51
|
Winther-Sørensen M, Holst JJ, Wewer Albrechtsen NJ. The feedback cycles between glucose, amino acids and lipids and alpha cell secretion and their role in metabolic fatty liver disease. Curr Opin Lipidol 2023; 34:27-31. [PMID: 36373738 DOI: 10.1097/mol.0000000000000857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE OF REVIEW Glucagon increases hepatic glucose production and in patients with metabolic diseases, glucagon secretion is increased contributing to diabetic hyperglycemia. This review explores the role of amino acids and lipids in the regulation of glucagon secretion and how it may be disturbed in metabolic diseases such as obesity and metabolic associated fatty liver disease (MAFLD). RECENT FINDINGS Human and animal studies have shown that MAFLD is associated with glucagon resistance towards amino acid catabolism, resulting in elevated plasma levels of amino acids. A recent clinical study showed that MAFLD is also associated with glucagon resistance towards lipid metabolism. In contrast, MAFLD may not decrease hepatic sensitivity to the stimulatory effects of glucagon on glucose production. SUMMARY Elevated plasma levels of amino acids and lipids associated with MAFLD may cause diabetogenic hyperglucagonemia. MAFLD and glucagon resistance may therefore be causally linked to hyperglycemia and the development of type 2 diabetes.
Collapse
Affiliation(s)
- Marie Winther-Sørensen
- Department of Biomedical Sciences
- NNF Center for Protein Research, Faculty of Health and Medical Sciences
| | | | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences
- NNF Center for Protein Research, Faculty of Health and Medical Sciences
- Department for Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
52
|
Scheen AJ, Lefèbvre PJ. Glucagon, from past to present: a century of intensive research and controversies. Lancet Diabetes Endocrinol 2023; 11:129-138. [PMID: 36566754 DOI: 10.1016/s2213-8587(22)00349-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022]
Abstract
2022 corresponds to the 100th anniversary of the discovery of glucagon. This TimeCapsule aims to recall the main steps leading to the discovery, characterisation, and clinical importance of the so-called second pancreatic hormone. We describe the early historical findings in basic research (ie, discovery, purification, structure, α-cell origin, radioimmunoassay, glucagon gene [GCG], and glucagon receptor [GLR]), in which three future Nobel Prize laureates were actively involved. Considered as an anti-insulin hormone, glucagon was rapidly used to treat insulin-induced hypoglycaemic coma episodes in people with type 1 diabetes. A key step in the story of glucagon was the discovery of its role and the role of α cells in the physiology and pathophysiology (ie, paracrinopathy) of type 2 diabetes. This concept led to the design of different strategies targeting glucagon, among which GLP-1 receptor (GLP1R) agonists were a major breakthrough, and combination of inhibition of glucagon secretion with stimulation of insulin secretion (both in a glucose-dependent manner). Taking advantage of the glucagon-induced increase in energy metabolism, biased coagonists were developed. Besides the GLP-1 receptor, these coagonists also target the glucagon receptor to further promote weight loss. Thus, the 100-year story of glucagon has most probably not come to an end.
Collapse
Affiliation(s)
- André J Scheen
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of medicine, CHU Liège, Liège University, Liège, Belgium.
| | - Pierre J Lefèbvre
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of medicine, CHU Liège, Liège University, Liège, Belgium
| |
Collapse
|
53
|
Jastreboff AM, Kushner RF. New Frontiers in Obesity Treatment: GLP-1 and Nascent Nutrient-Stimulated Hormone-Based Therapeutics. Annu Rev Med 2023; 74:125-139. [PMID: 36706749 DOI: 10.1146/annurev-med-043021-014919] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Nearly half of Americans are projected to have obesity by 2030, underscoring the pressing need for effective treatments. Glucagon-like peptide 1 receptor agonists (GLP-1 RAs) represent the first agents in a rapidly evolving, highly promising landscape of nascent hormone-based obesity therapeutics. With the understanding of the neurobiology of obesity rapidly expanding, these emerging entero-endocrine and endo-pancreatic agents combined or coformulated with GLP-1 RAs herald a new era of targeted, mechanism-based treatment of obesity. This article reviews GLP-1 RAs in the treatment of obesity and previews the imminent future of nutrient-stimulated hormone-based anti-obesity therapeutics.
