroendocrinol 2019; 31:e12664. [PMID: 30466162 PMCID: PMC6563152 DOI: 10.1111/jne.12664] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/11/2018] [Accepted: 11/17/2018] [Indexed: 12/21/2022]
Abstract
Obesity and type 2 diabetes are a veritable global pandemic. There is an imperative to develop new therapies for these conditions that can be delivered at scale to patients, which deliver effective and titratable weight loss, amelioration of diabetes, prevention of diabetic complications and improvements in cardiovascular health. Although agents based on glucagon-like peptide-1 (GLP-1) are now in routine use for diabetes and obesity, the limited efficacy of such drugs means that newer agents are required. By combining the effects of GLP-1 with other gut and metabolic hormones such as glucagon (GCG), oxyntomodulin, glucose-dependent insulinotropic peptide (GIP) and peptide YY (PYY), we may obtain improved weight loss, increased energy expenditure and improved metabolic profiles. Drugs based on dual agonism of GLP1R/GCGR and GLP1R/GIPR are being actively developed in clinical trials. Triple agonism, for example with GLPR1/GCGR/GIPR unimolecular agonists or using GLP-1/oxyntomodulin/PYY, is also being explored. Multi-agonist drugs seem set to deliver the next generation of therapies for diabetes and obesity soon.
Collapse
Affiliation(s)
| | - Oluwaseun Anyiam
- Section of Investigative MedicineImperial College LondonLondonUK
| | - Tricia Tan
- Section of Investigative MedicineImperial College LondonLondonUK
| |
Collapse
|
153
|
Camps-Bossacoma M, Garcia-Aloy M, Saldaña-Ruiz S, Cambras T, González-Domínguez R, Franch À, Pérez-Cano FJ, Andres-Lacueva C, Castell M. Role of Theobromine in Cocoa's Metabolic Properties in Healthy Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3605-3614. [PMID: 30855143 DOI: 10.1021/acs.jafc.8b07248] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cocoa is rich in polyphenols and methylxanthines, and it has been reported that its consumption, among other properties, has beneficial effects on metabolism. This study aimed to investigate the role of theobromine in cocoa's metabolic properties in healthy rats. In addition to morphometric measurements, biochemical markers of lipids and glucose metabolism and gene expression of molecules related to immune cells in adipose and hepatic tissues were assessed after 7 or 18 days of diet. Additionally, a metabolomic analysis was carried out at day 7. This study revealed the presence of six discriminant metabolites in plasma due to the diets. Moreover, the results showed that theobromine is the main responsible factor for cocoa's effects on body weight gain as well as on lipid and glucose metabolism. The effects on body weight and lipids appeared as early as after 7 days of diet, whereas those affecting glucose metabolism required a longer intervention.
Collapse
Affiliation(s)
- Mariona Camps-Bossacoma
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació; Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB) , Universitat de Barcelona (UB) , Barcelona 08028 , Spain
| | - Mar Garcia-Aloy
- Biomarkers and Nutrimetabolomics Laboratory, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, XaRTA, INSA-UB, Facultat de Farmàcia i Ciències de l'Alimentació, UB, Barcelona; CIBER de Fragilidad y Envejecimiento (CIBERFES) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Sandra Saldaña-Ruiz
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació; Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB) , Universitat de Barcelona (UB) , Barcelona 08028 , Spain
| | - Trinitat Cambras
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació; Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB) , Universitat de Barcelona (UB) , Barcelona 08028 , Spain
| | - Raúl González-Domínguez
- Biomarkers and Nutrimetabolomics Laboratory, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, XaRTA, INSA-UB, Facultat de Farmàcia i Ciències de l'Alimentació, UB, Barcelona; CIBER de Fragilidad y Envejecimiento (CIBERFES) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Àngels Franch
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació; Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB) , Universitat de Barcelona (UB) , Barcelona 08028 , Spain
| | - Francisco J Pérez-Cano
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació; Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB) , Universitat de Barcelona (UB) , Barcelona 08028 , Spain
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, XaRTA, INSA-UB, Facultat de Farmàcia i Ciències de l'Alimentació, UB, Barcelona; CIBER de Fragilidad y Envejecimiento (CIBERFES) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Margarida Castell
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació; Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB) , Universitat de Barcelona (UB) , Barcelona 08028 , Spain
| |
Collapse
|
154
|
Jung CH, Mok JO. The Effects of Hypoglycemic Agents on Non-alcoholic Fatty Liver Disease: Focused on Sodium-Glucose Cotransporter 2 Inhibitors and Glucagon-Like Peptide-1 Receptor Agonists. J Obes Metab Syndr 2019; 28:18-29. [PMID: 31089576 PMCID: PMC6484938 DOI: 10.7570/jomes.2019.28.1.18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/24/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023] Open
Abstract
The only known, effective intervention for non-alcoholic fatty liver disease (NAFLD) is weight loss, and there is no approved pharmacotherapy. Recently, new hypoglycemic agents, such as sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide-1 receptor agonists (GLP-1RAs), and their effects on NAFLD have received substantial interest. Herein, we review the currently available human studies regarding the effects of SGLT2 inhibitors and GLP-1RAs on NAFLD/non-alcoholic steatohepatitis in patients with type 2 diabetes mellitus, and we describe the possible mechanisms explaining the positive effects of these agents on NAFLD.
Collapse
Affiliation(s)
- Chan-Hee Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Ji-Oh Mok
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| |
Collapse
|
155
|
Georgia A, Asnis MCC, Febres G, Tsang A, Bessler M, Korner J. Roux-en-Y Gastric Bypass Is Associated With Hyperinsulinemia But Not Increased Maximal β-Cell Function. J Endocr Soc 2019; 3:632-642. [PMID: 30834358 PMCID: PMC6391719 DOI: 10.1210/js.2018-00213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/18/2019] [Indexed: 12/29/2022] Open
Abstract
Context Roux-en-Y gastric bypass (RYGB) is associated with postprandial hyperinsulinemia. Objective This study assessed whether increased blood insulin levels may be due to an increase in maximal β-cell function. Design, Setting, and Participants We performed a cross-sectional study at Columbia University Medical Center, New York, New York. Subjects without a history of diabetes were studied after surgery (n = 12) and were compared with nonsurgical controls (n = 10) who were mean matched for body mass index, insulin sensitivity, and hemoglobin A1c and with nonobese controls (n = 8). Methods Subjects underwent a mixed-meal tolerance test and on a separate day an intravenous glucose tolerance test followed by a hyperglycemic clamp (450 mg/dL; 25 mM blood glucose) and arginine stimulation. The main outcome measure was maximal insulin secretion quantified after arginine stimulation (AinsRmax). Results The RYGB group exhibited greater peak postprandial glucose levels and fourfold greater peak insulin levels than control groups; however, there were no significant differences in insulinogenic index or AinsRmax. Another finding was significantly greater postprandial glucagon levels in the RYGB group compared with controls. Conclusions Our results suggest that after RYGB, the increase in postprandial levels of insulin are not due to changes in maximal β-cell function but appear to be an appropriate response to altered nutrient flow and absorption.
Collapse
Affiliation(s)
- Annette Georgia
- Columbia University College of Physicians and Surgeons, New York, New York.,Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York
| | - Maria Cecilia Catilo Asnis
- Columbia University College of Physicians and Surgeons, New York, New York.,Stamford Health Medical Group, Stamford, Connecticut
| | - Gerardo Febres
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Amanda Tsang
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Marc Bessler
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Judith Korner
- Columbia University College of Physicians and Surgeons, New York, New York
| |
Collapse
|
156
|
Moh Moh MA, Jung CH, Lee B, Choi D, Kim BY, Kim CH, Kang SK, Mok JO. Association of glucagon-to-insulin ratio and nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. Diab Vasc Dis Res 2019; 16:186-195. [PMID: 30428692 DOI: 10.1177/1479164118810691] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE The aim of this study is to investigate the association between glucagon-to-insulin ratio and the presence of nonalcoholic fatty liver disease on ultrasonography in participants with type 2 diabetes mellitus. RESEARCH DESIGN AND METHODS This cross-sectional study was performed with data obtained from 172 participants with type 2 diabetes mellitus admitted to a University hospital of Korea. Participants were assessed for serum fasting and postprandial serum glucagon-to-insulin ratio and divided into tertiles. Nonalcoholic fatty liver disease was defined as ultrasonographically detected fatty liver. RESULTS Prevalence of nonalcoholic fatty liver disease was significantly decreased across tertile of fasting and postprandial glucagon-to-insulin ratio ( p = 0.009 for trend, p = 0.001 for trend, respectively). Lower glucagon-to-insulin ratio was significantly associated with the presence of nonalcoholic fatty liver disease even after adjustment for potential confounding variables [fasting glucagon-to-insulin ratio: odds ratio (95% confidence interval), 2.68 (1.08-6.86)], postprandial glucagon-to-insulin ratio: [2.72 (1.03-7.35)]. The participants in the lowest tertile of fasting glucagon-to-insulin ratio had higher body mass index, visceral fat thickness, subcutaneous fat thickness, homeostasis model assessment-insulin resistance and shorter duration of diabetes mellitus. CONCLUSION This study suggests that lower glucagon relative insulin may be independently associated with nonalcoholic fatty liver disease in participants with type 2 diabetes.
Collapse
Affiliation(s)
- Myint Aung Moh Moh
- 1 Department of Medicine, University of Medicine 2, Yangon, Yangon, Myanmar
| | - Chan-Hee Jung
- 2 Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Bora Lee
- 3 Department of Biostatistics, Soonchunhyang University College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Dughyun Choi
- 2 Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Bo-Yeon Kim
- 2 Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Chul-Hee Kim
- 2 Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Sung-Koo Kang
- 2 Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Ji-Oh Mok
- 2 Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| |
Collapse
|
157
|
Quiñones M, Fernø J, Diéguez C, Nogueiras R, Al-Massadi O. Exciting advances in GPCR-based drugs discovery for treating metabolic disease and future perspectives. Expert Opin Drug Discov 2019; 14:421-431. [PMID: 30821530 DOI: 10.1080/17460441.2019.1583642] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Current pharmacological therapies that target single receptors have limited efficacy for the treatment of diabetes and obesity. Novel approaches with hybrid peptides that activate more than one receptor at once to generate beneficial effects through synergistic effects have shown promising results. Several unimolecular dual and tri-agonists, mainly associated with GPCR like GLP-1/GCG/GIP receptors, have shown exceptional efficacy in preclinical models, and are currently being evaluated in clinical trials to investigate their safety and beneficial effects in humans. Areas covered: Herein, the authors review the development of drugs used in the treatment of metabolic disease, from single agonists to the new generation of tri-agonist peptides and compile the latest knowledge available on GPCR-based drug discovery. The authors also provide the reader with their expert perspectives on this exciting area of drug development. Expert opinion: The co-agonists that have been clinically tested so far have been well tolerated and reduce body weight as well as fasting glucose levels in patients with Type 2 Diabetes Mellitus to a higher degree than single agonists alone. The promising data collected so far now warrant large scale randomized clinical trials to assess whether a unimolecular polypharmacy-based approach could translate into safe and efficacious treatments for obesity and its comorbidities.
