1
|
Abe T, Takeda Y, Sakuma I, Okada M, Kurigaki A, Bessho R, Sato M, Kitsunai H, Takiyama Y, Sakurai M. Efficacy of Alogliptin/Metformin Fixed-Dose Combination Tablets and Vildagliptin/Metformin Fixed-Dose Combination Tablets on Glycemic Control in Real-World Clinical Practice for the Patients with Type 2 Diabetes: A Multicenter, Open-Label, Randomized, Parallel Group, Comparative Trial. Metab Syndr Relat Disord 2024; 22:651-660. [PMID: 39421912 DOI: 10.1089/met.2024.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Background: This study was aimed to compare the efficacy of two combination tablets of dipeptidyl peptidase-4 (DPP-4) inhibitors and metformin with different dosages, alogliptin/metformin (AM) and vildagliptin/metformin (VM), on glycemic control in patients with type 2 diabetes (T2D). Methods: This was a prospective, multicenter, open-label, randomized, parallel group, comparative trial. After a run-in period of treatment with metformin alone, a total of 59 Japanese outpatients with T2D, aged 20-79 years with glycated hemoglobin (HbA1c) levels of 6.5%-10% were randomly assigned to 12-week AM treatment, alogliptin 25 mg/metformin 500 mg combination tablet orally once a day, or VM treatment, vildagliptin 50 mg/metformin 250 mg combination tablet orally twice a day. The primary endpoints were the changes in HbA1c and fasting plasma glucose (FPG) levels from baseline to week 12 between the two groups. Blinded intermittently scanned continuous glucose monitoring (isCGM) was performed between weeks 10 and 12. The incidence of adverse events during the study was also evaluated. Results: In all, 52 participants were analyzed. Significant decreases in HbA1c and FPG levels from baseline to week 12 were observed in both treatment groups. However, there were no significant differences between the AM and VM groups in the change in HbA1c level (-0.3% and -0.4%, P = 0.309) or the FPG level (-9.0 and -15.0 mg/dL, P = 0.789). The isCGM revealed that both treatments achieved the recommended glycemic target range. No adverse events, such as severe hypoglycemia, were observed in either group. Conclusions: We concluded that there were no significant differences in the efficacy of two combination tablets of DPP-4 inhibitors and metformin with different dosages on glycemic control in patients with T2D.
Collapse
Affiliation(s)
- Tomoe Abe
- Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
- Sapporo Diabetes and Thyroid Clinic, Sapporo, Japan
| | - Yasutaka Takeda
- Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Ichiro Sakuma
- Caress Sapporo Hokko Memorial Clinic, Sapporo, Japan
| | | | - Ayaka Kurigaki
- Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Ryoichi Bessho
- Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Mao Sato
- Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Hiroya Kitsunai
- Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yumi Takiyama
- Department of Internal Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Masaru Sakurai
- Department of Social and Environmental Medicine, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
2
|
Aldehoff AS, Karkossa I, Goerdeler C, Krieg L, Schor J, Engelmann B, Wabitsch M, Landgraf K, Hackermüller J, Körner A, Rolle-Kampczyk U, Schubert K, von Bergen M. Unveiling the dynamics of acetylation and phosphorylation in SGBS and 3T3-L1 adipogenesis. iScience 2024; 27:109711. [PMID: 38840842 PMCID: PMC11152682 DOI: 10.1016/j.isci.2024.109711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/11/2023] [Accepted: 04/06/2024] [Indexed: 06/07/2024] Open
Abstract
Obesity, characterized by enlarged and dysfunctional adipose tissue, is among today's most pressing global public health challenges with continuously increasing prevalence. Despite the importance of post-translational protein modifications (PTMs) in cellular signaling, knowledge of their impact on adipogenesis remains limited. Here, we studied the temporal dynamics of transcriptome, proteome, central carbon metabolites, and the acetyl- and phosphoproteome during adipogenesis using LC-MS/MS combined with PTM enrichment strategies on human (SGBS) and mouse (3T3-L1) adipocyte models. Both cell lines exhibited unique PTM profiles during adipogenesis, with acetylated proteins being enriched for central energy metabolism, while phosphorylated proteins related to insulin signaling and organization of cellular structures. As candidates with strong correlation to the adipogenesis timeline we identified CD44 and the acetylation sites FASN_K673 and IDH_K272. While results generally aligned between SGBS and 3T3-L1 cells, details appeared cell line specific. Our datasets on SGBS and 3T3-L1 adipogenesis dynamics are accessible for further mining.
Collapse
Affiliation(s)
- Alix Sarah Aldehoff
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Cornelius Goerdeler
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Laura Krieg
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Jana Schor
- Department of Computational Biology and Chemistry, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Beatrice Engelmann
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, University Hospital for Children and Adolescents Ulm, Ulm, Germany
| | - Kathrin Landgraf
- University Hospital for Children and Adolescents, Center for Pediatric Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Jörg Hackermüller
- Department of Computational Biology and Chemistry, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
- Department of Computer Science, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- University Hospital for Children and Adolescents, Center for Pediatric Research, Medical Faculty, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG) of the Helmholtz-Centre Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- LIFE–Leipzig Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| |
Collapse
|
3
|
Coate KC, Ramnanan CJ, Smith M, Winnick JJ, Kraft G, Irimia-Dominguez J, Farmer B, Donahue EP, Roach PJ, Cherrington AD, Edgerton DS. Integration of metabolic flux with hepatic glucagon signaling and gene expression profiles in the conscious dog. Am J Physiol Endocrinol Metab 2024; 326:E428-E442. [PMID: 38324258 PMCID: PMC11193521 DOI: 10.1152/ajpendo.00316.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/08/2024]
Abstract
Glucagon rapidly and profoundly stimulates hepatic glucose production (HGP), but for reasons that are unclear, this effect normally wanes after a few hours, despite sustained plasma glucagon levels. This study characterized the time course of glucagon-mediated molecular events and their relevance to metabolic flux in the livers of conscious dogs. Glucagon was either infused into the hepato-portal vein at a sixfold basal rate in the presence of somatostatin and basal insulin, or it was maintained at a basal level in control studies. In one control group, glucose remained at basal, whereas in the other, glucose was infused to match the hyperglycemia that occurred in the hyperglucagonemic group. Elevated glucagon caused a rapid (30 min) and largely sustained increase in hepatic cAMP over 4 h, a continued elevation in glucose-6-phosphate (G6P), and activation and deactivation of glycogen phosphorylase and synthase activities, respectively. Net hepatic glycogenolysis increased rapidly, peaking at 15 min due to activation of the cAMP/PKA pathway, then slowly returned to baseline over the next 3 h in line with allosteric inhibition by glucose and G6P. Glucagon's stimulatory effect on HGP was sustained relative to the hyperglycemic control group due to continued PKA activation. Hepatic gluconeogenic flux did not increase due to the lack of glucagon's effect on substrate supply to the liver. Global gene expression profiling highlighted glucagon-regulated activation of genes involved in cellular respiration, metabolic processes, and signaling, as well as downregulation of genes involved in extracellular matrix assembly and development.NEW & NOTEWORTHY Glucagon rapidly stimulates hepatic glucose production, but these effects are transient. This study links the molecular and metabolic flux changes that occur in the liver over time in response to a rise in glucagon, demonstrating the strength of the dog as a translational model to couple findings in small animals and humans. In addition, this study clarifies why the rapid effects of glucagon on liver glycogen metabolism are not sustained.
Collapse
Affiliation(s)
- Katie C Coate
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Christopher J Ramnanan
- Department of Innovation in Medical Education, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Marta Smith
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Jason J Winnick
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States
| | - Guillaume Kraft
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Jose Irimia-Dominguez
- Department of Molecular and Cellular Endocrinology, Beckman Research Institute, Duarte, California, United States
| | - Ben Farmer
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - E Patrick Donahue
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Peter J Roach
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Dale S Edgerton
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| |
Collapse
|
4
|
Coate KC, Ramnanan CJ, Smith M, Winnick JJ, Kraft G, Irimia JM, Farmer B, Donahue P, Roach PJ, Cherrington AD, Edgerton DS. Integration of metabolic flux with hepatic glucagon signaling and gene expression profiles in the conscious dog. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559999. [PMID: 37808670 PMCID: PMC10557670 DOI: 10.1101/2023.09.28.559999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Glucagon rapidly and profoundly simulates hepatic glucose production (HGP), but for reasons which are unclear, this effect normally wanes after a few hours, despite sustained plasma glucagon levels. This study characterized the time course and relevance (to metabolic flux) of glucagon mediated molecular events in the livers of conscious dogs. Glucagon was either infused into the hepato-portal vein at a 6-fold basal rate in the presence of somatostatin and basal insulin, or it was maintained at a basal level in control studies. In one control group glucose remained at basal while in the other glucose was infused to match the hyperglycemia that occurred in the hyperglucagonemic group. Elevated glucagon caused a rapid (30 min) but only partially sustained increase in hepatic cAMP over 4h, a continued elevation in G6P, and activation and deactivation of glycogen phosphorylase and synthase activities, respectively. Net hepatic glycogenolysis and HGP increased rapidly, peaking at 30 min, then returned to baseline over the next 3h (although glucagons stimulatory effect on HGP was sustained relative to the hyperglycemic control group). Hepatic gluconeogenic flux did not increase due to lack of glucagon effect on substrate supply to the liver. Global gene expression profiling highlighted glucagon-regulated activation of genes involved in cellular respiration, metabolic processes, and signaling, and downregulation of genes involved in extracellular matrix assembly and development.
Collapse
|
5
|
Novikoff A, Müller TD. The molecular pharmacology of glucagon agonists in diabetes and obesity. Peptides 2023; 165:171003. [PMID: 36997003 PMCID: PMC10265134 DOI: 10.1016/j.peptides.2023.171003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Within recent decades glucagon receptor (GcgR) agonism has drawn attention as a therapeutic tool for the treatment of type 2 diabetes and obesity. In both mice and humans, glucagon administration enhances energy expenditure and suppresses food intake suggesting a promising metabolic utility. Therefore synthetic optimization of glucagon-based pharmacology to further resolve the physiological and cellular underpinnings mediating these effects has advanced. Chemical modifications to the glucagon sequence have allowed for greater peptide solubility, stability, circulating half-life, and understanding of the structure-function potential behind partial and "super"-agonists. The knowledge gained from such modifications has provided a basis for the development of long-acting glucagon analogues, chimeric unimolecular dual- and tri-agonists, and novel strategies for nuclear hormone targeting into glucagon receptor-expressing tissues. In this review, we summarize the developments leading toward the current advanced state of glucagon-based pharmacology, while highlighting the associated biological and therapeutic effects in the context of diabetes and obesity.
Collapse
Affiliation(s)
- Aaron Novikoff
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
6
|
Caruso I, Marrano N, Biondi G, Genchi VA, D'Oria R, Sorice GP, Perrini S, Cignarelli A, Natalicchio A, Laviola L, Giorgino F. Glucagon in type 2 diabetes: Friend or foe? Diabetes Metab Res Rev 2023; 39:e3609. [PMID: 36637256 DOI: 10.1002/dmrr.3609] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/02/2022] [Accepted: 12/30/2022] [Indexed: 01/14/2023]
Abstract
Hyperglucagonemia is one of the 'ominous' eight factors underlying the pathogenesis of type 2 diabetes (T2D). Glucagon is a peptide hormone involved in maintaining glucose homoeostasis by increasing hepatic glucose output to counterbalance insulin action. Long neglected, the introduction of dual and triple agonists exploiting glucagon signalling pathways has rekindled the interest in this hormone beyond its classic effect on glycaemia. Glucagon can promote weight loss by regulating food intake, energy expenditure, and brown and white adipose tissue functions through mechanisms still to be fully elucidated, thus its role in T2D pathogenesis should be further investigated. Moreover, the role of glucagon in the development of T2D micro- and macro-vascular complications is elusive. Mounting evidence suggests its beneficial effect in non-alcoholic fatty liver disease, while few studies postulated its favourable role in peripheral neuropathy and retinopathy. Contrarily, glucagon receptor agonism might induce renal changes resembling diabetic nephropathy, and data concerning glucagon actions on the cardiovascular system are conflicting. This review aims to summarise the available findings on the role of glucagon in the pathogenesis of T2D and its complications. Further experimental and clinical data are warranted to better understand the implications of glucagon signalling modulation with new antidiabetic drugs.