Collapse
Affiliation(s)
- Ania M Jastreboff
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Robert F Kushner
- Departments of Medicine (Endocrinology) and Medical Education, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| |
Collapse
|
54
|
Vadmand AC, Nissen AA, Mathiesen S, Soerum ME, Gerbek T, Fridh MK, Sørensen K, Hartmann B, Holst JJ, Müller K. Metabolic Dysregulation in Adult Survivors of Pediatric Hematopoietic Stem Cell Transplantation: The Role of Incretins. J Clin Endocrinol Metab 2023; 108:453-462. [PMID: 36181459 DOI: 10.1210/clinem/dgac561] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/19/2022] [Indexed: 01/20/2023]
Abstract
CONTEXT Survivors of pediatric hematopoietic stem cell transplantation (HSCT) have increased risk of developing metabolic syndrome (MetS), but the mechanisms are poorly understood. OBJECTIVE We aimed to test the hypothesis that insufficient secretion of glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) plays a pathogenetic role in HSCT survivors with MetS. METHODS This cross-sectional cohort study, conducted at the Danish national referral center for HSCT, studied 42 male HSCT survivors (median age 28.9 years) for a median 21.2 years from HSCT, along with 15 age- and sex-matched healthy controls. Main outcome measures were glucose metabolism and incretin hormones (by oral glucose tolerance test [OGTT]) and MetS criteria. The hypothesis was formulated before data collection. RESULTS GLP-1, GIP, and glucagon during an OGTT were similar in patients and controls, with no overall difference between survivors with (24%) and without MetS. However, fasting glucagon was significantly higher in patients with hypertriglyceridemia (mean difference [MD]: 6.1 pmol/L; 95% CI, 1.5-10.8; P = 0.01), and correlated with HDL (MD: 4.7 mmol/L; 95% CI, -0.6 to 9.9; P = 0.08), android-gynoid ratio (correlation coefficient [r] = 0.6, P = 0.0001) and waist-hip ratio (r = 0.5, P = 0.002). A similar pattern was seen for GIP, correlating positively with triglyceride (MD: 60%; 95% CI, 44-82; P = 0.002). GIP levels were significantly increased in patients treated with total body irradiation (TBI) (MD: 165%; 95% CI, 118-230; P = 0.004), which was found to be a significant risk factor for MetS. CONCLUSION This study demonstrates an altered production of incretin hormones in HSCT survivors previously treated with TBI, developing dyslipidemia and abdominal adiposity.
Collapse
Affiliation(s)
- Amalia Christina Vadmand
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Anne Anker Nissen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Sidsel Mathiesen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Maria Ebbesen Soerum
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Tina Gerbek
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Martin Kaj Fridh
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Kaspar Sørensen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Bolette Hartmann
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jens Juul Holst
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
- Institute for Inflammation Research, University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| |
Collapse
|
55
|
GLP-1 Receptor Agonists in Non-Alcoholic Fatty Liver Disease: Current Evidence and Future Perspectives. Int J Mol Sci 2023; 24:ijms24021703. [PMID: 36675217 PMCID: PMC9865319 DOI: 10.3390/ijms24021703] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
To date, non-alcoholic fatty liver disease (NAFLD) is the most frequent liver disease, affecting up to 70% of patients with diabetes. Currently, there are no specific drugs available for its treatment. Beyond their anti-hyperglycemic effect and the surprising role of cardio- and nephroprotection, GLP-1 receptor agonists (GLP-1 RAs) have shown a significant impact on body weight and clinical, biochemical and histological markers of fatty liver and fibrosis in patients with NAFLD. Therefore, GLP-1 RAs could be a weapon for the treatment of both diabetes mellitus and NAFLD. The aim of this review is to summarize the evidence currently available on the role of GLP-1 RAs in the treatment of NAFLD and to hypothesize potential future scenarios.