Collapse
Affiliation(s)
- Mar Quiñones
- a Department of Physiology, CIMUS , University of Santiago de Compostela-Instituto de Investigación Sanitaria , Santiago de Compostela , Spain.,b CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) , Madrid , Spain
| | - Johan Fernø
- c Hormone Laboratory , Haukeland University Hospital , Bergen , Norway
| | - Carlos Diéguez
- a Department of Physiology, CIMUS , University of Santiago de Compostela-Instituto de Investigación Sanitaria , Santiago de Compostela , Spain.,b CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) , Madrid , Spain
| | - Ruben Nogueiras
- a Department of Physiology, CIMUS , University of Santiago de Compostela-Instituto de Investigación Sanitaria , Santiago de Compostela , Spain.,b CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) , Madrid , Spain
| | - Omar Al-Massadi
- d Neurotransmission et signalisation laboratoire , Inserm UMR-S 839 , Paris , France.,e Faculté des Sciences et d'Ingénierie , Sorbonne Université , Paris , France.,f Institut du Fer a Moulin , Paris , France
| |
Collapse
|
158
|
Beaudry JL, Kaur KD, Varin EM, Baggio LL, Cao X, Mulvihill EE, Stern JH, Campbell JE, Scherer PE, Drucker DJ. The brown adipose tissue glucagon receptor is functional but not essential for control of energy homeostasis in mice. Mol Metab 2019; 22:37-48. [PMID: 30772257 PMCID: PMC6437632 DOI: 10.1016/j.molmet.2019.01.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 02/02/2023] Open
Abstract
Objective Administration of glucagon (GCG) or GCG-containing co-agonists reduces body weight and increases energy expenditure. These actions appear to be transduced by multiple direct and indirect GCG receptor (GCGR)-dependent mechanisms. Although the canonical GCGR is expressed in brown adipose tissue (BAT) the importance of BAT GCGR activity for the physiological control of body weight, or the response to GCG agonism, has not been defined. Methods We studied the mechanisms linking GCG action to acute increases in oxygen consumption using wildtype (WT), Ucp1−/− and Fgf21−/− mice. The importance of basal GCGR expression within the Myf5+ domain for control of body weight, adiposity, glucose and lipid metabolism, food intake, and energy expenditure was examined in GcgrBAT−/− mice housed at room temperature or 4 °C, fed a regular chow diet (RCD) or after a prolonged exposure to high fat diet (HFD). Results Acute GCG administration induced lipolysis and increased the expression of thermogenic genes in BAT cells, whereas knockdown of Gcgr reduced expression of genes related to thermogenesis. GCG increased energy expenditure (measured by oxygen consumption) both in vivo in WT mice and ex vivo in BAT and liver explants. GCG also increased acute energy expenditure in Ucp1−/− mice, but these actions were partially blunted in Ffg21−/− mice. However, acute GCG administration also robustly increased oxygen consumption in GcgrBAT−/− mice. Moreover, body weight, glycemia, lipid metabolism, body temperature, food intake, activity, energy expenditure and adipose tissue gene expression profiles were normal in GcgrBAT−/− mice, either on RCD or HFD, whether studied at room temperature, or chronically housed at 4 °C. Conclusions Exogenous GCG increases oxygen consumption in mice, also evident both in liver and BAT explants ex vivo, through UCP1-independent, FGF21-dependent pathways. Nevertheless, GCGR signaling within BAT is not physiologically essential for control of body weight, whole body energy expenditure, glucose homeostasis, or the adaptive metabolic response to cold or prolonged exposure to an energy dense diet. A functional glucagon receptor is expressed in brown adipose tissue and BAT cells. Glucagon increases energy expenditure in mice, as well as in liver and BAT. Glucagon increases whole body energy expenditure through FGF21-dependent and BAT glucagon receptor-independent pathways. Loss of the BAT glucagon receptor does not impair glucose or energy homeostasis in mice.
Collapse
Affiliation(s)
- Jacqueline L Beaudry
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Kiran Deep Kaur
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Elodie M Varin
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Laurie L Baggio
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Xiemin Cao
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Erin E Mulvihill
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Jennifer H Stern
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Jonathan E Campbell
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Phillip E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, USA
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
159
|
Stereochemical inversion as a route to improved biophysical properties of therapeutic peptides exemplified by glucagon. Commun Chem 2019. [DOI: 10.1038/s42004-018-0100-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
160
|
Papalou O, Kandaraki EA, Papadakis G, Diamanti-Kandarakis E. Endocrine Disrupting Chemicals: An Occult Mediator of Metabolic Disease. Front Endocrinol (Lausanne) 2019; 10:112. [PMID: 30881345 PMCID: PMC6406073 DOI: 10.3389/fendo.2019.00112] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs), a heterogeneous group of exogenous chemicals that can interfere with any aspect of endogenous hormones, represent an emerging global threat for human metabolism. There is now considerable evidence that the observed upsurge of metabolic disease cannot be fully attributed to increased caloric intake, physical inactivity, sleep deficit, and ageing. Among environmental factors implicated in the global deterioration of metabolic health, EDCs have drawn the biggest attention of scientific community, and not unjustifiably. EDCs unleash a coordinated attack toward multiple components of human metabolism, including crucial, metabolically-active organs such as hypothalamus, adipose tissue, pancreatic beta cells, skeletal muscle, and liver. Specifically, EDCs' impact during critical developmental windows can promote the disruption of individual or multiple systems involved in metabolism, via inducing epigenetic changes that can permanently alter the epigenome in the germline, enabling changes to be transmitted to the subsequent generations. The clear effect of this multifaceted attack is the manifestation of metabolic disease, clinically expressed as obesity, metabolic syndrome, diabetes mellitus, and non-alcoholic fatty liver disease. Although limitations of EDCs research do exist, there is no doubt that EDCs constitute a crucial parameter of the global deterioration of metabolic health we currently encounter.
Collapse
Affiliation(s)
- Olga Papalou
- Department of Endocrinology & Diabetes, Hygeia Hospital, Athens, Greece
| | | | | | - Evanthia Diamanti-Kandarakis
- Department of Endocrinology & Diabetes, Hygeia Hospital, Athens, Greece
- *Correspondence: Evanthia Diamanti-Kandarakis
| |
Collapse
|
161
|
Khare P, Mangal P, Baboota RK, Jagtap S, Kumar V, Singh DP, Boparai RK, Sharma SS, Khardori R, Bhadada SK, Kondepudi KK, Chopra K, Bishnoi M. Involvement of Glucagon in Preventive Effect of Menthol Against High Fat Diet Induced Obesity in Mice. Front Pharmacol 2018; 9:1244. [PMID: 30505271 PMCID: PMC6250823 DOI: 10.3389/fphar.2018.01244] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/12/2018] [Indexed: 11/13/2022] Open
Abstract
Glucagon mediated mechanisms have been shown to play clinically significant role in energy expenditure. The present study was designed to understand whether pharmacological mimicking of cold using menthol (TRPM8 modulator) can induce glucagon-mediated energy expenditure to prevent weight gain and related complications. Acute oral and topical administration of TRPM8 agonists (menthol and icilin) increased serum glucagon concentration which was prevented by pre-treatment with AMTB, a TRPM8 blocker. Chronic administration of menthol (50 and 100 mg/kg/day for 12 weeks) to HFD fed animals prevented weight gain, insulin resistance, adipose tissue hypertrophy and triacylglycerol deposition in liver. These effects were not restricted to oral administration, but also observed upon the topical application of menthol (10% w/v). The metabolic alterations caused by menthol in liver and adipose tissue mirrored the known effects of glucagon, such as increased glycogenolysis and gluconeogenesis in the liver, and enhanced thermogenic activity of white and brown adipose tissue. Correlation analysis suggests a strong correlation between glucagon dependent changes and energy expenditure markers. Interestingly, in-vitro treatment of the serum of menthol treated mice increased energy expenditure markers in mature 3T3L1 adipocytes, which was prevented in the presence of non-competitive glucagon receptor antagonist, L-168,049, indicating that menthol-induced increase in serum glucagon is responsible for increase in energy expenditure phenotype. In conclusion, the present work provides evidence that glucagon plays an important role in the preventive effect of menthol against HFD-induced weight gain and related complications.
Collapse
Affiliation(s)
- Pragyanshu Khare
- National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar, India.,Department of Pharmacology, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Priyanka Mangal
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Ritesh K Baboota
- National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar, India
| | - Sneha Jagtap
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Vijay Kumar
- National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar, India
| | | | - Ravneet K Boparai
- Department of Biotechnology, Government College for Girls, Chandigarh, India
| | - Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar, India
| | - Romesh Khardori
- Division of Endocrinology and Metabolism, The EVMS Sterling Centre of Diabetes and Endocrine Disorders, Department of Internal Medicine, East Virginia Medical School, Norfolk, VA, United States
| | - Sanjay K Bhadada
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Kanthi K Kondepudi
- National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar, India
| | - Kanwaljit Chopra
- Department of Pharmacology, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar, India
| |
Collapse
|
162
|
Rivero-Gutierrez B, Haller A, Holland J, Yates E, Khrisna R, Habegger K, Dimarchi R, D'Alessio D, Perez-Tilve D. Deletion of the glucagon receptor gene before and after experimental diabetes reveals differential protection from hyperglycemia. Mol Metab 2018; 17:28-38. [PMID: 30170980 PMCID: PMC6197675 DOI: 10.1016/j.molmet.2018.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Mice with congenital loss of the glucagon receptor gene (Gcgr-/- mice) remain normoglycemic in insulinopenic conditions, suggesting that unopposed glucagon action is the driving force for hyperglycemia in Type-1 Diabetes Mellitus (T1DM). However, chronic loss of GCGR results in a neomorphic phenotype that includes hormonal signals with hypoglycemic activity. We combined temporally-controlled GCGR deletion with pharmacological treatments to dissect the direct contribution of GCGR signaling to glucose control in a common mouse model of T1DM. METHODS We induced experimental T1DM by injecting the beta-cell cytotoxin streptozotocin (STZ) in mice with congenital or temporally-controlled Gcgr loss-of-function using tamoxifen (TMX). RESULTS Disruption of Gcgr expression, using either an inducible approach in adult mice or animals with congenital knockout, abolished the response to a long-acting Gcgr agonist. Mice with either developmental Gcgr disruption or inducible deletion several weeks before STZ treatment maintained normoglycemia. However, mice with inducible knockout of the Gcgr one week after the onset of STZ diabetes had only partial correction of hyperglycemia, an effect that was reversed by GLP-1 receptor blockade. Mice with Gcgr deletion for either 2 or 6 weeks had similar patterns of gene expression, although the changes were generally larger with longer GCGR knockout. CONCLUSIONS These findings demonstrate that the effects of glucagon to mitigate diabetic hyperglycemia are not through acute signaling but require compensations that take weeks to develop.