Collapse
Affiliation(s)
- Irene Caruso
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Nicola Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppina Biondi
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Valentina Annamaria Genchi
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Rossella D'Oria
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Gian Pio Sorice
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Sebastio Perrini
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Angelo Cignarelli
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Luigi Laviola
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
7
|
Jia Y, Liu Y, Feng L, Sun S, Sun G. Role of Glucagon and Its Receptor in the Pathogenesis of Diabetes. Front Endocrinol (Lausanne) 2022; 13:928016. [PMID: 35784565 PMCID: PMC9243425 DOI: 10.3389/fendo.2022.928016] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/13/2022] [Indexed: 11/24/2022] Open
Abstract
Various theories for the hormonal basis of diabetes have been proposed and debated over the past few decades. Insulin insufficiency was previously regarded as the only hormone deficiency directly leading to metabolic disorders associated with diabetes. Although glucagon and its receptor are ignored in this framework, an increasing number of studies have shown that they play essential roles in the development and progression of diabetes. However, the molecular mechanisms underlying the effects of glucagon are still not clear. In this review, recent research on the mechanisms by which glucagon and its receptor contribute to the pathogenesis of diabetes as well as correlations between GCGR mutation rates in populations and the occurrence of diabetes are summarized. Furthermore, we summarize how recent research clearly establishes glucagon as a potential therapeutic target for diabetes.
Collapse
Affiliation(s)
- Yunbo Jia
- Innovative Engineering Technology Research Center for Cell Therapy, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- Innovative Engineering Technology Research Center for Cell Therapy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Linlin Feng
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Siyu Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Siyu Sun, ; Guangwei Sun,
| | - Guangwei Sun
- Innovative Engineering Technology Research Center for Cell Therapy, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Siyu Sun, ; Guangwei Sun,
| |
Collapse
|
8
|
A Pilot Proteomic Study of Normal Human Tears: Leptin as a Potential Biomarker of Metabolic Disorders. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11125755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The concentrations of insulin, leptin, active ghrelin, C-peptide and gastric inhibitory polypeptide (GIP) and their inter-day variations were examined in normal human tears. In addition, correlations between the concentrations of these metabolic proteins and ocular surface parameters were determined. Subjects with healthy ocular surfaces attended three visits, with 7-day intervals. Tear evaporation rate (TER) and non-invasive tear break-up time (NITBUT) were assessed, and a total of 2 µL tears were collected from all subjects. Tear fluid concentrations of insulin, leptin, active ghrelin, C-peptide and GIP were measured by multiplex bead analysis. Insulin was the most highly expressed metabolic protein, followed by leptin, C-peptide, active ghrelin and GIP. Of these, only active ghrelin had a significant inter-day variation (p < 0.05). There was no inter-day variation in the mean concentrations of the other metabolic proteins. Leptin had a strong intra-class reproducibility. No correlation was detected between tear metabolic protein concentrations and ocular surface parameters. This pilot study shows, for the first time, that active ghrelin and GIP are detectable in healthy tears. The strong intra-class reproducibility for leptin shows that it could be used as a potential tear fluid biomarker and, possibly, in determining the effects of metabolic disorders on the ocular surface.
Collapse
|
9
|
Meister J, Wang L, Pydi SP, Wess J. Chemogenetic approaches to identify metabolically important GPCR signaling pathways: Therapeutic implications. J Neurochem 2021; 158:603-620. [PMID: 33540469 DOI: 10.1111/jnc.15314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 12/16/2022]
Abstract
DREADDs (Designer Receptors Exclusively Activated by a Designer Drug) are designer G protein-coupled receptors (GPCRs) that are widely used in the neuroscience field to modulate neuronal activity. In this review, we will focus on DREADD studies carried out with genetically engineered mice aimed at elucidating signaling pathways important for maintaining proper glucose and energy homeostasis. The availability of muscarinic receptor-based DREADDs endowed with selectivity for one of the four major classes of heterotrimeric G proteins (Gs , Gi , Gq , and G12 ) has been instrumental in dissecting the physiological and pathophysiological roles of distinct G protein signaling pathways in metabolically important cell types. The novel insights gained from this work should inform the development of novel classes of drugs useful for the treatment of several metabolic disorders including type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| |
Collapse
|
10
|
Vincent RK, Williams DM, Evans M. A look to the future in non-alcoholic fatty liver disease: Are glucagon-like peptide-1 analogues or sodium-glucose co-transporter-2 inhibitors the answer? Diabetes Obes Metab 2020; 22:2227-2240. [PMID: 32945071 DOI: 10.1111/dom.14196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/19/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022]
Abstract
The increasing prevalence of diabetes and non-alcoholic fatty liver disease (NAFLD) is a growing public health concern associated with significant morbidity, mortality and economic cost, particularly in those who progress to cirrhosis. Medical treatment is frequently limited, with no specific licensed treatments currently available for people with NAFLD. Its association with diabetes raises the possibility of shared mechanisms of disease progression and treatment. With the ever-growing interest in the non-glycaemic effects of diabetes medications, studies and clinical trials have investigated hepatic outcomes associated with the use of drug classes used for people with type 2 diabetes (T2D), such as glucagon-like peptide-1 (GLP-1) analogues or sodium-glucose co-transporter-2 (SGLT2) inhibitors. Studies exploring the use of GLP-1 analogues or SGLT2 inhibitors in people with NAFLD have observed improved measures of hepatic inflammation, liver enzymes and radiological features over short periods. However, these studies tend to have variable study populations and inconsistent reported outcomes, limiting comparison between drugs and drug classes. As these drugs appear to improve biomarkers of NAFLD, clinicians should consider their use in patients with NAFLD and T2D. However, further evidence with greater participant numbers and longer trial durations is required to support specific licensing for people with NAFLD. Larger trials would allow reporting of major adverse hepatic events, akin to cardiovascular and renal outcome trials, to be determined. This would provide a more meaningful evaluation of the impact of these drugs in NAFLD. Nevertheless, these drugs represent a future potential therapeutic avenue in this difficult-to-treat population and may beget significant health and economic impacts.
Collapse
Affiliation(s)
- Rebecca K Vincent
- Department of Gastroenterology, University Hospital Llandough, Cardiff, UK
| | - David M Williams
- Department of Diabetes and Endocrinology, University Hospital Llandough, Cardiff, UK
| | - Marc Evans
- Department of Diabetes and Endocrinology, University Hospital Llandough, Cardiff, UK
| |
Collapse
|
11
|
Baggio LL, Drucker DJ. Glucagon-like peptide-1 receptor co-agonists for treating metabolic disease. Mol Metab 2020; 46:101090. [PMID: 32987188 PMCID: PMC8085566 DOI: 10.1016/j.molmet.2020.101090] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022] Open
Abstract
Background Glucagon-like peptide-1 receptor (GLP-1R) agonists are approved to treat type 2 diabetes and obesity. They elicit robust improvements in glycemic control and weight loss, combined with cardioprotection in individuals at risk of or with pre-existing cardiovascular disease. These attributes make GLP-1 a preferred partner for next-generation therapies exhibiting improved efficacy yet retaining safety to treat diabetes, obesity, non-alcoholic steatohepatitis, and related cardiometabolic disorders. The available clinical data demonstrate that the best GLP-1R agonists are not yet competitive with bariatric surgery, emphasizing the need to further improve the efficacy of current medical therapy. Scope of review In this article, we discuss data highlighting the physiological and pharmacological attributes of potential peptide and non-peptide partners, exemplified by amylin, glucose-dependent insulinotropic polypeptide (GIP), and steroid hormones. We review the progress, limitations, and future considerations for translating findings from preclinical experiments to competitive efficacy and safety in humans with type 2 diabetes and obesity. Major conclusions Multiple co-agonist combinations exhibit promising clinical efficacy, notably tirzepatide and investigational amylin combinations. Simultaneously, increasing doses of GLP-1R agonists such as semaglutide produces substantial weight loss, raising the bar for the development of new unimolecular co-agonists. Collectively, the available data suggest that new co-agonists with robust efficacy should prove superior to GLP-1R agonists alone to treat metabolic disorders. GLP-1 is a preferred partner for co-agonist development. Co-agonist combinations must exhibit improved weight loss beyond GLP-1 alone. Unimolecular coagonists must exhibit retained or improved cardioprotection. Obesity represents an optimal condition for the development of new GLP-1 co-agonists.
Collapse
Affiliation(s)
- Laurie L Baggio
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mt. Sinai Hospital, Toronto, Ontario, M5G 1X5 Canada
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mt. Sinai Hospital, Toronto, Ontario, M5G 1X5 Canada.
| |
Collapse
|
12
|
Wang L, Zhu L, Meister J, Bone DBJ, Pydi SP, Rossi M, Wess J. Use of DREADD Technology to Identify Novel Targets for Antidiabetic Drugs. Annu Rev Pharmacol Toxicol 2020; 61:421-440. [PMID: 32746768 DOI: 10.1146/annurev-pharmtox-030220-121042] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
G protein-coupled receptors (GPCRs) form a superfamily of plasma membrane receptors that couple to four major families of heterotrimeric G proteins, Gs, Gi, Gq, and G12. GPCRs represent excellent targets for drug therapy. Since the individual GPCRs are expressed by many different cell types, the in vivo metabolic roles of a specific GPCR expressed by a distinct cell type are not well understood. The development of designer GPCRs known as DREADDs (designer receptors exclusively activated by a designer drug) that selectively couple to distinct classes of heterotrimeric G proteins has greatly facilitated studies in this area. This review focuses on the use of DREADD technology to explore the physiological and pathophysiological roles of distinct GPCR/G protein cascades in several metabolically important cell types. The novel insights gained from these studies should stimulate the development of GPCR-based treatments for major metabolic diseases such as type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Derek B J Bone
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Mario Rossi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA;
| |
Collapse
|
13
|
Ng JCM, Schooling CM. Effect of Glucagon on Ischemic Heart Disease and Its Risk Factors: A Mendelian Randomization Study. J Clin Endocrinol Metab 2020; 105:5837127. [PMID: 32407514 DOI: 10.1210/clinem/dgaa259] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/08/2020] [Indexed: 01/10/2023]
Abstract
CONTEXT Glucagon acts reciprocally with insulin to regular blood glucose. However, the effect of glucagon on cardiovascular disease has not been widely studied. It has been suggested that insulin may increase the risk of ischemic heart disease. OBJECTIVE To investigate whether glucagon, the main counteracting hormone of insulin, plays a role in development of ischemic heart disease. DESIGN, SETTING, AND PARTICIPANTS In this 2-sample Mendelian randomization study, we estimated the causal effect of glucagon on ischemic heart disease and its risk factors using the inverse-variance weighted method with multiplicative random effects and multiple sensitivity analyses. Genetic associations with glucagon and ischemic heart disease and its risk factors, including type 2 diabetes and fasting insulin, were obtained from publicly available genome-wide association studies. MAIN OUTCOME MEASURE Odds ratio for ischemic heart disease and its risk factors per 1 standard deviation change in genetically predicted glucagon. RESULTS Twenty-four single-nucleotide polymorphisms strongly (P < 5 × 10-6) and independently (r2 < 0.05) predicting glucagon were obtained. Genetically predicted higher glucagon was associated with an increased risk of ischemic heart disease (inverse-variance weighted odds ratio, 1.03; 95% confidence interval, 1.0003-1.05) but not with type 2 diabetes (inverse-variance weighted odds ratio, 0.998, 95% confidence interval, 0.97-1.03), log-transformed fasting insulin (inverse-variance weighted beta, 0.002, 95% confidence interval, -0.01 to 0.01), other glycemic traits, blood pressure, reticulocyte, or lipids. CONCLUSION Glucagon might have an adverse impact on ischemic heart disease. Relevance of the underlying pathway to existing and potential interventions should be investigated.