Collapse
|
56
|
Gao J, Li H, Xu H, Liu Y, Cai M, Shi Y, Zhang J, Wang H. High glucose-induced glucagon resistance and membrane distribution of GCGR revealed by super-resolution imaging. iScience 2023; 26:105967. [PMID: 36824278 PMCID: PMC9941209 DOI: 10.1016/j.isci.2023.105967] [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: 11/22/2022] [Revised: 12/29/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The glucagon receptor (GCGR) is a member of the class B G protein-coupled receptor family. Many research works have been carried out on GCGR structure, glucagon signaling pathway, and GCGR antagonists. However, the expression and fine distribution of GCGR proteins in response to glucagon under high glucose remain unclear. Using direct stochastic optical reconstruction microscopy (dSTORM) imaging, nanoscale GCGR clusters were observed on HepG2 cell membranes, and high glucose promoted GCGR expression and the formation of more and larger clusters. Moreover, glucagon stimulation under high glucose did not inhibit GCGR levels as significantly as that under low glucose and did not increase the downstream cyclic 3,5'-adenosine monophosphate-protein kinase A (cAMP-PKA) signal, and there were still large-size clusters on the membranes, indicating that high glucose induced glucagon resistance. In addition, high glucose induced stronger glucagon resistance in hepatoma cells compared with hepatic cells. Our work will pave a way to further our understanding of the pathogenesis of diabetes and develop more effective drugs targeting GCGR.
Collapse
Affiliation(s)
- Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China,Corresponding author
| | - Hongru Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China,University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Yong Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Yan Shi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Jingrui Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China,University of Science and Technology of China, Hefei, Anhui 230027, China,Laboratory for Marine Biology and Biotechnology, Qing dao National Laboratory for Marine Science and Technology, Wenhai Road, Aoshanwei, Jimo, Qingdao, Shandong 266237, China,Corresponding author
| |
Collapse
|
57
|
Rodgers RL. A reappraisal of the role of cyclic AMP in the physiological action of glucagon. Peptides 2023; 159:170906. [PMID: 36396082 DOI: 10.1016/j.peptides.2022.170906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Effects of the metabolic hormone glucagon can be physiological or supraphysiological, based on agonist concentration and the mediating cellular signal. The threshold concentration (TC) for activating the AC/cAMP signal pathway in liver is ≥ 100 pM. By contrast, mean plasma concentrations are around 20-45 pM, depending on the vascular bed. Accordingly, effects produced at TCs below 100 pM are physiological and mediated by cellular signal pathways other than AC/cAMP. Effects generated at concentrations above 100 pM are supraphysiological, often mediated by simultaneous activation of cAMP-independent and -dependent pathways. Physiological responses, and their established or implicated signal pathways, include stimulation of: glucose mobilization, fatty acid oxidation, and urea synthesis in liver (PLC/IP3/Ca2+/CaM); lipolysis in white and brown adipose tissue and oxygen consumption in brown adipose of the rat but not in humans (PLC/IP3/Ca2+/CaM); renal potassium and phosphate excretion in rodents and GFR in humans (signal undetermined); and glucose utilization in rat heart (PI3K/akt). Supraphysiological responses involve the AC/cAMP pathway and include: enhanced stimulation of glucose mobilization and stimulation of urea synthesis in liver; further stimulation of white and brown adipose lipolysis and thermogenesis in brown adipose tissue; stimulation of renal Cl- transport; and increased rat heart contractility. The AC/cAMP pathway is likely recruited when plasma glucagon rises above 100 pM during periods of elevated metabolic stress and systemic glucose demand, such as in the early neonate or strenuously exercising adult. The current cAMP-centered model should therefore be reconsidered and replaced with one that places more emphasis on cAMP-independent pathways.