Collapse
Affiliation(s)
- Belen Rivero-Gutierrez
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - April Haller
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - Jenna Holland
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - Emily Yates
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - Radha Khrisna
- Department of Medicine, Duke University School of Medicine, NC, USA
| | - Kirk Habegger
- Comprehensive Diabetes Center and Department of Medicine - Endocrinology, Diabetes & and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard Dimarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - David D'Alessio
- Department of Medicine, Duke University School of Medicine, NC, USA
| | - Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA.
| |
Collapse
|
163
|
Kim T, Holleman CL, Nason S, Arble DM, Ottaway N, Chabenne J, Loyd C, Kim JA, Sandoval D, Drucker DJ, DiMarchi R, Perez-Tilve D, Habegger KM. Hepatic Glucagon Receptor Signaling Enhances Insulin-Stimulated Glucose Disposal in Rodents. Diabetes 2018; 67:2157-2166. [PMID: 30150304 PMCID: PMC6198333 DOI: 10.2337/db18-0068] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022]
Abstract
Glucagon receptor (GCGR) agonists cause hyperglycemia but also weight loss. However, GCG-like peptide 1 receptor (GLP1R)/GCGR mixed agonists do not exhibit the diabetogenic effects often attributed to GCGR activity. Thus, we sought to investigate the effect of glucagon agonism on insulin action and glucose homeostasis. Acute GCGR agonism induced immediate hyperglycemia, followed by improved glucose tolerance and enhanced glucose-stimulated insulin secretion. Moreover, acute GCGR agonism improved insulin tolerance in a dose-dependent manner in both lean and obese mice. Improved insulin tolerance was independent of GLP1R, FGF21, and hepatic glycogenolysis. Moreover, we observed increased glucose infusion rate, disposal, uptake, and suppressed endogenous glucose production during euglycemic clamps. Mice treated with insulin and GCGR agonist had enhanced phosphorylation of hepatic AKT at Ser473; this effect was reproduced in isolated mouse primary hepatocytes and resulted in increased AKT kinase activity. These data reveal that GCGR agonism enhances glucose tolerance, in part, by augmenting insulin action, with implications for the use of GCGR agonism in therapeutic strategies for diabetes.
Collapse
Affiliation(s)
- Teayoun Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Cassie L Holleman
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Shelly Nason
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Deanna M Arble
- Department of Biological Sciences, Marquette University, Milwaukee, WI
| | - Nickki Ottaway
- Metabolic Diseases Institute and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, OH
| | | | - Christine Loyd
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Jeong-A Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Richard DiMarchi
- Novo Nordisk Research Center, Indianapolis, IN
- Department of Chemistry, Indiana University, Bloomington, IN
| | - Diego Perez-Tilve
- Metabolic Diseases Institute and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, OH
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
164
|
Kazierad DJ, Chidsey K, Somayaji VR, Bergman AJ, Calle RA. Efficacy and safety of the glucagon receptor antagonist PF-06291874: A 12-week, randomized, dose-response study in patients with type 2 diabetes mellitus on background metformin therapy. Diabetes Obes Metab 2018; 20:2608-2616. [PMID: 29923286 DOI: 10.1111/dom.13440] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 12/14/2022]
Abstract
AIMS To conduct a dose-response assessment of the efficacy and safety of the glucagon receptor antagonist PF-06291874 in adults with type 2 diabetes (T2DM) using stable doses of metformin. MATERIALS AND METHODS This randomized, double-blind, statin-stratified, placebo-controlled, 4-arm, parallel-group study was conducted in patients with T2DM who were receiving background metformin. After an 8-week, non-metformin oral antidiabetic agent washout period, 206 patients were randomized to placebo or PF-06291874 (30, 60 or 100 mg once daily) for 12 weeks. Glycosylated haemoglobin (HbA1c), fasting plasma glucose (FPG) and safety endpoints were assessed at baseline and post baseline. RESULTS Dose-dependent mean reductions from baseline in HbA1c for PF-06291874 ranged from -0.67% (-7.29 mmol/mol) to -0.93% (-10.13 mmol/mol), and for FPG from -16.6 to -33.3 mg/dL after 12 weeks of dosing. The incidence of hypoglycaemia was low and was similar between groups receiving PF-06291874 and placebo. Small, non-dose-dependent increases in LDL cholesterol (<10%) and blood pressure (BP) (systolic BP > 2 mm Hg; diastolic BP > 1 mm Hg) were observed with PF-06291874. Modest non-dose-dependent median increases were observed across PF-06291874 groups at 12 weeks for alanine aminotransferase (range, 37.6-48.7 U/L vs placebo) and aspartate aminotransferase (range, 33.3-36.6 U/L vs placebo); these were not associated with bilirubin changes. Small increases were observed in body weight (< 0.5 kg) in each PF-06291874 group vs placebo. CONCLUSIONS In patients with T2DM, PF-06291874 significantly lowered HbA1c and glucose, was well tolerated and carried a low risk of hypoglycaemia. Small, non-dose-related increases in BP, lipids and hepatic transaminases were observed.
Collapse
Affiliation(s)
- David J Kazierad
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Kristin Chidsey
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Veena R Somayaji
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Arthur J Bergman
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Roberto A Calle
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| |
Collapse
|
165
|
The first pediatric case of glucagon receptor defect due to biallelic mutations in GCGR is identified by newborn screening of elevated arginine. Mol Genet Metab Rep 2018; 17:46-52. [PMID: 30294546 PMCID: PMC6171159 DOI: 10.1016/j.ymgmr.2018.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022] Open
Abstract
Glucagon receptor (GCGR) defect (Mahvash disease) is an autosomal recessive hereditary pancreatic neuroendocrine tumor (PNET) syndrome that has only been reported in adults with pancreatic α cell hyperplasia and PNETs. We describe a 7-year-old girl with persistent hyperaminoacidemia, notable for elevations of glutamine (normal ammonia), alanine (normal lactate), dibasic amino acids (arginine, lysine and ornithine), threonine and serine. She initially was brought to medical attention by an elevated arginine on newborn screening (NBS) and treated for presumed arginase deficiency with a low protein diet, essential amino acids formula and an ammonia scavenger drug. This treatment normalized plasma amino acids. She had intermittent emesis and anorexia, but was intellectually normal. Arginase enzyme assay and ARG1 sequencing and deletion/duplication analysis were normal. Treatments were stopped, but similar pattern of hyperaminoacidemia recurred. She also had hypercholesterolemia type IIa, with only elevated LDL cholesterol, despite an extremely lean body habitus. Exome sequencing was initially non-diagnostic. Through a literature search, we recognized the pattern of hyperaminoacidemia was strikingly similar to that reported in the Gcgr−/− knockout mice. Subsequently the patient was found to have an extremely elevated plasma glucagon and a novel, homozygous c.958_960del (p.Phe320del) variant in GCGR. Functional studies confirmed the pathogenicity of this variant. This case expands the clinical phenotype of GCGR defect in children and emphasizes the clinical utility of plasma amino acids in screening, diagnosis and monitoring glucagon signaling interruption. Early identification of a GCGR defect may provide an opportunity for potential beneficial treatment for an adult onset tumor predisposition disease. Describe the first case of glucagon receptor defect uniquely identified by abnormal newborn screening for elevated arginine. Characterize the pattern of hyperaminoacidemia in GCGR defect. Expand the clinical spectrum of GCGR defect from adult to childhood with a unique gastrointestinal manifestation.
Collapse
|
166
|
Sharma D, Verma S, Vaidya S, Kalia K, Tiwari V. Recent updates on GLP-1 agonists: Current advancements & challenges. Biomed Pharmacother 2018; 108:952-962. [PMID: 30372907 DOI: 10.1016/j.biopha.2018.08.088] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/04/2018] [Accepted: 08/15/2018] [Indexed: 12/16/2022] Open
Abstract
Glucagon-like peptide (GLP)-1 is an incretin hormone exhibiting several pharmacological actions such as neuroprotection, increased cognitive function, cardio-protection, decreased hypertension, suppression of acid secretion, increase in lyposis, and protection from inflammation. The most potent actions are glucose-dependent insulinotropic and glucagonostatic actions, stimulation of β-cell proliferation, enhanced insulin secretion and reduced weight gain in patients with type-2 diabetes pertaining to blood glucose control. Despite all these actions, its short half-life (around 2∼min) and degradation by a dipeptidyl peptidase-4 enzyme (DPP-4) limits the therapeutic utility of GLP1. In this review, we have discussed DPP IV-resistant analogs of GLP-1 currently present in clinical trials such as Exenatide, Liraglutide, Semaglutide, Efpeglenatide, Exenatide ER, Ittca 650 (Intarcia), Dulaglutide, Albiglutide, and Lixisenatide. Moreover, we have also discussed in detail the pharmacology, signaling mechanisms, and pharmacokinetic properties (Cmax, Tmax, T1/2, Vd, and Bioavailability) of DPP IV-resistant analogs of (GLP-1). Interestingly, GLP-1 agonist drugs have shown better potential to treat type-2 diabetes mellitus (T2DM) as compared to currently used drugs in clinics without causing the side effects of hypoglycemia and weight gain.
Collapse
Affiliation(s)
- Dilip Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Suril Verma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Shivani Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India.
| | - Vinod Tiwari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar 382355, Gujarat, India.
| |
Collapse
|
167
|
Loh K, Shi YC, Bensellam M, Lee K, Laybutt DR, Herzog H. Y1 receptor deficiency in β-cells leads to increased adiposity and impaired glucose metabolism. Sci Rep 2018; 8:11835. [PMID: 30177746 PMCID: PMC6120893 DOI: 10.1038/s41598-018-30140-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/24/2018] [Indexed: 01/12/2023] Open
Abstract
Insulin secretion from pancreatic β-cells is critical for maintaining glucose homeostasis and deregulation of circulating insulin levels is associated with the development of metabolic diseases. While many factors have been implicated in the stimulation of insulin secretion, the mechanisms that subsequently reduce insulin secretion remain largely unexplored. Here we demonstrate that mice with β-cell specific ablation of the Y1 receptor exhibit significantly upregulated serum insulin levels associated with increased body weight and adiposity. Interestingly, when challenged with a high fat diet these β-cell specific Y1-deficient mice also develop hyperglycaemia and impaired glucose tolerance. This is most likely due to enhanced hepatic lipid synthesis, resulting in an increase of lipid accumulation in the liver. Together, our study demonstrates that Y1 receptor signaling negatively regulates insulin release, and pharmacological inhibition of Y1 receptor signalling for the treatment of non-insulin dependent diabetes should be taken into careful consideration.
Collapse
Affiliation(s)
- Kim Loh
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010, Australia. .,Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia. .,St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia.
| | - Yan-Chuan Shi
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010, Australia.,Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - Mohammed Bensellam
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010, Australia
| | - Kailun Lee
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010, Australia.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010, Australia.,Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - D Ross Laybutt
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010, Australia.,Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, 2010, Australia. .,Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia.
| |
Collapse
|
168
|
Kim T, Nason S, Holleman C, Pepin M, Wilson L, Berryhill TF, Wende AR, Steele C, Young ME, Barnes S, Drucker DJ, Finan B, DiMarchi R, Perez-Tilve D, Tschöp M, Habegger KM. Glucagon Receptor Signaling Regulates Energy Metabolism via Hepatic Farnesoid X Receptor and Fibroblast Growth Factor 21. Diabetes 2018; 67:1773-1782. [PMID: 29925501 PMCID: PMC6110317 DOI: 10.2337/db17-1502] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Glucagon, an essential regulator of glucose and lipid metabolism, also promotes weight loss, in part through potentiation of fibroblast growth factor 21 (FGF21) secretion. However, FGF21 is only a partial mediator of metabolic actions ensuing from glucagon receptor (GCGR) activation, prompting us to search for additional pathways. Intriguingly, chronic GCGR agonism increases plasma bile acid levels. We hypothesized that GCGR agonism regulates energy metabolism, at least in part, through farnesoid X receptor (FXR). To test this hypothesis, we studied whole-body and liver-specific FXR-knockout (Fxr∆liver) mice. Chronic GCGR agonist (IUB288) administration in diet-induced obese (DIO) Gcgr, Fgf21, and Fxr whole-body or liver-specific knockout (∆liver) mice failed to reduce body weight when compared with wild-type (WT) mice. IUB288 increased energy expenditure and respiration in DIO WT mice, but not Fxr∆liver mice. GCGR agonism increased [14C]palmitate oxidation in hepatocytes isolated from WT mice in a dose-dependent manner, an effect blunted in hepatocytes from Fxr∆liver mice. Our data clearly demonstrate that control of whole-body energy expenditure by GCGR agonism requires intact FXR signaling in the liver. This heretofore-unappreciated aspect of glucagon biology has implications for the use of GCGR agonism in the therapy of metabolic disorders.