Collapse
Affiliation(s)
- Jack C M Ng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Graduate School of Public Health and Health Policy, The City University of New York, New York, USA
| |
Collapse
|
14
|
Activation of SIK1 by phanginin A inhibits hepatic gluconeogenesis by increasing PDE4 activity and suppressing the cAMP signaling pathway. Mol Metab 2020; 41:101045. [PMID: 32599076 PMCID: PMC7381706 DOI: 10.1016/j.molmet.2020.101045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/06/2020] [Accepted: 06/22/2020] [Indexed: 12/25/2022] Open
Abstract
Objective Salt-induced kinase 1 (SIK1) acts as a key modulator in many physiological processes. However, the effects of SIK1 on gluconeogenesis and the underlying mechanisms have not been fully elucidated. In this study, we found that a natural compound phanginin A could activate SIK1 and further inhibit gluconeogenesis. The mechanisms by which phanginin A activates SIK1 and inhibits gluconeogenesis were explored in primary mouse hepatocytes, and the effects of phanginin A on glucose homeostasis were investigated in ob/ob mice. Methods The effects of phanginin A on gluconeogenesis and SIK1 phosphorylation were examined in primary mouse hepatocytes. Pan-SIK inhibitor and siRNA-mediated knockdown were used to elucidate the involvement of SIK1 activation in phanginin A-reduced gluconeogenesis. LKB1 knockdown was used to explore how phanginin A activated SIK1. SIK1 overexpression was used to evaluate its effect on gluconeogenesis, PDE4 activity, and the cAMP pathway. The acute and chronic effects of phanginin A on metabolic abnormalities were observed in ob/ob mice. Results Phanginin A significantly increased SIK1 phosphorylation through LKB1 and further suppressed gluconeogenesis by increasing PDE4 activity and inhibiting the cAMP/PKA/CREB pathway in primary mouse hepatocytes, and this effect was blocked by pan-SIK inhibitor HG-9-91-01 or siRNA-mediated knockdown of SIK1. Overexpression of SIK1 in hepatocytes increased PDE4 activity, reduced cAMP accumulation, and thereby inhibited gluconeogenesis. Acute treatment with phanginin A reduced gluconeogenesis in vivo, accompanied by increased SIK1 phosphorylation and PDE4 activity in the liver. Long-term treatment of phanginin A profoundly reduced blood glucose levels and improved glucose tolerance and dyslipidemia in ob/ob mice. Conclusion We discovered an unrecognized effect of phanginin A in suppressing hepatic gluconeogenesis and revealed a novel mechanism that activation of SIK1 by phanginin A could inhibit gluconeogenesis by increasing PDE4 activity and suppressing the cAMP/PKA/CREB pathway in the liver. We also highlighted the potential value of phanginin A as a lead compound for treating type 2 diabetes. Phanginin A inhibits gluconeogenesis in primary mouse hepatocytes. Phanginin A increases hepatic SIK1 phosphorylation both in vitro and in vivo. Activation of SIK1 increases PDE4 activity and suppresses the cAMP signaling pathway. Activation of SIK1 inhibits gluconeogenesis by regulating the PDE4/cAMP/PKA/CREB pathway. Phanginin A improves metabolic disorders in ob/ob mice.
Collapse
|
15
|
Ban Q, Cheng J, Sun X, Jiang Y, Zhao S, Song X, Guo M. Effects of a synbiotic yogurt using monk fruit extract as sweetener on glucose regulation and gut microbiota in rats with type 2 diabetes mellitus. J Dairy Sci 2020; 103:2956-2968. [PMID: 32089310 DOI: 10.3168/jds.2019-17700] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/18/2019] [Indexed: 12/25/2022]
Abstract
We developed a synbiotic yogurt using monk fruit extract as a sweetener and investigated the effects of feeding the yogurt to rats with type 2 diabetes induced by streptozotocin and a high-fat diet. The rats fed the synbiotic yogurt showed greater blood glucose regulation and a significant decrease in insulin resistance and glycosylated hemoglobin compared with rats fed yogurt sweetened with sucrose, and they showed a remarkable improvement in short-chain fatty acid levels and gut microbiota status. Liver and kidney damage was also ameliorated in the rats fed the synbiotic yogurt. Immunohistochemistry analysis showed that the synbiotic yogurt inhibited β-cell loss compared with the control yogurt. Consuming the synbiotic yogurt helped to restore the islets of Langerhans. Our results indicated that monk fruit extract may be a good alternative to sucrose for synbiotic yogurt products in people with type 2 diabetes to delay the progression of diabetes and associated complications.
Collapse
Affiliation(s)
- Qingfeng Ban
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
| | - Jianjun Cheng
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Xiaomeng Sun
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China
| | - Yunqing Jiang
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
| | - Shanbo Zhao
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
| | - Xiao Song
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
| | - Mingruo Guo
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin, 150030, China; Department of Nutrition and Food Sciences, College of Agriculture and Life Sciences, University of Vermont, Burlington 05405.
| |
Collapse
|
16
|
Liu Y, Yang L, Zhang Y, Liu X, Wu Z, Gilbert RG, Deng B, Wang K. Dendrobium officinale polysaccharide ameliorates diabetic hepatic glucose metabolism via glucagon-mediated signaling pathways and modifying liver-glycogen structure. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112308. [PMID: 31622745 DOI: 10.1016/j.jep.2019.112308] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/24/2019] [Accepted: 10/12/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dendrobium officinale polysaccharide (DOP) is the main active ingredient of Dendrobium officinale Kimura & Migo, which is a precious traditional Chinese medicine and often used in treatment of hepatitis, diabetes, obesity and rheumatoid arthritis. AIM OF THE STUDY DOP exhibits significant hypoglycemic activity, while its mechanism remains unclear. The present study aims to investigate the hypoglycemic mechanisms of DOP based on the glucagon-mediated signaling pathways and the liver glycogen structure, which catalyze hepatic glucose metabolism, and provide new knowledge about the antidiabetic mechanism of DOP and further evidence for its clinical use for diabetes. MATERIALS AND METHODS DOP were obtained from the dry stems of Dendrobium officinale by water extraction and alcohol precipitation method. T2DM mice model was established by high-fat diet combined with streptozotocin. Liver histopathological changes were observed by H&E and PAS straining. Pancreatic histology was studied by H&E staining and immunofluorescence analysis. The levels of glucagon and insulin were detected by Elisa Kit and the hepatic glycogen content was detected by GOPOD. The expressions of the hepatic glycogen-related metabolism enzymes, hepatic gluconeogenesis enzymes, and the related protein in cAMP-PKA and Akt/FoxO1 signaling pathways were detected by western blots. Liver glycogen was extracted from the liver tissues by sucrose density gradient centrifugation, and size exclusion chromatography (SEC) was used to analyze the structure of liver glycogen. RESULTS DOP could significantly affect the glucagon-mediated signaling pathways, cAMP-PKA and Akt/FoxO1, to further promote hepatic glycogen synthesis, inhibit hepatic glycogen degradation and hepatic gluconeogenesis. Moreover, DOP could reverse the instability of the liver glycogen structure and thus probably suppressed glycogen degradation. Thus, DOP finally would ameliorate hepatic glucose metabolism via glucagon-mediated signaling pathways and modifying liver-glycogen structure in diabetic mice. CONCLUSIONS The hypoglycemic mechanism of DOP might be associated with the regulation of glucagon-mediated hepatic glycogen metabolism and gluconeogenesis, and of liver glycogen structure, contributing to improved hepatic glucose metabolism in diabetic mice.
Collapse
Affiliation(s)
- Yage Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Linlin Yang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1227, Jiefang Road, 430030, Wuhan, China
| | - Xiaocui Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Zhijing Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1227, Jiefang Road, 430030, Wuhan, China
| | - Robert G Gilbert
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, College of Agriculture, Yangzhou University, 225009, Yangzhou, Jiangsu Province, China; Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Bin Deng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1227, Jiefang Road, 430030, Wuhan, China.
| | - Kaiping Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| |
Collapse
|
17
|
Belete TM. A Recent Achievement In the Discovery and Development of Novel Targets for the Treatment of Type-2 Diabetes Mellitus. J Exp Pharmacol 2020; 12:1-15. [PMID: 32021494 PMCID: PMC6959499 DOI: 10.2147/jep.s226113] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes (T2DM) is a chronic metabolic disorder. Impaired insulin secretion, enhanced hepatic glucose production, and suppressed peripheral glucose use are the main defects responsible for developing the disease. Besides, the pathophysiology of T2DM also includes enhanced glucagon secretion, decreased incretin secretion, increased renal glucose reabsorption, and adipocyte, and brain insulin resistance. The increasing prevalence of T2DM in the world beseeches an urgent need for better treatment options. The antidiabetic drugs focus on control of blood glucose concentration, but the future treatment goal is to delay disease progression and treatment failure, which causes poorer glycemic regulation. Recent treatment approaches target on several novel pathophysiological defects present in T2DM. Some of the promising novel targets being under clinical development include those that increase insulin sensitization (antagonists of glucocorticoids receptor), decreasing hepatic glucose production (glucagon receptor antagonist, inhibitors of glycogen phosphorylase and fructose-1,6-biphosphatase). This review summarizes studies that are available on novel targets being studied to treat T2DM with an emphasis on the small molecule drug design. The experience gathered from earlier studies and knowledge of T2DM pathways can guide the anti-diabetic drug development toward the discovery of drugs essential to treat T2DM.
Collapse
Affiliation(s)
- Tafere Mulaw Belete
- Department of Pharmacology, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
18
|
Pettus JH, D'Alessio D, Frias JP, Vajda EG, Pipkin JD, Rosenstock J, Williamson G, Zangmeister MA, Zhi L, Marschke KB. Efficacy and Safety of the Glucagon Receptor Antagonist RVT-1502 in Type 2 Diabetes Uncontrolled on Metformin Monotherapy: A 12-Week Dose-Ranging Study. Diabetes Care 2020; 43:161-168. [PMID: 31694861 DOI: 10.2337/dc19-1328] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/15/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Evaluate the safety and efficacy of RVT-1502, a novel oral glucagon receptor antagonist, in subjects with type 2 diabetes inadequately controlled on metformin. RESEARCH DESIGN AND METHODS In a phase 2, double-blind, randomized, placebo-controlled study, subjects with type 2 diabetes (n = 166) on a stable dose of metformin were randomized (1:1:1:1) to placebo or RVT-1502 5, 10, or 15 mg once daily for 12 weeks. The primary end point was change from baseline in HbA1c for each dose of RVT-1502 compared with placebo. Secondary end points included change from baseline in fasting plasma glucose (FPG) and safety assessments. RESULTS Over 12 weeks, RVT-1502 significantly reduced HbA1c relative to placebo by 0.74%, 0.76%, and 1.05% in the 5-, 10-, and 15-mg groups (P < 0.001), respectively, and FPG decreased by 2.1, 2.2, and 2.6 mmol/L (P < 0.001). The proportions of subjects achieving an HbA1c <7.0% were 19.5%, 39.5%, 39.5%, and 45.0% with placebo and RVT-1502 5, 10, and 15 mg (P ≤ 0.02 vs. placebo). The frequency of hypoglycemia was low, and no episodes were severe. Mild increases in mean aminotransferase levels remaining below the upper limit of normal were observed with RVT-1502 but were reversible and did not appear to be dose related, with no other liver parameter changes. Weight and lipid changes were similar between RVT-1502 and placebo. RVT-1502-associated mild increases in blood pressure were not dose related or consistent across time. CONCLUSIONS Glucagon receptor antagonism with RVT-1502 significantly lowers HbA1c and FPG, with a safety profile that supports further clinical development with longer-duration studies (NCT02851849).
Collapse
Affiliation(s)
| | | | | | - Eric G Vajda
- Ligand Pharmaceuticals Incorporated, San Diego, CA
| | | | | | | | | | - Lin Zhi
- Ligand Pharmaceuticals Incorporated, San Diego, CA
| | | |
Collapse
|
19
|
Jia L, Jiang Y, Li X, Chen Z. Purβ promotes hepatic glucose production by increasing Adcy6 transcription. Mol Metab 2019; 31:85-97. [PMID: 31918924 PMCID: PMC6920194 DOI: 10.1016/j.molmet.2019.11.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/03/2019] [Accepted: 11/10/2019] [Indexed: 02/07/2023] Open
Abstract
Objective Enhanced glucagon signaling and hepatic glucose production (HGP) can account for hyperglycemia in patients with obesity and type 2 diabetes. However, the detailed molecular mechanisms underlying the enhanced HGP in these patients are not fully understood. Here, we identify Purβ as a positive regulator of HGP and study its molecular mechanisms in the regulation of HGP both in vivo and in vitro. Methods Adenovirus-mediated knockdown or overexpression of Purβ was performed in either primary hepatocytes or the livers of db/db mice. Glucose metabolism, insulin sensitivity, and HGP were determined by glucose, insulin, and lactate tolerance tests, respectively. Purβ/ADCY6 protein levels, glucagon signaling (p-CREB/CREB), and insulin signaling (p-Akt/Akt) were measured by immunoblotting. Gene expression was measured by RNA-seq and real-time quantitative polymerase chain reaction. Luciferase reporter and chromatin immunoprecipitation assays were used to study the interaction between Purβ and the Adcy6 promoter. Results Purβ was abnormally elevated in obese mice and was also increased under fasting conditions or via the glucagon signaling pathway, which promoted HGP by increasing Adcy6 expression. Liver-specific knockdown of Purβ in db/db mice significantly ameliorated hyperglycemia and glucose intolerance by suppressing the glucagon/ADCY6/cAMP/PKA/CREB signaling pathway. Consistent with this observation, the knockdown of Purβ also inhibited glucose production in isolated primary hepatocytes by inhibiting the glucagon/ADCY6/cAMP/PKA/CREB signaling pathway, whereas the overexpression of Purβ promoted glucose production by activating this signaling pathway. Mechanistically, Purβ directly binds to the promoter of the Adcy6 gene and thereby promotes its transcription. Conclusions Taken together, these results illustrate a new model in which Purβ functions to regulate the glucagon/ADCY6/cAMP/PKA/CREB signaling pathway to help maintain glucose homeostasis. Purβ was identified as a novel positive regulator of hepatic glucose production. Purβ directly binds to the promoter of the Adcy6 gene, inducing its expression and activating the cAMP/PKA/CREB signaling pathway. Liver-specific knockdown of Purβ in db/db mice significantly ameliorates hyperglycemia and glucose intolerance by suppressing the ADCY6/cAMP/PKA/CREB signaling pathway.