Collapse
Affiliation(s)
- Robert L Rodgers
- Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02935, USA.
| |
Collapse
|
58
|
Feris F, McRae A, Kellogg TA, McKenzie T, Ghanem O, Acosta A. Mucosal and hormonal adaptations after Roux-en-Y gastric bypass. Surg Obes Relat Dis 2023; 19:37-49. [PMID: 36243547 PMCID: PMC9797451 DOI: 10.1016/j.soard.2022.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/12/2023]
Abstract
The aim of this study was to perform a comprehensive literature review regarding the relevant hormonal and histologic changes observed after Roux-en-Y gastric bypass (RYGB). We aimed to describe the relevant hormonal (glucagon-like peptides 1 and 2 [GLP-1 and GLP-2], peptide YY [PYY], oxyntomodulin [OXM], bile acids [BA], cholecystokinin [CCK], ghrelin, glucagon, gastric inhibitory polypeptide [GIP], and amylin) profiles, as well as the histologic (mucosal cellular) adaptations happening after patients undergo RYGB. Our review compiles the current evidence and furthers the understanding of the rationale behind the food intake regulatory adaptations occurring after RYGB surgery. We identify gaps in the literature where the potential for future investigations and therapeutics may lie. We performed a comprehensive database search without language restrictions looking for RYGB bariatric surgery outcomes in patients with pre- and postoperative blood work hormonal profiling and/or gut mucosal biopsies. We gathered the relevant study results and describe them in this review. Where human findings were lacking, we included animal model studies. The amalgamation of physiologic, metabolic, and cellular adaptations following RYGB is yet to be fully characterized. This constitutes a fundamental aspiration for enhancing and individualizing obesity therapy.
Collapse
Affiliation(s)
- Fauzi Feris
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alison McRae
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Todd A Kellogg
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Travis McKenzie
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Omar Ghanem
- Division of Endocrine and Metabolic Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
59
|
Dobosz AM, Janikiewicz J, Krogulec E, Dziewulska A, Ajduk A, Szpila M, Nieznańska H, Szczepankiewicz AA, Wypych D, Dobrzyn A. Inhibition of stearoyl-CoA desaturase 1 in the mouse impairs pancreatic islet morphogenesis and promotes loss of β-cell identity and α-cell expansion in the mature pancreas. Mol Metab 2022; 67:101659. [PMID: 36529318 PMCID: PMC9801219 DOI: 10.1016/j.molmet.2022.101659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/02/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Abnormalities that characterize the pathophysiology of type 2 diabetes (T2D) include deficiencies of β-cells and the expansion of α-cells in pancreatic islets, manifested by lower insulin release and glucagon oversecretion. The molecular mechanisms that determine intra-islet interactions between pancreatic α- and β-cells are still not fully understood. The present study showed that stearoyl-coenzyme A (CoA) desaturase 1 (SCD1), an enzyme that is implicated in fatty acid metabolism, serves as a checkpoint in the control of endocrine cell equilibrium in pancreatic islets. Our data showed that SCD1 activity is essential for proper α-cell and β-cell lineage determination during morphogenesis of the pancreas and the maintenance of mature β-cell identity. The inhibition of SCD1 expression/activity led to both a decrease in the expression of β-cell signature genes (e.g., Pdx1, Nkx6.1, MafA, and Neurod1, among others) and induction of the expression of the dedifferentiation marker Sox9 in mature pancreatic islets. The transcriptional repression of Pdx1 and MafA in SCD1-deficient β-cells was related to the excessive methylation of promoter regions of these transcription factors. In contrast, SCD1 ablation favored the formation of α-cells over β-cells throughout pancreas organogenesis and did not compromise α-cell identity in adult pancreatic islets. Such molecular changes that were caused by SCD1 downregulation resulted in the mislocalization of α-cells within the core of islets and increased the ratio of pancreatic α- to β-cell mass. This was followed by islet dysfunction, including impairments in glucose-stimulated insulin release, simultaneously with elevations of basal glucagon secretion. Altogether, these findings provide additional mechanistic insights into the role of SCD1 in the pathogenesis of T2D.