Collapse
MESH Headings
- Adiposity/drug effects
- Animals
- Anti-Obesity Agents/therapeutic use
- Calorimetry, Indirect
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Energy Metabolism/drug effects
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Gene Expression Regulation/drug effects
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Liver/drug effects
- Mitochondria, Liver/enzymology
- Mitochondria, Liver/metabolism
- Obesity/drug therapy
- Obesity/etiology
- Obesity/metabolism
- Obesity/pathology
- Organ Specificity
- Oxidative Phosphorylation/drug effects
- Peptides/therapeutic use
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Glucagon/agonists
- Receptors, Glucagon/genetics
- Receptors, Glucagon/metabolism
- Signal Transduction/drug effects
- Weight Gain/drug effects
Collapse
Affiliation(s)
- Teayoun Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Shelly Nason
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Cassie Holleman
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Mark Pepin
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, AL
| | - Landon Wilson
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Taylor F Berryhill
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, AL
| | - Chad Steele
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Stephen Barnes
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN
| | - Richard DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN
- Department of Chemistry, Indiana University, Bloomington, IN
| | - Diego Perez-Tilve
- Division of Endocrinology, Diabetes and Metabolism, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
169
|
Elvert R, Bossart M, Herling AW, Weiss T, Zhang B, Kannt A, Wagner M, Haack T, Evers A, Dudda A, Keil S, Lorenz M, Lorenz K, Riz M, Hennerici W, Larsen PJ. Team Players or Opponents: Coadministration of Selective Glucagon and GLP-1 Receptor Agonists in Obese Diabetic Monkeys. Endocrinology 2018; 159:3105-3119. [PMID: 29992313 DOI: 10.1210/en.2018-00399] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/03/2018] [Indexed: 01/06/2023]
Abstract
We assessed the therapeutic contribution of the individual components of glucagon-like peptide-1 receptor (GLP-1R) and glucagon receptor (GCGR) agonists alone and in combination upon energy homeostasis and glycemic control in diet-induced obese, diabetic nonhuman primates. The pharmacological active dose ranges of selective agonists were established through a dose-finding study, followed by a 6-week chronic study. Repeated subcutaneous administration of a selective GCGR agonist (30 µg/kg once daily) did not affect food intake or body weight, whereas the selective GLP-1R agonist (3 µg/kg once daily) alone decreased energy intake by 18% and body weight by 3.8% ± 0.9%. Combination of both agonists reduced significantly cumulative food intake by 27% and body weight by 6.6% ± 0.9%. Fasting plasma glucose (FPG) was improved by GLP-1R agonist (baseline vs end of study, 176.7 ± 34.0 vs 115.9 ± 16.1 mg/dL). In contrast, groups exposed to GCGR agonist experienced nonsignificant elevations of FPG. More accurate assessment of therapeutic interventions on glucose homeostasis was tested by an IV glucose tolerance test. Glucose excursion was significantly elevated by chronic GCGR agonist administration, whereas it was significantly decreased in GLP-1R agonist-treated monkeys. In the combination group, a nonsignificant increase of glucose excursion was seen, concomitantly with significantly increased insulin secretion. We conclude that chronic glucagon agonism does not affect energy homeostasis in nonhuman primates. In combination with GLP-1R agonism, glucagon agonism synergistically enhances negative energy balance with resulting larger body weight loss. However, adding GCGR to GLP-1R agonism diminishes glycemic control in diabetic monkeys. Therefore, long-term therapeutic implications of using GLP-1R/GCGR coagonists for weight management in diabetes warrants further scrutiny.
Collapse
Affiliation(s)
- Ralf Elvert
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Martin Bossart
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Andreas W Herling
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Tilo Weiss
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | | | - Aimo Kannt
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Wagner
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Torsten Haack
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Andreas Evers
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Angela Dudda
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Stefanie Keil
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Martin Lorenz
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Katrin Lorenz
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Michela Riz
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Wolfgang Hennerici
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| | - Philip J Larsen
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, German
| |
Collapse
|
170
|
Álvarez-Rodríguez M, Pereiro P, Reyes-López FE, Tort L, Figueras A, Novoa B. Analysis of the Long-Lived Responses Induced by Immunostimulants and Their Effects on a Viral Infection in Zebrafish ( Danio rerio). Front Immunol 2018; 9:1575. [PMID: 30038625 PMCID: PMC6047052 DOI: 10.3389/fimmu.2018.01575] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022] Open
Abstract
In recent years, the innate immune response has gained importance since evidence indicates that after an adequate priming protocol, it is possible to obtain some prolonged and enhanced immune responses. Nevertheless, several factors, such as the timing and method of administration of the immunostimulants, must be carefully considered. An inappropriate protocol can transform the treatments into a double-edged sword for the teleost immune system, resulting in a stressful and immunosuppressive state. In this work, we analyzed the long-term effects of different stimuli (β-glucans, lipopolysaccharide, and polyinosinic:polycytidylic acid) on the transcriptome modulation induced by Spring Viremia Carp Virus (SVCV) in adult zebrafish (Danio rerio) and on the mortality caused by this infection. At 35 days post-immunostimulation, the transcriptome was found to be highly altered compared to that of the control fish, and these stimuli also conditioned the response to SVCV challenge, especially in the case of β-glucans. No protection against SVCV was found with any of the stimuli, and non-significant higher mortalities were even observed, especially with β-glucans. However, in the short term (pre-stimulation with β-glucan and infection after 7 days), slight protection was observed after infection. The transcriptome response in the zebrafish kidney at 35 days posttreatment with β-glucans revealed a significant response associated with stress and immunosuppression. The identification of genes that were differentially expressed before and after the infection seemed to indicate a high energy cost of the immunostimulation that was prolonged over time and could explain the lack of protection against SVCV. Differential responses to stress and alterations in lipid metabolism, the tryptophan–kynurenine pathway, and interferon-gamma signaling seem to be some of the mechanisms involved in this response, which represents the end of trained immunity and the beginning of a stressful state characterized by immunosuppression.
Collapse
Affiliation(s)
| | - Patricia Pereiro
- Institute of Marine Research (IIM), National Research Council (CSIC), Vigo, Spain
| | - Felipe E Reyes-López
- Department of Cell Biology, Physiology and Immunology, Universidad Autónoma de Barcelona, Bellaterra, Spain
| | - Lluis Tort
- Department of Cell Biology, Physiology and Immunology, Universidad Autónoma de Barcelona, Bellaterra, Spain
| | - Antonio Figueras
- Institute of Marine Research (IIM), National Research Council (CSIC), Vigo, Spain
| | - Beatriz Novoa
- Institute of Marine Research (IIM), National Research Council (CSIC), Vigo, Spain
| |
Collapse
|
171
|
Seok S, Kim YC, Byun S, Choi S, Xiao Z, Iwamori N, Zhang Y, Wang C, Ma J, Ge K, Kemper B, Kemper JK. Fasting-induced JMJD3 histone demethylase epigenetically activates mitochondrial fatty acid β-oxidation. J Clin Invest 2018; 128:3144-3159. [PMID: 29911994 DOI: 10.1172/jci97736] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 04/24/2018] [Indexed: 12/21/2022] Open
Abstract
Jumonji D3 (JMJD3) histone demethylase epigenetically regulates development and differentiation, immunity, and tumorigenesis by demethylating a gene repression histone mark, H3K27-me3, but a role for JMJD3 in metabolic regulation has not been described. SIRT1 deacetylase maintains energy balance during fasting by directly activating both hepatic gluconeogenic and mitochondrial fatty acid β-oxidation genes, but the underlying epigenetic and gene-specific mechanisms remain unclear. In this study, JMJD3 was identified unexpectedly as a gene-specific transcriptional partner of SIRT1 and epigenetically activated mitochondrial β-oxidation, but not gluconeogenic, genes during fasting. Mechanistically, JMJD3, together with SIRT1 and the nuclear receptor PPARα, formed a positive autoregulatory loop upon fasting-activated PKA signaling and epigenetically activated β-oxidation-promoting genes, including Fgf21, Cpt1a, and Mcad. Liver-specific downregulation of JMJD3 resulted in intrinsic defects in β-oxidation, which contributed to hepatosteatosis as well as glucose and insulin intolerance. Remarkably, the lipid-lowering effects by JMJD3 or SIRT1 in diet-induced obese mice were mutually interdependent. JMJD3 histone demethylase may serve as an epigenetic drug target for obesity, hepatosteatosis, and type 2 diabetes that allows selective lowering of lipid levels without increasing glucose levels.
Collapse
Affiliation(s)
- Sunmi Seok
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Young-Chae Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sangwon Byun
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sunge Choi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Zhen Xiao
- Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Naoki Iwamori
- Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Yang Zhang
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Chaochen Wang
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Jian Ma
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Kai Ge
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Byron Kemper
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jongsook Kim Kemper
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
172
|
Patel VJ, Joharapurkar AA, Kshirsagar SG, Sutariya BK, Patel MS, Patel HM, Pandey DK, Bahekar RH, Jain MR. Coagonist of glucagon-like peptide-1 and glucagon receptors ameliorates kidney injury in murine models of obesity and diabetes mellitus. World J Diabetes 2018; 9:80-91. [PMID: 29988851 PMCID: PMC6033704 DOI: 10.4239/wjd.v9.i6.80] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/10/2018] [Accepted: 05/15/2018] [Indexed: 02/05/2023] Open
Abstract
AIM To investigate the role of glucagon-like peptide-1 (GLP-1)/glucagon receptors coagonist on renal dysfunction associated with diabetes and obesity.
METHODS Chronic high-fat diet fed C57BL/6J mice, streptozotocin-treated high-fat diet fed C57BL/6J mice and diabetic C57BLKS/J db/db mice were used as models of diabetes-induced renal dysfunction. The streptozotocin-treated high-fat diet fed mice and db/db mice were treated with the GLP-1 and glucagon receptors coagonist (Aib2 C24 Chimera2, 150 μg/kg, sc) for twelve weeks, while in chronic high-fat diet fed mice, coagonist (Aib2 C24 Chimera2, 150 μg/kg, sc) treatment was continued for forty weeks. Kidney function, histology, fibrosis, inflammation, and plasma biochemistry were assessed at the end of the treatment.