Collapse
Affiliation(s)
- Linna Jia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, School of Life Sciences, Northeast Normal University, Changchun, Jilin, 130024, China; HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yunfeng Jiang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
20
|
Zeng Z, Yuan Q, Yu R, Zhang J, Ma H, Chen S. Ameliorative Effects of Probiotic Lactobacillus paracasei NL41 on Insulin Sensitivity, Oxidative Stress, and Beta-Cell Function in a Type 2 Diabetes Mellitus Rat Model. Mol Nutr Food Res 2019; 63:e1900457. [PMID: 31433912 DOI: 10.1002/mnfr.201900457] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/17/2019] [Indexed: 01/31/2023]
Abstract
SCOPE The present study aims to assess the antidiabetic effect of Lactobacillus paracasei strain NL41 and its potential mechanisms in rats with type 2 diabetes mellitus (T2DM) induced by a high-fat diet and low-dose streptozotocin administration (HFD/STZ). METHODS AND RESULTS Eighteen Sprague-Dawley (SD) rats are randomly assigned to three groups: one control, one HFD/STZ model, and one HFD/STZ-Lactobacillus protection group with administration of strain NL41 for 12 weeks. Blood is collected for biochemical parameters analysis and tissue samples for histological analysis. Treatment with strain NL41 results in excellent blood glucose regulation and significantly decreases insulin resistance, and HbA1c, glucagon, and leptin levels, accompanied by remarkable improvement of dyslipidemia and oxidative stress status in the animals. Islets of Langerhans, liver, and kidney are significantly protected in the NL41-treated rats compared to the HFD/STZ-T2DM model rats. Histochemistry shows that strain NL41 inhibits beta-cell loss and alpha-cell expansion, indicating pancreatic islets as the targeted tissues for the primary ameliorative effect of the probiotic strain on HFD/STZ-T2DM rats. Crosstalk between the gut-liver and liver-pancreas endocrine axes is discussed. CONCLUSION Probiotic strain NL41 prevents HFD/STZ-T2DM by decreasing insulin resistance and oxidative stress status, and protecting beta-cell function.
Collapse
Affiliation(s)
- Zhu Zeng
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Biotechnology, Southwest University, Chongqing, 400715, P. R. China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Qipeng Yuan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Rui Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Jinlan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| | - Huiqin Ma
- College of Horticulture, China Agricultural University, Beijing, 100193, P. R. China
| | - Shangwu Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, P. R. China
| |
Collapse
|
21
|
Gao C, Ren SV, Yu J, Baal U, Thai D, Lu J, Zeng C, Yan H, Wang Y. Glucagon Receptor Antagonism Ameliorates Progression of Heart Failure. JACC Basic Transl Sci 2019; 4:161-172. [PMID: 31061918 PMCID: PMC6488764 DOI: 10.1016/j.jacbts.2018.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 02/08/2023]
Abstract
Mice were treated with a fully human monoclonal glucagon receptor antagonistic antibody REMD2.59 following myocardial infarction or pressure overload. REMD2.59 treatment blunted cardiac hypertrophy and fibrotic remodeling, and attenuated contractile dysfunction at 4 weeks after myocardial infarction. In addition, REMD2.59 treatment at the onset of pressure overload significantly suppressed cardiac hypertrophy and chamber dilation with marked preservation of cardiac systolic and diastolic function. Initiation of REMD2.59 treatment 2 weeks after pressure overload significantly blunted the progression of cardiac pathology. These results provide the first in vivo proof-of-concept evidence that glucagon receptor antagonism is a potentially efficacious therapy to ameliorate both onset and progression of heart failure.
Collapse
Affiliation(s)
- Chen Gao
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Address for correspondence: Dr. Yibin Wang or Dr. Chen Gao, Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, 650 Charles E. Young Drive, Room CHS 37-200J, Los Angeles, California 90095.
| | - Shuxun Vincent Ren
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Junyi Yu
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Ulysis Baal
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Dung Thai
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - John Lu
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Hai Yan
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Address for correspondence: Dr. Yibin Wang or Dr. Chen Gao, Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, 650 Charles E. Young Drive, Room CHS 37-200J, Los Angeles, California 90095.
| |
Collapse
|
22
|
Affiliation(s)
- Alessandro Pocai
- Cardiovascular and Metabolism, Janssen Research and Development, Spring House, Pennsylvania
| |
Collapse
|
23
|
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
|
24
|
Gumbiner B, Esteves B, Dell V, Joh T, Garzone PD, Forgie A, Udata C. Single and multiple ascending-dose study of glucagon-receptor antagonist RN909 in type 2 diabetes: a phase 1, randomized, double-blind, placebo-controlled trial. Endocrine 2018; 62:371-380. [PMID: 30203123 DOI: 10.1007/s12020-018-1597-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE This first-in-human study assessed safety, immunogenicity, pharmacokinetics, and pharmacodynamics of RN909, a monoclonal antibody antagonist of the glucagon receptor, in type 2 diabetes (T2DM) subjects. METHODS This study enrolled 84 T2DM subjects receiving stable metformin regimens. Forty-four subjects were randomized to receive single escalating doses of RN909 (0.3 to 6 mg/kg subcutaneously (SC), or 1 mg/kg intravenously (IV)), or placebo; 40 subjects were randomized to receive multiple escalating doses (50 to 150 mg SC) or placebo every 4 weeks for 12 weeks. RESULTS RN909 was well tolerated; treatment-related elevated liver function tests (LFTs) were observed in 4/33 (12.1%) and 5/32 (15.6%) subjects treated with single and multiple doses, respectively, versus 1/10 (10%) and 0 in the respective placebo groups. RN909 dose-normalized AUCinf increased more than dose-proportionally following single SC doses, and after multiple doses, accumulation ratios ranged from 1.3 to 3.4. The incidence of antidrug antibodies (ADA) was 33% after single doses and 50% after multiple doses. RN909 produced dose-dependent, durable fasting plasma glucose (FPG)-lowering at day 29 (mean change -20.6 to -97.5 mg/dL) and day 85 (mean change; -27.2 to -43.5 mg/dL) after single and multiple doses, respectively. HbA1c also was reduced after single (mean change -0.30% to -1.44%), and multiple doses (-0.83% to -1.56%). CONCLUSION RN909 was well tolerated after single and multiple doses in T2DM subjects, with diarrhea and elevated LFTs the most frequent adverse events. The appearance of ADA did not affect pharmacokinetics or efficacy. Robust lowering of FPG and HbA1c was observed.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Blood Glucose/drug effects
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/immunology
- Dose-Response Relationship, Drug
- Double-Blind Method
- Female
- Humans
- Hypoglycemic Agents/administration & dosage
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/pharmacokinetics
- Hypoglycemic Agents/therapeutic use
- Male
- Metformin/therapeutic use
- Middle Aged
- Placebos
- Receptors, Glucagon/antagonists & inhibitors
- Receptors, Glucagon/immunology
- Young Adult
Collapse
Affiliation(s)
- Barry Gumbiner
- Pfizer Inc., 10777 Science Center Dr, San Diego, CA, 92121, USA.
| | - Brooke Esteves
- Pfizer Inc., 7 Shipley Circle, Westford, MA, 01886, USA
- ImmunoGen, Inc., 830 Winter Street, Waltham, MA, 02451, USA
| | - Vanessa Dell
- Pfizer Inc. Maine, 235 E. 42nd Street, New York, NY, 10017, USA
| | - Tenshang Joh
- Pfizer Inc., 10646 Science Center Drive, La Jolla, CA, 92121, USA
| | - Pamela D Garzone
- Pfizer Inc., 230 East Grand Ave, South San Francisco, CA, 94080, USA
| | - Alison Forgie
- Pfizer Inc., 10646 Science Center Drive, La Jolla, CA, 92121, USA
| | | |
Collapse
|
25
|
Wu Y, Pan Q, Yan H, Zhang K, Guo X, Xu Z, Yang W, Qi Y, Guo CA, Hornsby C, Zhang L, Zhou A, Li L, Chen Y, Zhang W, Sun Y, Zheng H, Wondisford F, He L, Guo S. Novel Mechanism of Foxo1 Phosphorylation in Glucagon Signaling in Control of Glucose Homeostasis. Diabetes 2018; 67:2167-2182. [PMID: 30201683 PMCID: PMC6198346 DOI: 10.2337/db18-0674] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022]
Abstract
Dysregulation of hepatic glucose production (HGP) serves as a major underlying mechanism for the pathogenesis of type 2 diabetes. The pancreatic hormone glucagon increases and insulin suppresses HGP, controlling blood glucose homeostasis. The forkhead transcription factor Foxo1 promotes HGP through increasing expression of genes encoding the rate-limiting enzymes responsible for gluconeogenesis. We previously established that insulin suppresses Foxo1 by Akt-mediated phosphorylation of Foxo1 at Ser256 in human hepatocytes. In this study, we found a novel Foxo1 regulatory mechanism by glucagon, which promotes Foxo1 nuclear translocation and stability via cAMP- and protein kinase A-dependent phosphorylation of Foxo1 at Ser276 Replacing Foxo1-S276 with alanine (A) or aspartate (D) to block or mimic phosphorylation, respectively, markedly regulates Foxo1 stability and nuclear localization in human hepatocytes. To establish in vivo function of Foxo1-Ser276 phosphorylation in glucose metabolism, we generated Foxo1-S273A and Foxo1-S273D knock-in (KI) mice. The KI mice displayed impaired blood glucose homeostasis, as well as the basal and glucagon-mediated HGP in hepatocytes. Thus, Foxo1-Ser276 is a new target site identified in the control of Foxo1 bioactivity and associated metabolic diseases.
Collapse
Affiliation(s)
- Yuxin Wu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Quan Pan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Hui Yan
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Kebin Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Xiaoqin Guo
- Department of Endocrinology, Third Military Medical University, Chongqing, China
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Zihui Xu
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Wanbao Yang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yajuan Qi
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Cathy A Guo
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Caitlyn Hornsby
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Lin Zhang
- Department of Chemistry, Cleveland State University, Cleveland, OH
| | - Aimin Zhou
- Department of Chemistry, Cleveland State University, Cleveland, OH
| | - Ling Li
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yunmei Chen
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Weiping Zhang
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yuxiang Sun
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Hongting Zheng
- Department of Endocrinology, Third Military Medical University, Chongqing, China
| | - Fred Wondisford
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Ling He
- Division of Endocrinology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Shaodong Guo
- Department of Nutrition and Food Science, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| |
Collapse
|
26
|
Murata M, Adachi H, Nakade T, Oshima S. Glucagon secretion determined by the RIA method is lower in patients with low left ventricular ejection fraction: The new glass study. Diabetes Res Clin Pract 2018; 144:260-269. [PMID: 30213772 DOI: 10.1016/j.diabres.2018.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/06/2018] [Accepted: 09/03/2018] [Indexed: 10/28/2022]
Abstract
AIMS We investigated the glucagon levels in patients with heart failure (HF), using long oral glucose tolerance test (OGTT). METHODS In this prospective observational study, we enrolled 30 undiagnosed diabetes patients (age 69 ± 10 years, 70% males, HbA1c 43 mmol/mol). A 4-h OGTT was performed. Glucose, insulin, and glucagon (radioimmunoassay [RIA] and sandwich ELISA [S-W] methods) were evaluated during 4-h. We compared glucagon levels between HF and non-HF patients. RESULTS There were 11 HF and 19 non-HF patients. In patients with HF, glucagon (S-W) during 4-h was lower than in patients without HF, with no significant difference. The area under the curve (AUC) of glucagon (RIA) during 4-h was significantly lower among HF patients. Moreover, in patients with reduced left ventricular ejection fraction (LVEF) (<40%), AUC glucagon (RIA) was significantly lower than in patients with non-reduced EF (≥40%). However, there was no difference in glucagon values between the high E/e' (≥13.0) and low E/e' (<13.0) groups. CONCLUSIONS Although glucagon (S-W) showed no significant difference in patients with and without HF, especially reduced LVEF, glucagon (RIA) secretion was significantly lower in HF patients than in patients without HF. It is suggested that low glucagon secretion might be correlated with low EF.