Collapse
Affiliation(s)
- Aneta M. Dobosz
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland,Corresponding author.
| | - Justyna Janikiewicz
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| | - Ewelina Krogulec
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| | - Anna Dziewulska
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| | - Anna Ajduk
- Department of Embryology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Marcin Szpila
- Department of Embryology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Hanna Nieznańska
- Laboratory of Electron Microscopy, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| | - Andrzej A. Szczepankiewicz
- Laboratory of Electron Microscopy, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Dorota Wypych
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
60
|
Urva S, Coskun T, Loh MT, Du Y, Thomas MK, Gurbuz S, Haupt A, Benson CT, Hernandez-Illas M, D'Alessio DA, Milicevic Z. LY3437943, a novel triple GIP, GLP-1, and glucagon receptor agonist in people with type 2 diabetes: a phase 1b, multicentre, double-blind, placebo-controlled, randomised, multiple-ascending dose trial. Lancet 2022; 400:1869-1881. [PMID: 36354040 DOI: 10.1016/s0140-6736(22)02033-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Treating hyperglycaemia and obesity in individuals with type 2 diabetes using multi-receptor agonists can improve short-term and long-term outcomes. LY3437943 is a single peptide with agonist activity for glucagon, glucose-dependent insulinotropic polypeptide (GIP), and glucagon-like peptide 1 (GLP-1) receptors that is currently in development for the treatment of type 2 diabetes and for the treatment of obesity and associated comorbidities. We investigated the safety, pharmacokinetics, and pharmacodynamics of multiple weekly doses of LY3437943 in people with type 2 diabetes in a 12-week study. METHODS In this phase 1b, proof-of-concept, double-blind, placebo-controlled, randomised, multiple-ascending dose trial, adults (aged 20-70 years) with type 2 diabetes for at least 3 months, a glycated haemoglobin A1c (HbA1c) value of 7·0-10·5%, body-mass index of 23-50 kg/m2, and stable bodyweight (<5% change in previous 3 months) were recruited at four centres in the USA. Using an interactive web-response system, participants were randomly assigned to receive once-weekly subcutaneous injections of LY3437943, placebo, or dulaglutide 1·5 mg over a 12-week period. Five ascending dose cohorts were studied, with randomisation in each cohort such that a minimum of nine participants received LY3437943, three received placebo, and one received dulaglutide 1·5 mg within each cohort. The top doses in the two highest dose cohorts were attained via stepwise dose escalations. The primary outcome was to investigate the safety and tolerability of LY3437943, and characterising the pharmacodynamics and pharmacokinetics were secondary outcomes. Safety was analysed in all participants who received at least one dose of study drug, and pharmacodynamics and pharmacokinetics in all participants who received at least one dose of study drug and had evaluable data. This trial is registered at ClinicalTrials.gov, NCT04143802. FINDINGS Between Dec 18, 2019, and Dec 28, 2020, 210 people were screened, of whom 72 were enrolled, received at least one dose of study drug, and were included in safety analyses. 15 participants had placebo, five had dulaglutide 1·5 mg and, for LY3437943, nine had 0·5 mg, nine had 1·5 mg, 11 had 3 mg, 11 had 3/6 mg, and 12 had 3/6/9/12 mg. 29 participants discontinued the study prematurely. Treatment-emergent adverse events were reported by 33 (63%), three (60%), and eight (54%) participants who received LY3437943, dulaglutide 1·5 mg, and placebo, respectively, with gastrointestinal disorders being the most frequently reported treatment-emergent adverse events. The pharmacokinetics of LY3437943 were dose proportional and its half-life was approximately 6 days. At week 12, placebo-adjusted mean daily plasma glucose significantly decreased from baseline at the three highest dose LY3437943 groups (least-squares mean difference -2·8 mmol/L [90% CI -4·63 to -0·94] for 3 mg; -3·1 mmol/L [-4·91 to -1·22] for 3/6 mg; and -2·9 mmol/L [-4·70 to -1·01] for 3/6/9/12 mg). Placebo-adjusted sHbA1c also decreased significantly in the three highest dose groups (-1·4% [90% CI -2·17 to -0·56] for 3 mg; -1·6% [-2·37 to -0·75] for 3/6 mg; and -1·2% [-2·05 to -0·45] for 3/6/9/12 mg). Placebo-adjusted bodyweight reduction with LY3437943 appeared to be dose dependent (up to -8·96 kg [90% CI -11·16 to -6·75] in the 3/6/9/12 mg group). INTERPRETATION In this early phase study, LY3437943 showed an acceptable safety profile, and its pharmacokinetics suggest suitability for once-weekly dosing. This finding, together with the pharmacodynamic findings of robust reductions in glucose and bodyweight, provides support for phase 2 development. FUNDING Eli Lilly and Company.