RESULTS Coagonist treatment decreased body weight, plasma lipids, insulin resistance, creatinine, blood urea nitrogen, urinary albumin excretion rate and renal lipids. In kidney, expression of lipogenic genes (SREBP-1C, FAS, and SCD-1) was decreased, and expression of genes involved in β-oxidation (CPT-1 and PPAR-α) was increased due to coagonist treatment. In plasma, coagonist treatment increased adiponectin and FGF21 and decreased IL-6 and TNF-α. Coagonist treatment reduced expression of inflammatory (TNF-α, MCP-1, and MMP-9) and pro-fibrotic (TGF-β, COL1A1, and α-SMA) genes and also improved histological derangement in renal tissue.
CONCLUSION Coagonist of GLP-1 and glucagon receptors alleviated diabetes and obesity-induced renal dysfunction by reducing glucose intolerance, obesity, and hyperlipidemia.
Collapse
Affiliation(s)
- Vishal J Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Amit A Joharapurkar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Samadhan G Kshirsagar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Brijesh K Sutariya
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Maulik S Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Hiren M Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Dheerendra K Pandey
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Rajesh H Bahekar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| | - Mukul R Jain
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad 382210, India
| |
Collapse
|
173
|
Shankar SS, Shankar RR, Mixson LA, Miller DL, Pramanik B, O'Dowd AK, Williams DM, Frederick CB, Beals CR, Stoch SA, Steinberg HO, Kelley DE. Native Oxyntomodulin Has Significant Glucoregulatory Effects Independent of Weight Loss in Obese Humans With and Without Type 2 Diabetes. Diabetes 2018; 67:1105-1112. [PMID: 29545266 DOI: 10.2337/db17-1331] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/06/2018] [Indexed: 12/20/2022]
Abstract
Oxyntomodulin (OXM), an enteroendocrine hormone, causes appetite suppression, increased energy expenditure, and weight loss in obese humans via activation of GLP-1 and glucagon receptors. However, the effects of OXM on glucose homeostasis remain ill defined. To address this gap, we evaluated the effects of an i.v. infusion of native OXM on insulin secretion rates (ISRs) and glycemic excursion in a graded glucose infusion (GGI) procedure in two separate randomized, placebo (PBO)-controlled, single-dose crossover trials in 12 overweight and obese subjects without diabetes and in 12 obese subjects with type 2 diabetes mellitus (T2DM), using the GLP-1 analog liraglutide (LIRA) as a comparator in T2DM. In both groups, in the GGI, 3.0 pmol/kg/min of OXM significantly increased ISR and blunted glycemic excursion relative to PBO. In T2DM, the effects of OXM were comparable to those of LIRA, including restoration of β-cell glucose responsiveness to that of nonobese subjects without diabetes. Our findings indicate that native OXM significantly augments glucose-dependent insulin secretion acutely in obese subjects with and without diabetes, with effects comparable to pharmacologic GLP-1 receptor activation and independent of weight loss. Native OXM has potential to improve hyperglycemia via complementary and independent induction of insulin secretion and weight loss.
Collapse
|
174
|
Optimization of peptide-based polyagonists for treatment of diabetes and obesity. Bioorg Med Chem 2018; 26:2873-2881. [DOI: 10.1016/j.bmc.2017.10.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/31/2017] [Indexed: 12/28/2022]
|
175
|
Basco D, Zhang Q, Salehi A, Tarasov A, Dolci W, Herrera P, Spiliotis I, Berney X, Tarussio D, Rorsman P, Thorens B. α-cell glucokinase suppresses glucose-regulated glucagon secretion. Nat Commun 2018; 9:546. [PMID: 29416045 PMCID: PMC5803227 DOI: 10.1038/s41467-018-03034-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 01/15/2018] [Indexed: 02/08/2023] Open
Abstract
Glucagon secretion by pancreatic α-cells is triggered by hypoglycemia and suppressed by high glucose levels; impaired suppression of glucagon secretion is a hallmark of both type 1 and type 2 diabetes. Here, we show that α-cell glucokinase (Gck) plays a role in the control of glucagon secretion. Using mice with α-cell-specific inactivation of Gck (αGckKO mice), we find that glucokinase is required for the glucose-dependent increase in intracellular ATP/ADP ratio and the closure of KATP channels in α-cells and the suppression of glucagon secretion at euglycemic and hyperglycemic levels. αGckKO mice display hyperglucagonemia in the fed state, which is associated with increased hepatic gluconeogenic gene expression and hepatic glucose output capacity. In adult mice, fed hyperglucagonemia is further increased and glucose intolerance develops. Thus, glucokinase governs an α-cell metabolic pathway that suppresses secretion at or above normoglycemic levels; abnormal suppression of glucagon secretion deregulates hepatic glucose metabolism and, over time, induces a pre-diabetic phenotype.
Collapse
Affiliation(s)
- Davide Basco
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Albert Salehi
- Department of Clinical Science, UMAS, Division of Islet Cell Physiology, Lund, Sweden
| | - Andrei Tarasov
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Wanda Dolci
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Pedro Herrera
- Department of Genetic Medicine and Development, 1200, Geneva, Switzerland
| | - Ioannis Spiliotis
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Xavier Berney
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
176
|
Patel V, Joharapurkar A, Kshirsagar S, Patel M, Sutariya B, Patel H, Pandey D, Patel D, Ranvir R, Kadam S, Bahekar R, Jain M. Coagonist of glucagon-like peptide-1 and glucagon receptors ameliorates nonalcoholic fatty liver disease. Can J Physiol Pharmacol 2018; 96:587-596. [PMID: 29406832 DOI: 10.1139/cjpp-2017-0683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is often associated with obesity and type 2 diabetes. Coagonists of glucagon-like peptide-1 receptor (GLP-1R) and glucagon receptor (GCGR) are under clinical investigation for the treatment of obesity and type 2 diabetes. In this study, we have demonstrated the effect of a balanced coagonist in the treatment of NAFLD using mouse models. GLP-1R agonist exendin-4, glucagon, and coagonist (Aib2 C24 chimera2) were administered to C57BL6/J mice, in which NAFLD was induced by carbon tetrachloride (CCl4) treatment after high-fat diet (HFD) feeding, and choline-deficient, L-amino-acid-defined HFD (CDAHFD) feeding. Repeated dose administration of coagonist significantly attenuated liver inflammation and steatosis induced by acute and long-term treatment with CCl4 in HFD-fed mice. Coagonist markedly attenuated the CDAHFD-induced expression of TIMP-1, MMP-9, TNF-α, MCP-1, COL1A1, and α-SMA. It also inhibited progression of hepatic steatosis and fibrosis in mice. Exendin-4 was better than glucagon, but coagonist was most effective in reduction of hepatic inflammation as well as steatosis. Coagonist of GLP-1R and GCGR improved NAFLD in C57BL6/J mice. This effect is mediated by reduction in lipotoxicity and inflammation in liver.
Collapse
Affiliation(s)
- Vishal Patel
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Amit Joharapurkar
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Samadhan Kshirsagar
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Maulik Patel
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Brijesh Sutariya
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Hiren Patel
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Dheerendra Pandey
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Dipam Patel
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Ramchandra Ranvir
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Shekhar Kadam
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Rajesh Bahekar
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| | - Mukul Jain
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India.,Zydus Research Centre, Cadila Healthcare Limited, Sarkhej-Bavla N.H. No. 8A, Moraiya, Ahmedabad, 382210, India
| |
Collapse
|
177
|
Wewer Albrechtsen NJ. Glucagon receptor signaling in metabolic diseases. Peptides 2018; 100:42-47. [PMID: 29412830 DOI: 10.1016/j.peptides.2017.11.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/23/2017] [Accepted: 11/24/2017] [Indexed: 01/25/2023]
Abstract
Glucagon is a peptide hormone secreted from the pancreatic alpha cells in response to hypoglycemia but in some patients with type 2 diabetes a paradoxical hypersecretion results from the intake of glucose. In rodent, antagonizing the actions of glucagon have been shown to be effective for lowering blood glucose levels and this has recently have been solidified in patients with type 2 diabetes. Although the reported increases of liver enzymes, hyperglucagonemia, and alpha cell hyperplasia resulting from glucagon receptor antagonism may potentially limit the clinical applicability of glucagon receptor antagonists, they may serve as an instrumental toolbox for delineating the physiology of glucagon. Agonizing glucagon receptor signaling may be relevant, in particular when combined with glucagon-like peptide-1 receptor analogues in the perspective of body weight lowering therapy. Here, we will focus on new conceptual aspects of glucagon biology and how this may led to new diagnostics and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, and the Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.
| |
Collapse
|
178
|
Polypharmacy through Phage Display: Selection of Glucagon and GLP-1 Receptor Co-agonists from a Phage-Displayed Peptide Library. Sci Rep 2018; 8:585. [PMID: 29330364 PMCID: PMC5766609 DOI: 10.1038/s41598-017-18494-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/16/2017] [Indexed: 02/07/2023] Open
Abstract
A promising emerging area for the treatment of obesity and diabetes is combinatorial hormone therapy, where single-molecule peptides are rationally designed to integrate the complementary actions of multiple endogenous metabolically-related hormones. We describe here a proof-of-concept study on developing unimolecular polypharmacy agents through the use of selection methods based on phage-displayed peptide libraries (PDL). Co-agonists of the glucagon (GCG) and GLP-1 receptors were identified from a PDL sequentially selected on GCGR- and GLP1R-overexpressing cells. After two or three rounds of selection, 7.5% of randomly picked clones were GLP1R/GCGR co-agonists, and a further 1.53% were agonists of a single receptor. The phages were sequenced and 35 corresponding peptides were synthesized. 18 peptides were potent co-agonists, 8 of whom showed EC50 ≤ 30 pM on each receptor, comparable to the best rationally designed co-agonists reported in the literature. Based on literature examples, two sequences were engineered to stabilize against dipeptidyl peptidase IV cleavage and prolong the in vivo half-life: the engineered peptides were comparably potent to the parent peptides on both receptors, highlighting the potential use of phage-derived peptides as therapeutic agents. The strategy described here appears of general value for the discovery of optimized polypharmacology paradigms across several metabolically-related hormones.
Collapse
|
179
|
Goldstein I, Hager GL. The Three Ds of Transcription Activation by Glucagon: Direct, Delayed, and Dynamic. Endocrinology 2018; 159:206-216. [PMID: 29077799 PMCID: PMC6283435 DOI: 10.1210/en.2017-00521] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022]
Abstract
Upon lowered blood glucose occurring during fasting, glucagon is secreted from pancreatic islets, exerting various metabolic effects to normalize glucose levels. A considerable portion of these effects is mediated by glucagon-activated transcription factors (TFs) in liver. Glucagon directly activates several TFs via immediate cyclic adenosine monophosphate (cAMP)- and calcium-dependent signaling events. Among these TFs, cAMP response element-binding protein (CREB) is a major factor. CREB recruits histone-modifying enzymes and cooperates with other TFs on the chromatin template to increase the rate of gene transcription. In addition to direct signal transduction, the transcriptional effects of glucagon are also influenced by dynamic TF cross talk. Specifically, assisted loading of one TF by a companion TF leads to increased binding and activity. Lastly, transcriptional regulation by glucagon is also exerted by TF cascades by which a primary TF induces the gene expression of secondary TFs that bring about their activity a few hours after the initial glucagon signal. This mechanism of a delayed response may be instrumental in establishing the temporal organization of the fasting response by which distinct metabolic events separate early from prolonged fasting. In this mini-review, we summarize recent advances and critical discoveries in glucagon-dependent gene regulation with a focus on direct TF activation, dynamic TF cross talk, and TF cascades.