Collapse
Affiliation(s)
- Makoto Murata
- Gunma Prefectural Cardiovascular Center, Department of Cardiology, 3-12 Kameizumimachi, Maebashi, Gunma 371-0004, Japan.
| | - Hitoshi Adachi
- Gunma Prefectural Cardiovascular Center, Department of Cardiology, 3-12 Kameizumimachi, Maebashi, Gunma 371-0004, Japan.
| | - Taishuke Nakade
- Gunma Prefectural Cardiovascular Center, Department of Cardiology, 3-12 Kameizumimachi, Maebashi, Gunma 371-0004, Japan.
| | - Shigeru Oshima
- Gunma Prefectural Cardiovascular Center, Department of Cardiology, 3-12 Kameizumimachi, Maebashi, Gunma 371-0004, Japan.
| |
Collapse
|
27
|
Handgraaf S, Dusaulcy R, Visentin F, Philippe J, Gosmain Y. 17-β Estradiol regulates proglucagon-derived peptide secretion in mouse and human α- and L cells. JCI Insight 2018; 3:98569. [PMID: 29618657 DOI: 10.1172/jci.insight.98569] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/28/2018] [Indexed: 01/11/2023] Open
Abstract
Clinical and experimental data indicate a beneficial effect of estrogens on energy and glucose homeostasis associated with improved insulin sensitivity and positive effects on insulin secretion. The aim of the study was to investigate the impact of estrogens on proglucagon-producing cells, pancreatic α cells, and enteroendocrine L cells. The consequences of sexual hormone deprivation were evaluated in ovariectomized mice (ovx). Ovx mice exhibited impaired glucose tolerance during oral glucose tolerance tests (OGTT), which was associated with decreased GLP-1 intestinal and pancreatic secretion and content, an effect that was reversed by estradiol (E2) treatment. Indeed, E2 increased oral glucose-induced GLP-1 secretion in vivo and GLP-1 secretion from primary culture of mouse and human α cells through the activation of all 3 estrogen receptors (ERs), whereas E2-induced GLP-1 secretion from mouse and human intestinal explants occurred only by ERβ activation. Underlying the implication of ERβ, its selective agonist WAY20070 was able to restore glucose tolerance in ovx mice at least partly through plasma GLP-1 increase. We conclude that E2 directly controls both α- and L cells to increase GLP-1 secretion, in addition to its effects on insulin and glucagon secretion, highlighting the potential beneficial role of the estrogenic pathway and, more particularly, of ERβ agonists to prevent type 2 diabetes.
Collapse
|
28
|
Guzman CB, Duvvuru S, Akkari A, Bhatnagar P, Battioui C, Foster W, Zhang XM, Shankar SS, Deeg MA, Chalasani N, Hardy TA, Kazda CM, Pillai SG. Coding variants in PNPLA3 and TM6SF2 are risk factors for hepatic steatosis and elevated serum alanine aminotransferases caused by a glucagon receptor antagonist. Hepatol Commun 2018; 2:561-570. [PMID: 29761171 PMCID: PMC5944587 DOI: 10.1002/hep4.1171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/05/2018] [Accepted: 02/13/2018] [Indexed: 01/04/2023] Open
Abstract
LY2409021 is a glucagon receptor antagonist that was associated with hepatic steatosis and elevated aminotransferases in phase 2 diabetes studies. We investigated the relationship between selected genetic variants and hepatic steatosis and elevated alanine aminotransferases (ALTs) associated with LY2409021. Patients participated in a 6‐week placebo‐controlled trial (I1R‐MC‐GLDI [GLDI], n = 246) and a 52‐week placebo‐ and active comparator‐controlled trial (I1R‐MC‐GLDJ [GLDJ], n = 158). GLDJ had endpoints at 6 months, including measures of hepatic fat fraction (HFF) by magnetic resonance imaging. The five genes tested were patatin‐like phospholipase domain containing 3 (PNPLA3) (rs738409 and rs738491), transmembrane 6 superfamily member 2 (TM6SF2) (rs58542926), peroxisome proliferative activated receptor gamma coactivator 1 alpha (PPARGC1A) (rs4361373, rs3774921, rs2970849), adenylate cyclase 3 (ADCY3) (rs713586), and insulin‐like growth factor 1 (IGF‐1) (rs1520220). In GLDI, PNPLA3 I148M (P = 0.001) and TM6SF2 E167K (P = 0.001) were significantly associated with an increase in ALT at 6 weeks for LY2409021 but not for placebo. In GLDJ, PNPLA3 I148M showed the same effect (P = 0.007) on ALT at 6 months but the placebo or sitagliptin did not. In GLDJ, both PNPLA3 and TM6SF2 risk‐allele carriers showed increases in HFF that were numerically greater but not statistically significant. The carriers of PNPLA3 and/or TM6SF2 risk alleles showed significantly increased ALT (GLDI, +13.28 U/L in carriers versus +4.84 U/L in noncarriers, P = 4 × 10–5; GLDJ, +14.6 U/L in carriers versus +1.7 in noncarriers, P = 0.0018) and HFF (GLDJ, +5.35% in carriers versus 2.38% in noncarriers, P = 0.048). Elevation of transaminase and HFF were also noted in the noncarriers but at a significantly lower degree. Conclusion: The carriers of PNPLA3 and/or TM6SF2 variant alleles are at risk for hepatic steatosis and elevated ALT levels caused by LY2409021, a glucagon receptor antagonist. More studies are needed to investigate if our observations are generalizable to hepatic steatosis caused by other medications. (Hepatology Communications 2018;2:561‐570)
Collapse
Affiliation(s)
- Cristina B Guzman
- Eli Lilly and Company Indianapolis IN.,Present address: Enanta Pharmaceuticals Inc Watertown MA
| | - Suman Duvvuru
- Eli Lilly and Company Indianapolis IN.,Present address: Amazon Seattle WA
| | | | | | | | | | | | - Sudha S Shankar
- Eli Lilly and Company Indianapolis IN.,Present address: NGM Biopharmaceuticals San Francisco CA
| | - Mark A Deeg
- Eli Lilly and Company Indianapolis IN.,Present address: Regulus Pharmaceuticals San Diego CA
| | - Naga Chalasani
- Indiana University Department of Medicine Indianapolis IN
| | | | | | | |
Collapse
|
29
|
Stachowicz A, Olszanecki R, Suski M, Wiśniewska A, Kuś K, Białas M, Jawień J, Korbut R. Quantitative proteomics reveals decreased expression of major urinary proteins in the liver of apoE/eNOS-DKO mice. Clin Exp Pharmacol Physiol 2018; 45:711-719. [DOI: 10.1111/1440-1681.12927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Aneta Stachowicz
- Chair of Pharmacology; Jagiellonian University Medical College; Krakow Poland
| | - Rafał Olszanecki
- Chair of Pharmacology; Jagiellonian University Medical College; Krakow Poland
| | - Maciej Suski
- Chair of Pharmacology; Jagiellonian University Medical College; Krakow Poland
| | - Anna Wiśniewska
- Chair of Pharmacology; Jagiellonian University Medical College; Krakow Poland
| | - Katarzyna Kuś
- Chair of Pharmacology; Jagiellonian University Medical College; Krakow Poland
| | - Magdalena Białas
- Chair of Pathomorphology; Jagiellonian University Medical College; Krakow Poland
| | - Jacek Jawień
- Chair of Pharmacology; Jagiellonian University Medical College; Krakow Poland
| | - Ryszard Korbut
- Chair of Pharmacology; Jagiellonian University Medical College; Krakow Poland
| |
Collapse
|
30
|
Pedersen C, Porsgaard T, Thomsen M, Rosenkilde MM, Roed NK. Sustained effect of glucagon on body weight and blood glucose: Assessed by continuous glucose monitoring in diabetic rats. PLoS One 2018; 13:e0194468. [PMID: 29558502 PMCID: PMC5860770 DOI: 10.1371/journal.pone.0194468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/02/2018] [Indexed: 11/18/2022] Open
Abstract
Insulin is a vital part of diabetes treatment, whereas glucagon is primarily used to treat insulin-induced hypoglycemia. However, glucagon is suggested to have a central role in the regulation of body weight, which would be beneficial for diabetic patients. Since the glucagon effect on blood glucose is known to be transient, it is relevant to investigate the pharmacodynamics of glucagon after repeated dosing. In the present study, we used telemetry to continuously measure blood glucose in streptozotocin induced diabetic Sprague-Dawley rats. This allowed for a more detailed analysis of glucose regulation compared to intermittent blood sampling. In particular, we evaluated the blood glucose-lowering effect of different insulin doses alone, and in combination with a long acting glucagon analog (LAG). We showed how the effect of the LAG accumulated and persisted over time. Furthermore, we found that addition of the LAG decreased body weight without affecting food intake. In a subsequent study, we focused on the glucagon effect on body weight and food intake during equal glycemic control. In order to obtain comparable maximum blood glucose lowering effect to insulin alone, the insulin dose had to be increased four times in combination with 1 nmol/kg of the LAG. In this set-up the LAG prevented further increase in body weight despite the four times higher insulin-dose. However, the body composition was changed. The insulin group increased both lean and fat mass, whereas the group receiving four times insulin in combination with the LAG only significantly increased the fat mass. No differences were observed in food intake, suggesting a direct effect on energy expenditure by glucagon. Surprisingly, we observed decreased levels of FGF21 in plasma compared to insulin treatment alone. With the combination of insulin and the LAG the blood glucose-lowering effect of insulin was prolonged, which could potentially be beneficial in diabetes treatment.
Collapse
Affiliation(s)
- Christina Pedersen
- Department of GLP-1 & T2D Biology, Novo Nordisk A/S, Maaloev, Denmark
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Trine Porsgaard
- Department of Insulin Pharmacology, Novo Nordisk A/S, Maaloev, Denmark
| | - Maria Thomsen
- Department of Modelling, Novo Nordisk A/S, Maaloev, Denmark
| | - Mette Marie Rosenkilde
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
31
|
Wood MJA, Talbot K, Bowerman M. Spinal muscular atrophy: antisense oligonucleotide therapy opens the door to an integrated therapeutic landscape. Hum Mol Genet 2018; 26:R151-R159. [PMID: 28977438 DOI: 10.1093/hmg/ddx215] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 01/03/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating neuromuscular disorder characterized by loss of spinal cord motor neurons, muscle atrophy and infantile death or severe disability. It is caused by severe reduction of the ubiquitously expressed survival motor neuron (SMN) protein, owing to loss of the SMN1 gene. This would be completely incompatible with survival without the presence of a quasi-identical duplicated gene, SMN2, specific to humans. SMN2 harbours a silent point mutation that favours the production of transcripts lacking exon 7 and a rapidly degraded non-functional SMNΔ7 protein, but from which functional full length SMN protein is produced at very low levels (∼10%). Since the seminal discovery of the SMA-causing gene in 1995, research has focused on the development of various SMN replacement strategies culminating, in December 2016, in the approval of the first precise molecularly targeted therapy for SMA (nusinersen), and a pivotal proof of principle that therapeutic antisense oligonucleotide (ASO) treatment can effectively target the central nervous system (CNS) to treat neurological and neuromuscular disease. Nusinersen is a steric block ASO that binds the SMN2 messenger RNA and promotes exon 7 inclusion and thus increases full length SMN expression. Here, we consider the implications of this therapeutic landmark for SMA therapeutics and discuss how future developments will need to address the challenges of delivering ASO therapies to the CNS, with appropriate efficiency and activity, and how SMN-based therapy should be used in combination with complementary strategies to provide an integrated approach to treat CNS and peripheral pathologies in SMA.