Collapse
Affiliation(s)
- Shweta Urva
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Yu Du
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - Axel Haupt
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | - David A D'Alessio
- Duke Division of Endocrinology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
61
|
Bomholt AB, Johansen CD, Christensen JB, Kjeldsen SAS, Galsgaard KD, Winther-Sørensen M, Serizawa R, Hornum M, Porrini E, Pedersen J, Ørskov C, Gluud LL, Sørensen CM, Holst JJ, Albrechtsen R, Wewer Albrechtsen NJ. Evaluation of commercially available glucagon receptor antibodies and glucagon receptor expression. Commun Biol 2022; 5:1278. [PMID: 36418521 PMCID: PMC9684523 DOI: 10.1038/s42003-022-04242-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Glucagon is a major regulator of metabolism and drugs targeting the glucagon receptor (GCGR) are being developed. Insight into tissue and cell-specific expression of the GCGR is important to understand the biology of glucagon and to differentiate between direct and indirect actions of glucagon. However, it has been challenging to localize the GCGR in tissue due to low expression levels and lack of specific methods. Immunohistochemistry has frequently been used for GCGR localization, but antibodies targeting G-protein-coupled-receptors may be inaccurate. We evaluated all currently commercially available GCGR antibodies. The antibody, ab75240 (Antibody no. 11) was found to perform best among the twelve antibodies tested and using this antibody we found expression of the GCGR in the kidney, liver, preadipocytes, pancreas, and heart. Three antibody-independent approaches all confirmed the presence of the GCGR within the pancreas, liver and the kidneys. GCGR expression should be evaluated by both antibody and antibody-independent approaches.
Collapse
Affiliation(s)
- Anna Billeschou Bomholt
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Dall Johansen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bager Christensen
- grid.5254.60000 0001 0674 042XDepartment of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sasha Alexandra Sampson Kjeldsen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Winther-Sørensen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Reza Serizawa
- grid.4973.90000 0004 0646 7373Department of Pathology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Mads Hornum
- grid.475435.4Department of Nephrology, Centre for Cancer and Organ Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Esteban Porrini
- grid.411220.40000 0000 9826 9219Instituto de Tecnologías Biomédicas, University of La Laguna, Research Unit, Hospital Universitario de Canarias, Tenerife, Spain
| | - Jens Pedersen
- grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.411900.d0000 0004 0646 8325Department of Internal Medicine, Endocrinology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Cathrine Ørskov
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Lotte Gluud
- grid.5254.60000 0001 0674 042XDepartment of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.4973.90000 0004 0646 7373Gastro Unit, Copenhagen University Hospital, Hvidovre, Denmark
| | - Charlotte Mehlin Sørensen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Reidar Albrechtsen
- grid.5254.60000 0001 0674 042XBiotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai Jacob Wewer Albrechtsen
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.512917.9Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| |
Collapse
|
62
|
Hope DCD, Hinds CE, Lopes T, Vincent ML, Shrewsbury JV, Yu ATC, Davies I, Scott R, Jones B, Murphy KG, Minnion JS, Sardini A, Carling D, Lutz TA, Bloom SR, Tan TMM, Owen BM. Hypoaminoacidemia underpins glucagon-mediated energy expenditure and weight loss. Cell Rep Med 2022; 3:100810. [PMID: 36384093 PMCID: PMC9729826 DOI: 10.1016/j.xcrm.2022.100810] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 09/26/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022]
Abstract
Glucagon analogs show promise as components of next-generation, multi-target, anti-obesity therapeutics. The biology of chronic glucagon treatment, in particular, its ability to induce energy expenditure and weight loss, remains poorly understood. Using a long-acting glucagon analog, G108, we demonstrate that glucagon-mediated body weight loss is intrinsically linked to the hypoaminoacidemia associated with its known amino acid catabolic action. Mechanistic studies reveal an energy-consuming response to low plasma amino acids in G108-treated mice, prevented by dietary amino acid supplementation and mimicked by a rationally designed low amino acid diet. Therefore, low plasma amino acids are a pre-requisite for G108-mediated energy expenditure and weight loss. However, preventing hypoaminoacidemia with additional dietary protein does not affect the ability of G108 to improve glycemia or hepatic steatosis in obese mice. These studies provide a mechanism for glucagon-mediated weight loss and confirm the hepatic glucagon receptor as an attractive molecular target for metabolic disease therapeutics.