Collapse
Affiliation(s)
- Ido Goldstein
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Correspondence: Gordon L. Hager, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B602, Bethesda, Maryland 20892. E-mail: ; or Ido Goldstein, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B307, Bethesda, Maryland 20892. E-mail:
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Correspondence: Gordon L. Hager, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B602, Bethesda, Maryland 20892. E-mail: ; or Ido Goldstein, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B307, Bethesda, Maryland 20892. E-mail:
| |
Collapse
|
180
|
DiMarchi RD, Mayer JP, Gelfanov VM, Tschöp M. Max Bergmann award lecture:Macromolecular medicinal chemistry as applied to metabolic diseases. J Pept Sci 2018; 24. [PMID: 29322647 DOI: 10.1002/psc.3056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 11/06/2022]
Abstract
This review presents the scope of research presented in an October 2016 lecture pertaining to the award of the 2015 Max Bergmann Medal. The advancement in synthetic and biosynthetic chemistry as applied to the discovery of novel macromolecular drug candidates is reviewed. The evolution of the technology from the design, synthesis, and development of the first biosynthetic peptides through the emergence of peptide-based incretin agonists that function by multiple biological mechanisms is exemplified by the progression of such peptides from preclinical to clinical study. A closing section highlights recent progress made in total chemical synthesis of insulin and related peptides.
Collapse
Affiliation(s)
- Richard D DiMarchi
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, 46241, USA
| | - John P Mayer
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, 46241, USA
| | - Vasily M Gelfanov
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, 46241, USA
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333, Munich, Germany
| |
Collapse
|
181
|
Thiessen SE, Derde S, Derese I, Dufour T, Vega CA, Langouche L, Goossens C, Peersman N, Vermeersch P, Vander Perre S, Holst JJ, Wouters PJ, Vanhorebeek I, Van den Berghe G. Role of Glucagon in Catabolism and Muscle Wasting of Critical Illness and Modulation by Nutrition. Am J Respir Crit Care Med 2017; 196:1131-1143. [PMID: 28475354 DOI: 10.1164/rccm.201702-0354oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RATIONALE Critical illness is hallmarked by muscle wasting and disturbances in glucose, lipid, and amino acid homeostasis. Circulating concentrations of glucagon, a catabolic hormone that affects these metabolic pathways, are elevated during critical illness. Insight in the nutritional regulation of glucagon and its metabolic role during critical illness is lacking. OBJECTIVES To evaluate whether macronutrient infusion can suppress plasma glucagon during critical illness and study the role of illness-induced glucagon abundance in the disturbed glucose, lipid, and amino acid homeostasis and in muscle wasting during critical illness. METHODS In human and mouse studies, we infused macronutrients and manipulated glucagon availability up and down to investigate its acute and chronic metabolic role during critical illness. MEASUREMENTS AND MAIN RESULTS In critically ill patients, infusing glucose with insulin did not lower glucagon, whereas parenteral nutrition containing amino acids increased glucagon. In critically ill mice, infusion of amino acids increased glucagon and up-regulated markers of hepatic amino acid catabolism without affecting muscle wasting. Immunoneutralizing glucagon in critically ill mice only transiently affected glucose and lipid metabolism, did not affect muscle wasting, but drastically suppressed markers of hepatic amino acid catabolism and reversed the illness-induced hypoaminoacidemia. CONCLUSIONS These data suggest that elevated glucagon availability during critical illness increases hepatic amino acid catabolism, explaining the illness-induced hypoaminoacidemia, without affecting muscle wasting and without a sustained impact on blood glucose. Furthermore, amino acid infusion likely results in a further breakdown of amino acids in the liver, mediated by increased glucagon, without preventing muscle wasting. Clinical trial registered with www.clinicaltrials.gov (NCT 00512122).
Collapse
Affiliation(s)
- Steven E Thiessen
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Sarah Derde
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Inge Derese
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Thomas Dufour
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Chloé Albert Vega
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Lies Langouche
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Chloë Goossens
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Nele Peersman
- 2 Department of Laboratory Medicine, KU Leuven, Leuven, Belgium; and
| | - Pieter Vermeersch
- 2 Department of Laboratory Medicine, KU Leuven, Leuven, Belgium; and
| | - Sarah Vander Perre
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Jens J Holst
- 3 Novo Nordisk Foundation Center for Basic Metabolic Research and.,4 Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Pieter J Wouters
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Ilse Vanhorebeek
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| | - Greet Van den Berghe
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, and
| |
Collapse
|
182
|
Grøndahl MF, Keating DJ, Vilsbøll T, Knop FK. Current Therapies That Modify Glucagon Secretion: What Is the Therapeutic Effect of Such Modifications? Curr Diab Rep 2017; 17:128. [PMID: 29080075 DOI: 10.1007/s11892-017-0967-z] [Citation(s) in RCA: 15] [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] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Hyperglucagonemia contributes significantly to hyperglycemia in type 2 diabetes and suppressed glucagon levels may increase the risk of hypoglycemia. Here, we give a brief overview of glucagon physiology and the role of glucagon in the pathophysiology of type 2 diabetes and provide insights into how antidiabetic drugs influence glucagon secretion as well as a perspective on the future of glucagon-targeting drugs. RECENT FINDINGS Several older as well as recent investigations have evaluated the effect of antidiabetic agents on glucagon secretion to understand how glucagon may be involved in the drugs' efficacy and safety profiles. Based on these findings, modulation of glucagon secretion seems to play a hitherto underestimated role in the efficacy and safety of several glucose-lowering drugs. Numerous drugs currently available to diabetologists are capable of altering glucagon secretion: metformin, sulfonylurea compounds, insulin, glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase-4 inhibitors, sodium-glucose cotransporter 2 inhibitors and amylin mimetics. Their diverse effects on glucagon secretion are of importance for their individual efficacy and safety profiles. Understanding how these drugs interact with glucagon secretion may help to optimize treatment.
Collapse
Affiliation(s)
- Magnus F Grøndahl
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Damien J Keating
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, University of Copenhagen, Gentofte, Denmark
| | - Filip K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical sciences, University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Metabolic Research, Faculty of Health and Medical sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
183
|
Sánchez-Garrido MA, Brandt SJ, Clemmensen C, Müller TD, DiMarchi RD, Tschöp MH. GLP-1/glucagon receptor co-agonism for treatment of obesity. Diabetologia 2017; 60:1851-1861. [PMID: 28733905 PMCID: PMC6448809 DOI: 10.1007/s00125-017-4354-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/28/2017] [Indexed: 12/25/2022]
Abstract
Over a relatively short period, obesity and type 2 diabetes have come to represent a large medical and economic burden to global societies. The epidemic rise in the prevalence of obesity has metabolic consequences and is paralleled by an increased occurrence of other diseases, such as diabetes, cancer and cardiovascular complications. Together, obesity and type 2 diabetes constitute one of the more preventable causes of premature death and the identification of novel, safe and effective anti-obesity drugs is of utmost importance. Pharmacological attempts to treat obesity have had limited success, with notable adverse effects, rendering bariatric surgery as the only current therapy for substantially improving body weight. Novel unimolecular, multifunctional peptides have emerged as one of the most promising medicinal approaches to enhance metabolic efficacy and restore normal body weight. In this review, we will mainly focus on the discovery and translational relevance of dual agonists that pharmacologically function at the receptors for glucagon and glucagon-like peptide-1. Such peptides have advanced to clinical evaluation and inspired the pursuit of multiple related approaches to achieving polypharmacy within single molecules.
Collapse
Affiliation(s)
- Miguel A Sánchez-Garrido
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Business Campus Garching, Parkring 13, 85748, Garching, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sara J Brandt
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Business Campus Garching, Parkring 13, 85748, Garching, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christoffer Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Business Campus Garching, Parkring 13, 85748, Garching, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Business Campus Garching, Parkring 13, 85748, Garching, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Richard D DiMarchi
- Department of Chemistry, Indiana University, 800 E Kirkwood Ave, Bloomington, IN, 47405, USA.
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Business Campus Garching, Parkring 13, 85748, Garching, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany.
| |
Collapse
|
184
|
Abstract
The liver is crucial for the maintenance of normal glucose homeostasis - it produces glucose during fasting and stores glucose postprandially. However, these hepatic processes are dysregulated in type 1 and type 2 diabetes mellitus, and this imbalance contributes to hyperglycaemia in the fasted and postprandial states. Net hepatic glucose production is the summation of glucose fluxes from gluconeogenesis, glycogenolysis, glycogen synthesis, glycolysis and other pathways. In this Review, we discuss the in vivo regulation of these hepatic glucose fluxes. In particular, we highlight the importance of indirect (extrahepatic) control of hepatic gluconeogenesis and direct (hepatic) control of hepatic glycogen metabolism. We also propose a mechanism for the progression of subclinical hepatic insulin resistance to overt fasting hyperglycaemia in type 2 diabetes mellitus. Insights into the control of hepatic gluconeogenesis by metformin and insulin and into the role of lipid-induced hepatic insulin resistance in modifying gluconeogenic and net hepatic glycogen synthetic flux are also discussed. Finally, we consider the therapeutic potential of strategies that target hepatosteatosis, hyperglucagonaemia and adipose lipolysis.
Collapse
Affiliation(s)
- Max C Petersen
- Department of Internal Medicine, Yale School of Medicine
- Department of Cellular &Molecular Physiology, Yale School of Medicine
| | | | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine
- Department of Cellular &Molecular Physiology, Yale School of Medicine
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
185
|
Velázquez-Villegas LA, Tovar-Palacio C, Palacios-González B, Torres N, Tovar AR, Díaz-Villaseñor A. Recycling of glucagon receptor to plasma membrane increases in adipocytes of obese rats by soy protein; implications for glucagon resistance. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201700116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/24/2017] [Accepted: 06/14/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Laura A. Velázquez-Villegas
- Departmento de Fisiología de la Nutrición; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; Mexico City Mexico
| | - Claudia Tovar-Palacio
- Departmento de Nefrología y Metabolismo Mineral; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; Mexico City Mexico
| | - Berenice Palacios-González
- Departmento de Fisiología de la Nutrición; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; Mexico City Mexico
| | - Nimbe Torres
- Departmento de Fisiología de la Nutrición; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; Mexico City Mexico
| | - Armando R. Tovar
- Departmento de Fisiología de la Nutrición; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; Mexico City Mexico
| | - Andrea Díaz-Villaseñor
- Departmento de Fisiología de la Nutrición; Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán; Mexico City Mexico
- Departmento de Medicina Genómica y Toxicología Ambiental; Instituto de Investigaciones Biomédicas; Universidad Nacional Autónoma de México; Mexico City Mexico
| |
Collapse
|
186
|
Zhou J, Cai X, Huang X, Dai Y, Sun L, Zhang B, Yang B, Lin H, Huang W, Qian H. A novel glucagon-like peptide-1/glucagon receptor dual agonist exhibits weight-lowering and diabetes-protective effects. Eur J Med Chem 2017; 138:1158-1169. [PMID: 28772236 DOI: 10.1016/j.ejmech.2017.07.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/19/2017] [Accepted: 07/22/2017] [Indexed: 10/19/2022]
Abstract
Glucagon has plenty of effects via a specific glucagon receptor(GCGR) like elevating the blood glucose, improving fatty acids metabolism, energy expenditure and increasing lipolysis in adipose tissue. The most important role of glucagon is to regulate the blood glucose, but the emergent possibilities of hyperglycaemia is exist. Glucagon could also slightly activate glucagon-like peptide-1 receptor(GLP-1R), which lead to blood glucose lowering effect. This study aims to erase the likelihood of hyperglycaemia and to remain the inherent catabolic effects through improving GLP-1R activation and deteriorating GCGR activation so as to lower the bodyweight and show diabetes-protective effects. Firstly, twelve cysteine modified GLP-1/GCGR dual agonists were synthesized (1-12). Then, the GLP-1R/GCGR mediated activation and biological activity in normal ICR mice were comprehensively performed. Compounds substituted by cysteine at positions 22, 23 and 25 in glucagon were observed to be better regulators of the body weight and blood glucose. To prolong the half-lives of derivatives, various fatty side chain maleimides were modified to optimal glucagon analogues. Laurate maleimide conjugate 4d was the most potent. Administration of 1000 nmol/kg 4d once every two days for a month normalized adiposity and glucose tolerance in diet-induced obese (DIO) mice. Improvements in plasma metabolic parameters including insulin, leptin, and adiponectin were observed. These studies suggest that compound 4d behaves well in lowering body weight and maintaining energy expenditure without a chance of hyperglycaemia, 4d has strong clinical potential as an efficient GLP-1/GCGR agonist in the prevention and treatment of obesity and dyslipidemia.