Collapse
Affiliation(s)
- Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford OX1 3QX, Oxford, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Melissa Bowerman
- Department of Physiology, Anatomy and Genetics, University of Oxford OX1 3QX, Oxford, UK
| |
Collapse
|
32
|
Pancreatic alpha cells in diabetic rats express active GLP-1 receptor: Endosomal co-localization of GLP-1/GLP-1R complex functioning through intra-islet paracrine mechanism. Sci Rep 2018; 8:3725. [PMID: 29487355 PMCID: PMC5829082 DOI: 10.1038/s41598-018-21751-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/09/2018] [Indexed: 01/03/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) stimulates insulin secretion from pancreatic beta cells and suppresses glucagon secretion from alpha cells. It remains controversial, however, whether GLP-1 receptor (GLP-1R) is expressed in mature alpha cells. In this study, unlike previous studies using non-diabetic animals, we demonstrated using diabetic model rats and confocal laser scanning microscopy that the GLP-1/GLP-1R complex was located in the endosome of diabetic islets. In addition, we showed that GLP-1 and GLP-1R co-localized with various endosomal markers and adenylate cyclase in the alpha cells of diabetic rats. Diabetic rats had endosomal signaling pathway but normal rats had classical signaling pathway for activated GLP-1R. Furthermore, we performed pancreatic perfusion to assess the functional activity of GLP-1R when stimulated by exendin-4 (EX4). In a pancreas perfusion study, EX4 significantly stimulated glucagon secretion in diabetic rats but not normal rats. However, such glucagon secretion was immediately suppressed, probably due to concomitantly secreted insulin. The GLP-1/GLP-1R complex appears to function through an intra-islet paracrine mechanism in diabetic conditions which could explain, at least in part, the mechanism of paradoxical hyperglucagonaemia in type 2 diabetes.
Collapse
|
33
|
Nunez DJ, D'Alessio D. Glucagon receptor as a drug target: A witches' brew of eye of newt (peptides) and toe of frog (receptors). Diabetes Obes Metab 2018; 20:233-237. [PMID: 28842950 DOI: 10.1111/dom.13102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/16/2022]
MESH Headings
- Animals
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Drugs, Investigational/adverse effects
- Drugs, Investigational/pharmacology
- Drugs, Investigational/therapeutic use
- Glucagon-Like Peptide-1 Receptor/agonists
- Glucagon-Like Peptide-1 Receptor/metabolism
- Humans
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Molecular Targeted Therapy
- Receptors, Glucagon/agonists
- Receptors, Glucagon/antagonists & inhibitors
- Receptors, Glucagon/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Derek J Nunez
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - David D'Alessio
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
34
|
Seghieri M, Christensen AS, Andersen A, Solini A, Knop FK, Vilsbøll T. Future Perspectives on GLP-1 Receptor Agonists and GLP-1/glucagon Receptor Co-agonists in the Treatment of NAFLD. Front Endocrinol (Lausanne) 2018; 9:649. [PMID: 30459715 PMCID: PMC6232120 DOI: 10.3389/fendo.2018.00649] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022] Open
Abstract
Along the obesity pandemic, the prevalence of non-alcoholic fatty liver disease (NAFLD), often regarded as the hepatic manifestation of the metabolic syndrome, increases worldwide representing now the prevalent liver disease in western countries. No pharmacotherapy is approved for the treatment of NAFLD and, currently, the cornerstone treatment is lifestyle modifications focusing on bodyweight loss, notoriously difficult to obtain and even more difficult to maintain. Thus, novel therapeutic approaches are highly demanded. Glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RAs) are approved for the treatment of type 2 diabetes and obesity. They exert their body weight-lowering effect by reducing satiety and food intake. GLP-1RAs have also been shown to reduce liver inflammation and fibrosis. Furthermore, glucagon receptor agonism is being investigated for the treatment of NAFLD due to its appetite and food intake-reducing effects, as well as its ability to increase lipid oxidation and thermogenesis. Recent studies suggest that glucagon receptor signaling is disrupted in NAFLD, indicating that supra-physiological glucagon receptor agonism might represent a new NAFLD treatment target. The present review provides (1) an overview in the pathophysiology of NAFLD, including the potential involvement of GLP-1 and glucagon, (2) an introduction to the currently available GLP-1RAs and (3) outlines the potential of emerging GLP-1RAs and GLP-1/glucagon receptor co-agonists in the treatment of NAFLD.
Collapse
Affiliation(s)
- Marta Seghieri
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alexander S. Christensen
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Andersen
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Filip K. Knop
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Tina Vilsbøll
| |
Collapse
|
35
|
Kim K, Ryu D, Dongiovanni P, Ozcan L, Nayak S, Ueberheide B, Valenti L, Auwerx J, Pajvani UB. Degradation of PHLPP2 by KCTD17, via a Glucagon-Dependent Pathway, Promotes Hepatic Steatosis. Gastroenterology 2017; 153:1568-1580.e10. [PMID: 28859855 PMCID: PMC5705280 DOI: 10.1053/j.gastro.2017.08.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Obesity-induced nonalcoholic fatty liver disease (NAFLD) develops, in part, via excess insulin-stimulated hepatic de novo lipogenesis, which increases, paradoxically, in patients with obesity-induced insulin resistance. Pleckstrin homology domain leucine-rich repeat protein phosphatase 2 (PHLPP2) terminates insulin signaling by dephosphorylating Akt; levels of PHLPP2 are reduced in livers from obese mice. We investigated whether loss of hepatic PHLPP2 is sufficient to induce fatty liver in mice, mechanisms of PHLPP2 degradation in fatty liver, and expression of genes that regulate PHLPP2 in livers of patients with NAFLD. METHODS C57BL/6J mice (controls), obese db/db mice, and mice with liver-specific deletion of PHLPP2 (L-PHLPP2) fed either normal chow or high-fat diet (HFD) were analyzed for metabolic phenotypes, including glucose tolerance and hepatic steatosis. PHLPP2-deficient primary hepatocytes or CRISPR/Cas9-mediated PHLPP2-knockout hepatoma cells were analyzed for insulin signaling and gene expression. We performed mass spectrometry analyses of liver tissues from C57BL/6J mice transduced with Ad-HA-Flag-PHLPP2 to identify posttranslational modifications to PHLPP2 and proteins that interact with PHLPP2. We measured levels of mRNAs by quantitative reverse transcription polymerase chain reaction in liver biopsies from patients with varying degrees of hepatic steatosis. RESULTS PHLPP2-knockout hepatoma cells and hepatocytes from L-PHLPP2 mice showed normal initiation of insulin signaling, but prolonged insulin action. Chow-fed L-PHLPP2 mice had normal glucose tolerance but hepatic steatosis. In HFD-fed C57BL/6J or db/db obese mice, endogenous PHLPP2 was degraded by glucagon and PKA-dependent phosphorylation of PHLPP2 (at Ser1119 and Ser1210), which led to PHLPP2 binding to potassium channel tetramerization domain containing 17 (KCTD17), a substrate-adaptor for Cul3-RING ubiquitin ligases. Levels of KCTD17 mRNA were increased in livers of HFD-fed C57BL/6J or db/db obese mice and in liver biopsies patients with NAFLD, compared with liver tissues from healthy control mice or patients without steatosis. Knockdown of KCTD17 with small hairpin RNA in primary hepatocytes increased PHLPP2 protein but not Phlpp2 mRNA, indicating that KCTD17 mediates PHLPP2 degradation. KCTD17 knockdown in obese mice prevented PHLPP2 degradation and decreased expression of lipogenic genes. CONCLUSIONS In mouse models of obesity, we found that PHLPP2 degradation induced lipogenesis without affecting gluconeogenesis. KCTD17, which is up-regulated in liver tissues of obese mice and patients with NAFLD, binds to phosphorylated PHLPP2 to target it for ubiquitin-mediated degradation; this increases expression of genes that regulate lipogenesis to promote hepatic steatosis. Inhibitors of this pathway might be developed for treatment of patients with NAFLD.
Collapse
Affiliation(s)
- KyeongJin Kim
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Dongryeol Ryu
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland,Department of Korean Medical Science, School of Korean Medicine and Healthy-Aging Korean Medical Research Center, Pusan National University, Republic of Korea
| | - Paola Dongiovanni
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, DEPT, Università degli Studi di Milano, Milano, Italy
| | - Lale Ozcan
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Shruti Nayak
- Proteomics Laboratory, Division of Advanced Research and Technologies, New York University School of Medicine
| | - Beatrix Ueberheide
- Proteomics Laboratory, Division of Advanced Research and Technologies, New York University School of Medicine,Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center
| | - Luca Valenti
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, DEPT, Università degli Studi di Milano, Milano, Italy
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Utpal B. Pajvani
- Department of Medicine, Columbia University, New York, NY 10032, USA
| |
Collapse
|
36
|
Guzman CB, Zhang XM, Liu R, Regev A, Shankar S, Garhyan P, Pillai SG, Kazda C, Chalasani N, Hardy TA. Treatment with LY2409021, a glucagon receptor antagonist, increases liver fat in patients with type 2 diabetes. Diabetes Obes Metab 2017; 19:1521-1528. [PMID: 28371155 DOI: 10.1111/dom.12958] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/15/2017] [Accepted: 03/27/2017] [Indexed: 12/19/2022]
Abstract
AIMS To evaluate whether treatment with LY2409021, a novel, selective glucagon receptor antagonist, is associated with changes in hepatic fat and other safety variables related to the benefit-risk profile for chronic use in patients with type 2 diabetes (T2D). METHODS Safety and efficacy were assessed in patients with T2D taking metformin and sulphonylurea who were randomized to LY2409021 20 mg (n = 65), placebo (n = 68), or sitagliptin 100 mg (n = 41). Key endpoints included change from baseline to month 6 in hepatic fat fraction (HFF), assessed by magnetic resonance imaging; hepatic aminotransferases; blood pressure; lipid profile; fasting plasma glucose; and glycated haemoglobin (HbA1c). RESULTS A significant increase in HFF was seen with LY2409021 vs sitagliptin (least squares [LS] mean difference 3.72%; P < .001) and placebo (4.44%; P < .001), accompanied by significant elevations in alanine aminotransferase levels with LY2409021 vs sitagliptin (6.8 U/L; P = .039) and vs placebo (10.7 U/L; P < .001). No patients had concomitant elevations in bilirubin levels. LY2409021 treatment showed significant HbA1c reductions vs placebo (LS mean difference -0.77%; P < .001) but not sitagliptin (-0.20%; P = .383). Similar results were observed for fasting plasma glucose. LY2409021 was also associated with significant increases in systolic blood pressure vs sitagliptin (4.9 mm Hg; P = .030) and vs placebo (4.3 mm Hg; P = .029), as well as significant increases in body weight and total cholesterol. All effects of LY2409021 were reversible. CONCLUSION In this cohort of patients with T2D, chronic glucagon receptor antagonism with LY2409021 was associated with glucose-lowering but also demonstrated increases in hepatic fat, hepatic aminotransferases, and other adverse effects.
Collapse
Affiliation(s)
| | | | - Rong Liu
- Eli Lilly and Company, Indianapolis, Indiana
| | - Arie Regev
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | - Naga Chalasani
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | | |
Collapse
|
37
|
Scheen AJ, Paquot N, Lefèbvre PJ. Investigational glucagon receptor antagonists in Phase I and II clinical trials for diabetes. Expert Opin Investig Drugs 2017; 26:1373-1389. [PMID: 29052441 DOI: 10.1080/13543784.2017.1395020] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Despite type 2 diabetes (T2D) being recognized as a bihormonal pancreatic disease, current therapies are mainly focusing on insulin, while targeting glucagon has been long dismissed. However, glucagon receptor (GCGr) antagonists are currently investigated in clinical trials. Area covered: Following a brief description of the rationale for antagonizing GCGr in T2D, lessons from GCGr knock-out mice and pharmacological means to antagonize GCGr, a detailed description of the main results obtained with GCGr antagonists in Phase I-II clinical trials is provided. The development of several small molecules has been discontinued, while new ones are currently considered as well as innovative approaches such as monoclonal antibodies or antisense oligonucleotides inhibiting GCGr gene expression. Their potential benefits but also limitations are discussed. Expert opinion: The proof-of-concept that antagonizing GCGr improves glucose control in T2D has been confirmed in humans. Nevertheless, some adverse events led to stopping the development of some of these GCGr antagonists. New approaches seem to have a better benefit/risk balance, although none has progressed to Phase III clinical trials so far. Pharmacotherapy of T2D is becoming a highly competitive field so that GCGr antagonists should provide clear advantages over numerous existing glucose-lowering medications before eventually reaching clinical practice.
Collapse
Affiliation(s)
- André J Scheen
- a Division of Clinical Pharmacology , Center for Interdisciplinary Research on Medicines (CIRM), University of Liège , Belgium.,b Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine , CHU , Liège , Belgium
| | - Nicolas Paquot
- b Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine , CHU , Liège , Belgium
| | - Pierre J Lefèbvre
- b Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine , CHU , Liège , Belgium
| |
Collapse
|
38
|
Kraft G, Coate KC, Winnick JJ, Dardevet D, Donahue EP, Cherrington AD, Williams PE, Moore MC. Glucagon's effect on liver protein metabolism in vivo. Am J Physiol Endocrinol Metab 2017; 313:E263-E272. [PMID: 28536182 PMCID: PMC5625084 DOI: 10.1152/ajpendo.00045.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 11/22/2022]
Abstract
The postprandial state is characterized by a storage of nutrients in the liver, muscle, and adipose tissue for later utilization. In the case of a protein-rich meal, amino acids (AA) stimulate glucagon secretion by the α-cell. The aim of the present study was to determine the impact of the rise in glucagon on AA metabolism, particularly in the liver. We used a conscious catheterized dog model to recreate a postprandial condition using a pancreatic clamp. Portal infusions of glucose, AA, and insulin were used to achieve postprandial levels, while portal glucagon infusion was either maintained at the basal level or increased by three-fold. The high glucagon infusion reduced the increase in arterial AA concentrations compared with the basal glucagon level (-23%, P < 0.05). In the presence of high glucagon, liver AA metabolism shifted toward a more catabolic state with less protein synthesis (-36%) and increased urea production (+52%). Net hepatic glucose uptake was reduced modestly (-35%), and AA were preferentially used in gluconeogenesis, leading to lower glycogen synthesis (-54%). The phosphorylation of AMPK was increased by the high glucagon infusion (+40%), and this could be responsible for increasing the expression of genes related to pathways producing energy and lowering those involved in energy consumption. In conclusion, the rise in glucagon associated with a protein-rich meal promotes a catabolic utilization of AA in the liver, thereby, opposing the storage of AA in proteins.
Collapse
Affiliation(s)
- Guillaume Kraft
- Department of Molecular Physiology and Biophysics,Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Katie C Coate
- Department of Molecular Physiology and Biophysics,Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Jason J Winnick
- Department of Molecular Physiology and Biophysics,Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Dominique Dardevet
- Université Clermont Auvergne, Institut National de la Recherche Agronomique, Unité de Nutrition Humaine, Clermont-Ferrand, France
| | - E Patrick Donahue
- Department of Molecular Physiology and Biophysics,Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Alan D Cherrington
- Department of Molecular Physiology and Biophysics,Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Phillip E Williams
- Department of Molecular Physiology and Biophysics,Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Mary Courtney Moore
- Department of Molecular Physiology and Biophysics,Vanderbilt University School of Medicine, Nashville, Tennessee; and
| |
Collapse
|
39
|
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: 244] [Impact Index Per Article: 30.5] [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
|
40
|
Song W, Cheng D, Hong S, Sappe B, Hu Y, Wei N, Zhu C, O'Connor MB, Pissios P, Perrimon N. Midgut-Derived Activin Regulates Glucagon-like Action in the Fat Body and Glycemic Control. Cell Metab 2017; 25:386-399. [PMID: 28178568 PMCID: PMC5373560 DOI: 10.1016/j.cmet.2017.01.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/03/2016] [Accepted: 01/04/2017] [Indexed: 01/08/2023]
Abstract
While high-caloric diet impairs insulin response to cause hyperglycemia, whether and how counter-regulatory hormones are modulated by high-caloric diet is largely unknown. We find that enhanced response of Drosophila adipokinetic hormone (AKH, the glucagon homolog) in the fat body is essential for hyperglycemia associated with a chronic high-sugar diet. We show that the activin type I receptor Baboon (Babo) autonomously increases AKH signaling without affecting insulin signaling in the fat body via, at least, increase of Akh receptor (AkhR) expression. Further, we demonstrate that Activin-β (Actβ), an activin ligand predominantly produced in the enteroendocrine cells (EEs) of the midgut, is upregulated by chronic high-sugar diet and signals through Babo to promote AKH action in the fat body, leading to hyperglycemia. Importantly, activin signaling in mouse primary hepatocytes also increases glucagon response and glucagon-induced glucose production, indicating a conserved role for activin in enhancing AKH/glucagon signaling and glycemic control.
Collapse
Affiliation(s)
- Wei Song
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Daojun Cheng
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Shangyu Hong
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Benoit Sappe
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Neil Wei
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Changqi Zhu
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Pavlos Pissios
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
41
|
Niclosamide reduces glucagon sensitivity via hepatic PKA inhibition in obese mice: Implications for glucose metabolism improvements in type 2 diabetes. Sci Rep 2017; 7:40159. [PMID: 28054648 PMCID: PMC5214666 DOI: 10.1038/srep40159] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes (T2D) is a global pandemic. Currently, the drugs used to treat T2D improve hyperglycemic symptom of the disease but the underlying mechanism causing the high blood glucose levels have not been fully resolved. Recently published data showed that salt form of niclosamide improved glucose metabolism in high fat fed mice via mitochondrial uncoupling. However, based on our previous work we hypothesised that niclosamide might also improve glucose metabolism via inhibition of the glucagon signalling in liver in vivo. In this study, mice were fed either a chow or high fat diet containing two different formulations of niclosamide (niclosamide ethanolamine salt - NENS or niclosamide - Nic) for 10 weeks. We identified both forms of niclosamide significantly improved whole body glucose metabolism without altering total body weight or body composition, energy expenditure or insulin secretion or sensitivity. Our study provides evidence that inhibition of the glucagon signalling pathway contributes to the beneficial effects of niclosamide (NENS or Nic) on whole body glucose metabolism. In conclusion, our results suggest that the niclosamide could be a useful adjunctive therapeutic strategy to treat T2D, as hepatic glucose output is elevated in people with T2D and current drugs do not redress this adequately.
Collapse
|
42
|
Felix Braga J, Ravizzoni Dartora D, Alenina N, Bader M, Santos RAS. Glucagon-producing cells are increased in Mas-deficient mice. Endocr Connect 2017; 6:27-32. [PMID: 27998954 PMCID: PMC5302165 DOI: 10.1530/ec-16-0098] [Citation(s) in RCA: 6] [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: 12/14/2016] [Accepted: 12/20/2016] [Indexed: 12/31/2022]
Abstract
It has been shown that angiotensin(1-7) (Ang(1-7)) produces several effects related to glucose homeostasis. In this study, we aimed to investigate the effects of genetic deletion of Ang(1-7), the GPCR Mas, on the glucagon-producing cells. C57BL6/N Mas-/- mice presented a significant and marked increase in pancreatic α-cells (number of cells: 146 ± 21 vs 67 ± 8 in WT; P < 0.001) and the percentage per islet (17.9 ± 0.91 vs 12.3 ± 0.9% in WT; P < 0.0001) with subsequent reduction of β-cells percentage (82.1 ± 0.91 vs 87.7 ± 0.9% in WT; P < 0.0001). Accordingly, glucagon plasma levels were increased (516.7 ± 36.35 vs 390.8 ± 56.45 pg/mL in WT; P < 0.05) and insulin plasma levels were decreased in C57BL6/N Mas-/- mice (0.25 ± 0.01 vs 0.31 ± 56.45 pg/mL in WT; P = 0.02). In order to eliminate the possibility of a background-related phenotype, we determined the number of glucagon-producing cells in FVB/N Mas-/- mice. In keeping with the observations in C57BL6/N Mas-/- mice, the number and percentage of pancreatic α-cells were also significantly increased in these mice (number of α-cells: 260 ± 22 vs 156 ± 12 in WT, P < 0.001; percentage per islet: 16 ± 0.8 vs 10 ± 0.5% in WT, P < 0.0001). These results suggest that Mas has a previously unexpected role on the pancreatic glucagon production.
Collapse
Affiliation(s)
- Janaína Felix Braga
- Department of Physiology and BiophysicsNational Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela Ravizzoni Dartora
- Cardiology Institute of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC)Porto Alegre, Rio Grande do Sul, Brazil
| | - Natalia Alenina
- Max-Delbruck Center of Molecular Medicine (MDC)Berlin-Buch, Berlin, Germany
| | - Michael Bader
- Cardiology Institute of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC)Porto Alegre, Rio Grande do Sul, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and BiophysicsNational Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Cardiology Institute of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC)Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
43
|
Vajda EG, Logan D, Lasseter K, Armas D, Plotkin DJ, Pipkin JD, Li YX, Zhou R, Klein D, Wei X, Dilzer S, Zhi L, Marschke KB. Pharmacokinetics and pharmacodynamics of single and multiple doses of the glucagon receptor antagonist LGD-6972 in healthy subjects and subjects with type 2 diabetes mellitus. Diabetes Obes Metab 2017; 19:24-32. [PMID: 27501510 PMCID: PMC5215471 DOI: 10.1111/dom.12752] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/26/2016] [Accepted: 07/27/2016] [Indexed: 01/21/2023]
Abstract
AIM To evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of single and multiple doses of a novel, oral glucagon receptor antagonist, LGD-6972, in healthy subjects and subjects with type 2 diabetes (T2DM). METHODS In the single ascending dose study, LGD-6972 (2-480 mg) was administered to healthy subjects (n = 48) and T2DM subjects (n = 8). In the multiple ascending dose study, healthy subjects (n = 12) received a dose of 15 mg LGD-6972 and T2DM subjects (n = 36) received doses of 5, 10 or 15 mg of LGD-6972 daily for 14 days. RESULTS LGD-6972 had linear plasma pharmacokinetics consistent with once-daily dosing that was comparable in healthy and T2DM subjects. Dose-dependent decreases in fasting plasma glucose were observed in all groups with a maximum of 3.15 mmol/L (56.8 mg/dL) on day 14 in T2DM subjects. LGD-6972 also reduced plasma glucose in the postprandial state. Dose-dependent increases in fasting plasma glucagon were observed, but glucagon levels decreased and insulin levels increased after an oral glucose load in T2DM subjects. LGD-6972 was well tolerated at the doses tested without dose-related or clinically meaningful changes in clinical laboratory parameters. No subject experienced hypoglycaemia. CONCLUSION Inhibition of glucagon action by LGD-6972 was associated with decreases in glucose in both healthy and T2DM subjects, the magnitude of which was sufficient to predict improvement in glycaemic control with longer treatment duration in T2DM patients. The safety and pharmacological profile of LGD-6972 after 14 days of dosing supports continued clinical development.
Collapse
Affiliation(s)
- Eric G. Vajda
- Ligand Pharmaceuticals IncorporatedSan DiegoCaliforniaUSA
| | - Douglas Logan
- Cincinnati VA Medical Center Ringgold Standard InstitutionCincinnatiOhioUSA
| | | | | | | | - JD Pipkin
- Ligand Pharmaceuticals IncorporatedSan DiegoCaliforniaUSA
| | | | | | | | | | - Stacy Dilzer
- Clinical Pharmacology of Miami, Inc.MiamiFloridaUSA
| | - Lin Zhi
- Ligand Pharmaceuticals IncorporatedSan DiegoCaliforniaUSA
| | | |
Collapse
|
44
|
Ceriello A, Genovese S, Mannucci E, Gronda E. Glucagon and heart in type 2 diabetes: new perspectives. Cardiovasc Diabetol 2016; 15:123. [PMID: 27568179 PMCID: PMC5002329 DOI: 10.1186/s12933-016-0440-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/13/2016] [Indexed: 02/06/2023] Open
Abstract
Increased levels of glucagon in type 2 diabetes are well known and, until now, have been considered deleterious. However, glucagon has an important role in the maintenance of both heart and kidney function. Moreover, in the past, glucagon has been therapeutically used for heart failure treatment. The new antidiabetic drugs, dipeptidyl peptidase-4 inhibitors and sodium-glucose co-transporter-2 inhibitors, are able to decrease and to increase glucagon levels, respectively, while contrasting data have been reported regarding the glucagon like peptide 1 receptors agonists. The cardiovascular outcome trials, requested by the FDA, raised some concerns about the possibility that the dipeptidyl peptidase-4 inhibitors can precipitate the heart failure, while, at least for empagliflozin, a positive effect has been shown in decreasing both cardiovascular death and heart failure. The recent LEADER Trial, showed a significant reduction of cardiovascular death with liraglutide, but a neutral effect on heart failure. A possible explanation of the results with the DPPIV inhibitors and empagliflozin might be related to their divergent effect on glucagon levels. Due to unclear effects of glucagon like peptide 1 receptor agonists on glucagon, the possible role of this hormone in the Leader trial remains unclear.