Collapse
Affiliation(s)
- Jie Zhou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China; Structural Biology Center, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Xingguang Cai
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xun Huang
- Division of Antitumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, PR China
| | - Yuxuan Dai
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lidan Sun
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bo Zhang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bo Yang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Haiyan Lin
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, PR China.
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China.
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China.
| |
Collapse
|
187
|
Tschöp M, DiMarchi R. Single-Molecule Combinatorial Therapeutics for Treating Obesity and Diabetes. Diabetes 2017; 66:1766-1769. [PMID: 28603138 DOI: 10.2337/dbi16-0068] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/01/2017] [Indexed: 11/13/2022]
Affiliation(s)
- Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, and German Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany, and Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Richard DiMarchi
- Department of Chemistry, Indiana University, Bloomington, IN, and Novo Nordisk Research Center Indianapolis, Indianapolis, IN
| |
Collapse
|
188
|
Neumann UH, Ho JSS, Chen S, Tam YYC, Cullis PR, Kieffer TJ. Lipid nanoparticle delivery of glucagon receptor siRNA improves glucose homeostasis in mouse models of diabetes. Mol Metab 2017; 6:1161-1172. [PMID: 29031717 PMCID: PMC5641600 DOI: 10.1016/j.molmet.2017.06.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 12/15/2022] Open
Abstract
Objective Hyperglucagonemia is present in many forms of diabetes and contributes to hyperglycemia, and glucagon suppression can ameliorate diabetes in mice. Leptin, a glucagon suppressor, can also reverse diabetes in rodents. Lipid nanoparticle (LNP) delivery of small interfering RNA (siRNA) effectively targets the liver and is in clinical trials for the treatment of various diseases. We compared the effectiveness of glucagon receptor (Gcgr)-siRNA delivered via LNPs to leptin in two mouse models of diabetes. Methods Gcgr siRNA encapsulated into LNPs or leptin was administered to mice with diabetes due to injection of the β-cell toxin streptozotocin (STZ) alone or combined with high fat diet (HFD/STZ). Results In STZ-diabetic mice, a single injection of Gcgr siRNA lowered blood glucose levels for 3 weeks, improved glucose tolerance, and normalized plasma ketones levels, while leptin therapy normalized blood glucose levels, oral glucose tolerance, and plasma ketones, and suppressed lipid metabolism. In contrast, in HFD/STZ-diabetic mice, Gcgr siRNA lowered blood glucose levels for 2 months, improved oral glucose tolerance, and reduced HbA1c, while leptin had no beneficial effects. Conclusions While leptin may be more effective than Gcgr siRNA at normalizing both glucose and lipid metabolism in STZ diabetes, Gcgr siRNA is more effective at reducing blood glucose levels in HFD/STZ diabetes. Gcgr siRNA improves glucose metabolism but not lipid metabolism in STZ diabetic mice. Leptin improves both glucose and lipid metabolism in STZ diabetic mice. Gcgr siRNA improves glucose metabolism in HFD/STZ diabetic mice. Leptin does not improve glucose metabolism in HFD/STZ diabetic mice.
Collapse
Affiliation(s)
- Ursula H Neumann
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Jessica S S Ho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Sam Chen
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Yuen Yi C Tam
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
189
|
Clemmensen C, Müller TD, Woods SC, Berthoud HR, Seeley RJ, Tschöp MH. Gut-Brain Cross-Talk in Metabolic Control. Cell 2017; 168:758-774. [PMID: 28235194 DOI: 10.1016/j.cell.2017.01.025] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/19/2016] [Accepted: 01/23/2017] [Indexed: 12/15/2022]
Abstract
Because human energy metabolism evolved to favor adiposity over leanness, the availability of palatable, easily attainable, and calorically dense foods has led to unprecedented levels of obesity and its associated metabolic co-morbidities that appear resistant to traditional lifestyle interventions. However, recent progress identifying the molecular signaling pathways through which the brain and the gastrointestinal system communicate to govern energy homeostasis, combined with emerging insights on the molecular mechanisms underlying successful bariatric surgery, gives reason to be optimistic that novel precision medicines that mimic, enhance, and/or modulate gut-brain signaling can have unprecedented potential for stopping the obesity and type 2 diabetes pandemics.
Collapse
Affiliation(s)
- Christoffer Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45220, USA
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70803, USA
| | - Randy J Seeley
- Departments of Surgery, Internal Medicine, and Nutritional Sciences at the University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Center for Diabetes Research (DZD), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany.
| |
Collapse
|
190
|
Evers A, Haack T, Lorenz M, Bossart M, Elvert R, Henkel B, Stengelin S, Kurz M, Glien M, Dudda A, Lorenz K, Kadereit D, Wagner M. Design of Novel Exendin-Based Dual Glucagon-like Peptide 1 (GLP-1)/Glucagon Receptor Agonists. J Med Chem 2017; 60:4293-4303. [DOI: 10.1021/acs.jmedchem.7b00174] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Andreas Evers
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Torsten Haack
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Martin Lorenz
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Martin Bossart
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Ralf Elvert
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Bernd Henkel
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Siegfried Stengelin
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Michael Kurz
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Maike Glien
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Angela Dudda
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Katrin Lorenz
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Dieter Kadereit
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Michael Wagner
- R&D, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| |
Collapse
|
191
|
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: 249] [Impact Index Per Article: 31.1] [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
|
192
|
Affiliation(s)
- Nori Geary
- Department of Psychiatry, Weill Cornell Medical College, New York, NY
| |
Collapse
|
193
|
Wagner R, Hakaste LH, Ahlqvist E, Heni M, Machann J, Schick F, Van Obberghen E, Stefan N, Gallwitz B, Tuomi T, Häring HU, Groop L, Fritsche A. Nonsuppressed Glucagon After Glucose Challenge as a Potential Predictor for Glucose Tolerance. Diabetes 2017; 66:1373-1379. [PMID: 27986831 DOI: 10.2337/db16-0354] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 12/02/2016] [Indexed: 11/13/2022]
Abstract
Glucagon levels are classically suppressed after glucose challenge. It is still not clear as to whether a lack of suppression contributes to hyperglycemia and thus to the development of diabetes. We investigated the association of postchallenge change in glucagon during oral glucose tolerance tests (OGTTs), hypothesizing that higher postchallenge glucagon levels are observed in subjects with impaired glucose tolerance (IGT). Glucagon levels were measured during OGTT in a total of 4,194 individuals without diabetes in three large European cohorts. Longitudinal changes in glucagon suppression were investigated in 50 participants undergoing a lifestyle intervention. Only 66-79% of participants showed suppression of glucagon at 120 min (fold change glucagon120/0 <1) during OGTT, whereas 21-34% presented with increasing glucagon levels (fold change glucagon120/0 ≥1). Participants with nonsuppressed glucagon120 had a lower risk of IGT in all cohorts (odds ratio 0.44-0.53, P < 0.01). They were also leaner and more insulin sensitive and had lower liver fat contents. In the longitudinal study, an increase of fold change glucagon120/0 was associated with an improvement in insulin sensitivity (P = 0.003). We characterize nonsuppressed glucagon120 during the OGTT. Lower glucagon suppression after oral glucose administration is associated with a metabolically healthier phenotype, suggesting that it is not an adverse phenomenon.
Collapse
Affiliation(s)
- Róbert Wagner
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease, and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Liisa H Hakaste
- Diabetes and Obesity Research Program, Research Programs Unit, Folkhälsan Research Centre, University of Helsinki, Helsinki, Finland
| | - Emma Ahlqvist
- Diabetes and Endocrinology, Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Martin Heni
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease, and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Fritz Schick
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Emmanuel Van Obberghen
- Université Côte d'Azur, CHU, INSERM, CNRS, Institute of Research on Cancer and Aging in Nice (IRCAN), Nice, France
| | - Norbert Stefan
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease, and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Baptist Gallwitz
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease, and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich, University of Tübingen, Tübingen, Germany
| | - Tiinamaija Tuomi
- Diabetes and Obesity Research Program, Research Programs Unit, Folkhälsan Research Centre, University of Helsinki, Helsinki, Finland
- Department of Endocrinology, Abdominal Centre, Helsinki University Hospital, Helsinki, Finland
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease, and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Leif Groop
- Diabetes and Endocrinology, Department of Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
- Finnish Institute of Molecular Medicine, Helsinki University, Helsinki, Finland
| | - Andreas Fritsche
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease, and Clinical Chemistry, Department of Internal Medicine, University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholz Centre Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|
194
|
Bergman A, Tan B, Somayaji VR, Calle RA, Kazierad DJ. A 4-week study assessing the pharmacokinetics, pharmacodynamics, safety, and tolerability of the glucagon receptor antagonist PF-06291874 administered as monotherapy in subjects with type 2 diabetes mellitus. Diabetes Res Clin Pract 2017; 126:95-104. [PMID: 28237861 DOI: 10.1016/j.diabres.2017.01.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/30/2017] [Indexed: 11/24/2022]
Abstract
AIMS The glucagon receptor antagonist PF-06291874 has demonstrated robust glucose reductions in subjects with type 2 diabetes mellitus (T2DM) on background metformin. This study assessed the pharmacokinetics, pharmacodynamics, safety, and tolerability of PF-06291874 administered as monotherapy in subjects with T2DM. METHODS After a ≥4-week antidiabetic therapy washout period, 172 subjects were randomized to placebo or PF-06291874 15, 35, 75, or 150mg once daily for 28days. Mean daily glucose (MDG), fasting plasma glucose (FPG), and predefined safety endpoints were assessed at baseline and day 28. RESULTS Dose-dependent reductions (placebo-adjusted) from baseline in MDG ranged from 40.3 to 68.8mg/dL and in FPG from 27.1 to 57.2mg/dL after 28days of dosing with PF-06291874. There were no significant changes in low-density lipoprotein cholesterol at doses ≤75mg relative to placebo. Small, dose-dependent increases in alanine aminotransferase and aspartate aminotransferase were observed; however, the incidence of these values >3×upper limit of normal was similar across doses. PF-06291874 exposures were consistent with previous studies and PF-06291874 was well tolerated, with minimal incidence of hypoglycemia. CONCLUSIONS PF-06291874 as monotherapy was well tolerated and produced robust reductions in plasma glucose following 4weeks of dosing in subjects with T2DM.