Collapse
Affiliation(s)
- Antonio Ceriello
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomedica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), C/Rosselló, 149-153, 08036 Barcelona, Spain
- Department of Cardiovascular and Metabolic Diseases, IRCCS Multimedica, Sesto San Giovanni, MI Italy
| | - Stefano Genovese
- Department of Cardiovascular and Metabolic Diseases, IRCCS Multimedica, Sesto San Giovanni, MI Italy
| | - Edoardo Mannucci
- Diabetology, Careggi Hospital, University of Florence, Florence, Italy
| | - Edoardo Gronda
- Department of Cardiovascular and Metabolic Diseases, IRCCS Multimedica, Sesto San Giovanni, MI Italy
| |
Collapse
|
45
|
Shu S, Cai X, Li J, Feng Y, Dai A, Wang J, Yang D, Wang MW, Liu H. Design, synthesis, structure–activity relationships, and docking studies of pyrazole-containing derivatives as a novel series of potent glucagon receptor antagonists. Bioorg Med Chem 2016; 24:2852-63. [DOI: 10.1016/j.bmc.2016.04.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 11/28/2022]
|
46
|
Li M, Dean ED, Zhao L, Nicholson WE, Powers AC, Chen W. Glucagon receptor inactivation leads to α-cell hyperplasia in zebrafish. J Endocrinol 2015; 227:93-103. [PMID: 26446275 PMCID: PMC4598637 DOI: 10.1530/joe-15-0284] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glucagon antagonism is a potential treatment for diabetes. One potential side effect is α-cell hyperplasia, which has been noted in several approaches to antagonize glucagon action. To investigate the molecular mechanism of the α-cell hyperplasia and to identify the responsible factor, we created a zebrafish model in which glucagon receptor (gcgr) signaling has been interrupted. The genetically and chemically tractable zebrafish, which provides a robust discovery platform, has two gcgr genes (gcgra and gcgrb) in its genome. Sequence, phylogenetic, and synteny analyses suggest that these are co-orthologs of the human GCGR. Similar to its mammalian counterparts, gcgra and gcgrb are mainly expressed in the liver. We inactivated the zebrafish gcgra and gcgrb using transcription activator-like effector nuclease (TALEN) first individually and then both genes, and assessed the number of α-cells using an α-cell reporter line, Tg(gcga:GFP). Compared to WT fish at 7 days postfertilization, there were more α-cells in gcgra-/-, gcgrb-/-, and gcgra-/-;gcgrb-/- fish and there was an increased rate of α-cell proliferation in the gcgra-/-;gcgrb-/- fish. Glucagon levels were higher but free glucose levels were lower in gcgra-/-, gcgrb-/-, and gcgra-/-;gcgrb-/- fish, similar to Gcgr-/- mice. These results indicate that the compensatory α-cell hyperplasia in response to interruption of glucagon signaling is conserved in zebrafish. The robust α-cell hyperplasia in gcgra-/-;gcgrb-/- larvae provides a platform to screen for chemical and genetic suppressors, and ultimately to identify the stimulus of α-cell hyperplasia and its signaling mechanism.
Collapse
Affiliation(s)
- Mingyu Li
- Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA
| | - E Danielle Dean
- Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA
| | - Liyuan Zhao
- Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA
| | - Wendell E Nicholson
- Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA
| | - Alvin C Powers
- Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA
| | - Wenbiao Chen
- Departments of Molecular Physiology and BiophysicsVanderbilt University School of Medicine, Light Hall, Room 711, 2215 Garland Avenue, Nashville, Tennessee 37232, USADivision of DiabetesEndocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USAThird Institute of OceanographyState Oceanic Administration, Xiamen 361005, ChinaVeterans Affairs Tennessee Valley Healthcare SystemNashville, Tennessee 37212, USA
| |
Collapse
|
47
|
Lefèbvre PJ, Paquot N, Scheen AJ. Inhibiting or antagonizing glucagon: making progress in diabetes care. Diabetes Obes Metab 2015; 17:720-5. [PMID: 25924114 DOI: 10.1111/dom.12480] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/27/2015] [Accepted: 04/27/2015] [Indexed: 01/07/2023]
Abstract
Absolute or relative hyperglucagonaemia has been recognized for years in all experimental or clinical forms of diabetes. It has been suggested that excess secretion of glucagon by the islet α cells is a direct consequence of intra-islet insulin secretory defects. Recent studies have shown that knockout of the glucagon receptor or administration of a monoclonal specific glucagon receptor antibody make insulin-deficient type 1 diabetic rodents thrive without insulin. These observations suggest that glucagon plays an essential role in the pathophysiology of diabetes and that targeting the α cell and glucagon are innovative approaches in the management of diabetes. Despite active research and identification of promising compounds, no one selective glucagon antagonist is presently used in the treatment of diabetes. Interestingly, besides insulin, several drugs used today in the management of diabetes appear to exert their effects, in part, by inhibiting glucagon secretion (glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase-4 inhibitors, α-glucosidase inhibitors and, possibly, sulphonylureas) or glucagon action (metformin). The potential risks associated with total glucagon suppression include α-cell hyperplasia, increased mass of the pancreas, increased susceptibility to hepatosteatosis and hepatocellular injury and increased risk of hypoglycaemia, and these should be considered in the search and development of new compounds reducing glucagon receptor signalling. More than 40 years after its initial description, hyperglucagonaemia in diabetes can no longer be ignored or minimized, and its correction represents an attractive way to improve diabetes management.
Collapse
Affiliation(s)
- P J Lefèbvre
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University of Liège, Liège, Belgium
| | - N Paquot
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University of Liège, Liège, Belgium
| | - A J Scheen
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University of Liège, Liège, Belgium
- Division of Clinical Pharmacology, Department of Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
48
|
Abstract
Plasma hormone peptides, including GLP-1, GIP, Glucagon, and OXM, possess multiple physiological roles and potential therapeutic and diagnostic utility as biomarkers in the research of metabolic disorders. These peptides are subject to proteolytic degradation causing preanalytical variations. Stabilization for accurate quantitation of these active peptides in ex vivo blood specimens is essential for drug and biomarker development. We investigated the protease-driven instability of these peptides in conventional serum, plasma, anticoagulated whole blood, as well as whole blood and plasma stabilized with protease inhibitors. The peptide was monitored by both time-course Matrix-Assisted Laser Desorption Ionization Time-to-Flight Mass Spectrometry (MALDI –TOF MS) and Ab-based assay (ELISA or RIA). MS enabled the identification of proteolytic fragments. In non-stabilized blood samples, the results clearly indicated that dipeptidyl peptidase-IV (DPP-IV) removed the N-terminal two amino acid residues from GLP-1, GIP and OXM(1-37) and not-yet identified peptidase(s) cleave(s) the full-length OXM(1-37) and its fragments. DPP-IV also continued to remove two additional N-terminal residues of processed OXM(3–37) to yield OXM(5–37). Importantly, both DPP-IV and other peptidase(s) activities were inhibited efficiently by the protease inhibitors included in the BD P800* tube. There was preservation of GLP-1, GIP, OXM and glucagon in the P800 plasma samples with half-lives > 96, 96, 72, and 45 hours at room temperature (RT), respectively. In the BD P700* plasma samples, the stabilization of GLP-1 was also achieved with half-life > 96 hours at RT. The stabilization of these variable peptides increased their utility in drug and/or biomarker development. While stability results of GLP-1 obtained with Ab-based assay were consistent with those obtained by MS analysis, the Ab-based results of GIP, Glucagon, and OXM did not reflect the time-dependent degradations revealed by MS analysis. Therefore, we recommended characterizing the degradation of the peptide using the MS-based method when investigating the stability of a specific peptide.
Collapse
Affiliation(s)
- Jizu Yi
- BD Diagnostics, One Becton Drive, Franklin Lakes, NJ, United States of America
- * E-mail: (JY); (DC)
| | - David Warunek
- BD Diagnostics, One Becton Drive, Franklin Lakes, NJ, United States of America
| | - David Craft
- BD Diagnostics, One Becton Drive, Franklin Lakes, NJ, United States of America
- * E-mail: (JY); (DC)
| |
Collapse
|
49
|
Bankir L, Roussel R, Bouby N. Protein- and diabetes-induced glomerular hyperfiltration: role of glucagon, vasopressin, and urea. Am J Physiol Renal Physiol 2015; 309:F2-23. [DOI: 10.1152/ajprenal.00614.2014] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/13/2015] [Indexed: 12/21/2022] Open
Abstract
A single protein-rich meal (or an infusion of amino acids) is known to increase the glomerular filtration rate (GFR) for a few hours, a phenomenon known as “hyperfiltration.” It is important to understand the factors that initiate this upregulation because it becomes maladaptive in the long term. Several mediators and paracrine factors have been shown to participate in this upregulation, but they are not directly triggered by protein intake. Here, we explain how a rise in glucagon and in vasopressin secretion, directly induced by protein ingestion, might be the initial factors triggering the hepatic and renal events leading to an increase in the GFR. Their effects include metabolic actions in the liver and stimulation of sodium chloride reabsorption in the thick ascending limb. Glucagon is not only a glucoregulatory hormone. It is also important for the excretion of nitrogen end products by stimulating both urea synthesis in the liver (along with gluconeogenesis from amino acids) and urea excretion by the kidney. Vasopressin allows the concentration of nitrogenous end products (urea, ammonia, etc.) and other protein-associated wastes in a hyperosmotic urine, thus allowing a very significant water economy characteristic of all terrestrial mammals. No hyperfiltration occurs in the absence of one or the other hormone. Experimental results suggest that the combined actions of these two hormones, along with the complex intrarenal handling of urea, lead to alter the composition of the tubular fluid at the macula densa and to reduce the intensity of the signal activating the tubuloglomerular feedback control of GFR, thus allowing GFR to raise. Altogether, glucagon, vasopressin, and urea contribute to set up the best compromise between efficient urea excretion and water economy.
Collapse
Affiliation(s)
- Lise Bankir
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
| | - Ronan Roussel
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
- Diabétologie Endocrinologie Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France
| | - Nadine Bouby
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Diderot, Sorbonne-Paris-Cité, Paris, France; and
| |
Collapse
|
50
|
Sandoval DA, D'Alessio DA. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease. Physiol Rev 2015; 95:513-48. [PMID: 25834231 DOI: 10.1152/physrev.00013.2014] [Citation(s) in RCA: 324] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preproglucagon gene (Gcg) is expressed by specific enteroendocrine cells (L-cells) of the intestinal mucosa, pancreatic islet α-cells, and a discrete set of neurons within the nucleus of the solitary tract. Gcg encodes multiple peptides including glucagon, glucagon-like peptide-1, glucagon-like peptide-2, oxyntomodulin, and glicentin. Of these, glucagon and GLP-1 have received the most attention because of important roles in glucose metabolism, involvement in diabetes and other disorders, and application to therapeutics. The generally accepted model is that GLP-1 improves glucose homeostasis indirectly via stimulation of nutrient-induced insulin release and by reducing glucagon secretion. Yet the body of literature surrounding GLP-1 physiology reveals an incompletely understood and complex system that includes peripheral and central GLP-1 actions to regulate energy and glucose homeostasis. On the other hand, glucagon is established principally as a counterregulatory hormone, increasing in response to physiological challenges that threaten adequate blood glucose levels and driving glucose production to restore euglycemia. However, there also exists a potential role for glucagon in regulating energy expenditure that has recently been suggested in pharmacological studies. It is also becoming apparent that there is cross-talk between the proglucagon derived-peptides, e.g., GLP-1 inhibits glucagon secretion, and some additive or synergistic pharmacological interaction between GLP-1 and glucagon, e.g., dual glucagon/GLP-1 agonists cause more weight loss than single agonists. In this review, we discuss the physiological functions of both glucagon and GLP-1 by comparing and contrasting how these peptides function, variably in concert and opposition, to regulate glucose and energy homeostasis.
Collapse
Affiliation(s)
- Darleen A Sandoval
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David A D'Alessio
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|