Collapse
|
195
|
Alonge KM, Meares GP, Hillgartner FB. Glucagon and Insulin Cooperatively Stimulate Fibroblast Growth Factor 21 Gene Transcription by Increasing the Expression of Activating Transcription Factor 4. J Biol Chem 2017; 292:5239-5252. [PMID: 28188284 DOI: 10.1074/jbc.m116.762922] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/02/2017] [Indexed: 01/07/2023] Open
Abstract
Previous studies have shown that glucagon cooperatively interacts with insulin to stimulate hepatic FGF21 gene expression. Here we investigated the mechanism by which glucagon and insulin increased FGF21 gene transcription in primary hepatocyte cultures. Transfection analyses demonstrated that glucagon plus insulin induction of FGF21 transcription was conferred by two activating transcription factor 4 (ATF4) binding sites in the FGF21 gene. Glucagon plus insulin stimulated a 5-fold increase in ATF4 protein abundance, and knockdown of ATF4 expression suppressed the ability of glucagon plus insulin to increase FGF21 expression. In hepatocytes incubated in the presence of insulin, treatment with a PKA-selective agonist mimicked the ability of glucagon to stimulate ATF4 and FGF21 expression. Inhibition of PKA, PI3K, Akt, and mammalian target of rapamycin complex 1 (mTORC1) suppressed the ability of glucagon plus insulin to stimulate ATF4 and FGF21 expression. Additional analyses demonstrated that chenodeoxycholic acid (CDCA) induced a 6-fold increase in ATF4 expression and that knockdown of ATF4 expression suppressed the ability of CDCA to increase FGF21 gene expression. CDCA increased the phosphorylation of eIF2α, and inhibition of eIF2α signaling activity suppressed CDCA regulation of ATF4 and FGF21 expression. These results demonstrate that glucagon plus insulin increases FGF21 transcription by stimulating ATF4 expression and that activation of cAMP/PKA and PI3K/Akt/mTORC1 mediates the effect of glucagon plus insulin on ATF4 expression. These results also demonstrate that CDCA regulation of FGF21 transcription is mediated at least partially by an eIF2α-dependent increase in ATF4 expression.
Collapse
Affiliation(s)
| | - Gordon P Meares
- Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia 26506
| | | |
Collapse
|
196
|
Sekar R, Wang L, Chow BKC. Central Control of Feeding Behavior by the Secretin, PACAP, and Glucagon Family of Peptides. Front Endocrinol (Lausanne) 2017; 8:18. [PMID: 28223965 PMCID: PMC5293785 DOI: 10.3389/fendo.2017.00018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/18/2017] [Indexed: 12/25/2022] Open
Abstract
Constituting a group of structurally related brain-gut peptides, secretin (SCT), pituitary adenylate cyclase-activating peptide (PACAP), and glucagon (GCG) family of peptide hormones exert their functions via interactions with the class B1 G protein-coupled receptors. In recent years, the roles of these peptides in neuroendocrine control of feeding behavior have been a specific area of research focus for development of potential therapeutic drug targets to combat obesity and metabolic disorders. As a result, some members in the family and their analogs have already been utilized as therapeutic agents in clinical application. This review aims to provide an overview of the current understanding on the important role of SCT, PACAP, and GCG family of peptides in central control of feeding behavior.
Collapse
Affiliation(s)
- Revathi Sekar
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Lei Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
197
|
Mroz PA, Perez-Tilve D, Liu F, Mayer JP, DiMarchi RD. Native Design of Soluble, Aggregation-Resistant Bioactive Peptides: Chemical Evolution of Human Glucagon. ACS Chem Biol 2016; 11:3412-3420. [PMID: 27797473 DOI: 10.1021/acschembio.6b00923] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Peptide-based therapeutics commonly suffer from biophysical properties that compromise pharmacology and medicinal use. Structural optimization of the primary sequence is the usual route to address such challenges while trying to maintain as much native character and avoiding introduction of any foreign element that might evoke an immunological response. Glucagon serves a seminal physiological role in buffering against hypoglycemia, but its low aqueous solubility, chemical instability, and propensity to self-aggregate severely complicate its medicinal use. Selective amide bond replacement with metastable ester bonds is a preferred approach to the preparation of peptides with biophysical properties that otherwise inhibit synthesis. We have recruited such chemistry in the design and development of unique glucagon prodrugs that have physical properties suitable for medicinal use and yet rapidly convert to native hormone upon exposure to slightly alkaline pH. These prodrugs demonstrate in vitro and in vivo pharmacology when formulated in physiological buffers that are nearly identical to native hormone when solubilized in conventional dilute hydrochloric acid. This approach provides the best of both worlds, where the pro-drug delivers chemical properties supportive of aqueous formulation and the native biological properties.
Collapse
Affiliation(s)
- Piotr A. Mroz
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Diego Perez-Tilve
- Department
of Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio 45267, United States
| | - Fa Liu
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | - John P. Mayer
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| | - Richard D. DiMarchi
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
- Novo Nordisk Research Center, Indianapolis, Indiana 46241, United States
| |
Collapse
|
198
|
Henderson SJ, Konkar A, Hornigold DC, Trevaskis JL, Jackson R, Fritsch Fredin M, Jansson‐Löfmark R, Naylor J, Rossi A, Bednarek MA, Bhagroo N, Salari H, Will S, Oldham S, Hansen G, Feigh M, Klein T, Grimsby J, Maguire S, Jermutus L, Rondinone CM, Coghlan MP. Robust anti-obesity and metabolic effects of a dual GLP-1/glucagon receptor peptide agonist in rodents and non-human primates. Diabetes Obes Metab 2016; 18:1176-1190. [PMID: 27377054 PMCID: PMC5129521 DOI: 10.1111/dom.12735] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 06/20/2016] [Accepted: 06/29/2016] [Indexed: 01/11/2023]
Abstract
AIMS To characterize the pharmacology of MEDI0382, a peptide dual agonist of glucagon-like peptide-1 (GLP-1) and glucagon receptors. MATERIALS AND METHODS MEDI0382 was evaluated in vitro for its ability to stimulate cAMP accumulation in cell lines expressing transfected recombinant or endogenous GLP-1 or glucagon receptors, to potentiate glucose-stimulated insulin secretion (GSIS) in pancreatic β-cell lines and stimulate hepatic glucose output (HGO) by primary hepatocytes. The ability of MEDI0382 to reduce body weight and improve energy balance (i.e. food intake and energy expenditure), as well as control blood glucose, was evaluated in mouse models of obesity and healthy cynomolgus monkeys following single and repeated daily subcutaneous administration for up to 2 months. RESULTS MEDI0382 potently activated rodent, cynomolgus and human GLP-1 and glucagon receptors and exhibited a fivefold bias for activation of GLP-1 receptor versus the glucagon receptor. MEDI0382 produced superior weight loss and comparable glucose lowering to the GLP-1 peptide analogue liraglutide when administered daily at comparable doses in DIO mice. The additional fat mass reduction elicited by MEDI0382 probably results from a glucagon receptor-mediated increase in energy expenditure, whereas food intake suppression results from activation of the GLP-1 receptor. Notably, the significant weight loss elicited by MEDI0382 in DIO mice was recapitulated in cynomolgus monkeys. CONCLUSIONS Repeated administration of MEDI0382 elicits profound weight loss in DIO mice and non-human primates, produces robust glucose control and reduces hepatic fat content and fasting insulin and glucose levels. The balance of activities at the GLP-1 and glucagon receptors is considered to be optimal for achieving weight and glucose control in overweight or obese Type 2 diabetic patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - S. Will
- MedImmune LLCGaithersburgMDUSA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Lund A, Bagger JI, Christensen M, Grøndahl M, van Hall G, Holst JJ, Vilsbøll T, Knop FK. Higher Endogenous Glucose Production During OGTT vs Isoglycemic Intravenous Glucose Infusion. J Clin Endocrinol Metab 2016; 101:4377-4384. [PMID: 27533305 DOI: 10.1210/jc.2016-1948] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Oral glucose ingestion elicits a larger insulin response and delayed suppression of glucagon compared to isoglycemic IV glucose infusion (IIGI). OBJECTIVE We studied whether these differences translate into effects on endogenous glucose production (EGP) and glucose disposal in patients with type 2 diabetes and nondiabetic control subjects. DESIGN This was a single-blinded, randomized, crossover study. SETTING The study was conducted at a specialized research unit. PARTICIPANTS Ten patients with type 2 diabetes (age, [mean ± SD] 57.1 ± 6.7 years; body mass index, 29.0 ± 4.3 kg/m2; hemoglobin A1c, 53.8 ± 11.0 mmol/mol; duration of diabetes, 9.2 ± 5.0 years) and 10 matched nondiabetic control subjects (age, 56.0±10.7 years; body mass index, 29.8 ± 2.9 kg/m2; hemoglobin A1c, 33.8 ± 5.5 mmol/mol) participated. INTERVENTIONS Three experimental days: 75 g-oral glucose tolerance test (OGTT), IIGI, and IIGI+glucagon (IIGI with a concomitant IV glucagon infusion [0.8 ng/kg/min from 0 to 25 minutes] designed to mimic portal glucagon concentrations during OGTT in the type 2 diabetic group) were undertaken. MAIN OUTCOME MEASURES Glucose kinetics were assessed by tracer methodology. RESULTS Glucose rate of disappearance was higher during the OGTT vs IIGI in the control group, but similar on all days in the diabetic group. Surprisingly, in both groups, EGP was more suppressed during IIGI than during OGTT, and exogenous glucagon infusion during IIGI did not restore EGP to the levels observed during OGTT. CONCLUSION EGP was less suppressed during OGTT than during IIGI in both patients with type 2 diabetes and in nondiabetic control subjects. Based on the present experimental design, it was not possible to attribute this difference to the delayed glucagon suppression observed in the initial phase of the OGTT.
Collapse
Affiliation(s)
- Asger Lund
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jonatan I Bagger
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Christensen
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Magnus Grøndahl
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Gerrit van Hall
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Center for Diabetes Research (A.L., J.I.B., M.C., M.G., T.V., F.K.K.), Gentofte Hospital, University of Copenhagen, Hellerup, Denmark; The NNF Center for Basic Metabolic Research (A.L., J.J.H., F.K.K.), Copenhagen, Denmark; Department of Clinical Medicine (T.V., F.K.K.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology (M.C.), Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences (G.v.H., J.J.H.), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; and Department of Clinical Biochemistry (G.v.H.), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
200
|
Chemical Hybridization of Glucagon and Thyroid Hormone Optimizes Therapeutic Impact for Metabolic Disease. Cell 2016; 167:843-857.e14. [PMID: 27720451 DOI: 10.1016/j.cell.2016.09.014] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 06/24/2016] [Accepted: 09/07/2016] [Indexed: 11/22/2022]
Abstract
Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.
Collapse
|