Collapse
Affiliation(s)
- David C D Hope
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Charlotte E Hinds
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tatiana Lopes
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Matthew L Vincent
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jed V Shrewsbury
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Arthur T C Yu
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Iona Davies
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Rebecca Scott
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ben Jones
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Kevin G Murphy
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - James S Minnion
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alessandro Sardini
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - David Carling
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Stephen R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia M M Tan
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Bryn M Owen
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
63
|
Lednovich KR, Gough S, Brenner M, Qadri T, Layden BT. G
Protein‐Coupled Receptors in Metabolic Disease. GPCRS AS THERAPEUTIC TARGETS 2022:521-552. [DOI: 10.1002/9781119564782.ch15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
64
|
Heebøll S, Risikesan J, Ringgaard S, Kumarathas I, Sandahl TD, Grønbæk H, Søndergaard E, Nielsen S. Impaired Glucagon-Mediated Suppression of VLDL-Triglyceride Secretion in Individuals With Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD). Diabetes 2022; 71:2402-2411. [PMID: 36001750 PMCID: PMC9630084 DOI: 10.2337/db22-0313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/22/2022] [Indexed: 01/25/2023]
Abstract
Individuals with metabolic dysfunction-associated fatty liver disease (MAFLD) have elevated plasma lipids as well as glucagon, although glucagon suppresses hepatic VLDL-triglyceride (TG) secretion. We hypothesize that the sensitivity to glucagon in hepatic lipid metabolism is impaired in MAFLD. We recruited 11 subjects with severe MAFLD (MAFLD+), 10 with mild MAFLD (MAFLD-), and 7 overweight control (CON) subjects. We performed a pancreatic clamp with a somatostatin analog (octreotide) to suppress endogenous hormone production, combined with infusion of low-dose glucagon (0.65 ng/kg/min, t = 0-270 min, LowGlucagon), followed by high-dose glucagon (1.5 ng/kg/min, t = 270-450 min, HighGlucagon). VLDL-TG and glucose tracers were used to evaluate VLDL-TG kinetics and endogenous glucose production (EGP). HighGlucagon suppressed VLDL-TG secretion compared with LowGlucagon. This suppression was markedly attenuated in MAFLD subjects compared with CON subjects (MAFLD+: 13% ± [SEM] 5%; MAFLD-: 10% ± 3%; CON: 36% ± 7%, P < 0.01), with no difference between MAFLD groups. VLDL-TG concentration and VLDL-TG oxidation rate increased between LowGlucagon and HighGlucagon in MAFLD+ subjects compared with CON subjects. EGP transiently increased during HighGlucagon without any difference between the three groups. Individuals with MAFLD have a reduced sensitivity to glucagon in the hepatic TG metabolism, which could contribute to the dyslipidemia seen in MAFLD patients. ClinicalTrials.gov: NCT04042142.
Collapse
Affiliation(s)
- Sara Heebøll
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Corresponding author: Sara Heebøll,
| | - Jeyanthini Risikesan
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Internal Medicine, Regional Hospital Viborg, Viborg, Denmark
| | - Steffen Ringgaard
- Magnetic Resonance Research Centre, Aarhus University, Aarhus, Denmark
| | - Indumathi Kumarathas
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thomas D. Sandahl
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Henning Grønbæk
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Esben Søndergaard
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
65
|
Vps37a regulates hepatic glucose production by controlling glucagon receptor localization to endosomes. Cell Metab 2022; 34:1824-1842.e9. [PMID: 36243006 DOI: 10.1016/j.cmet.2022.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/04/2022] [Accepted: 09/21/2022] [Indexed: 01/11/2023]
Abstract
During mammalian energy homeostasis, the glucagon receptor (Gcgr) plays a key role in regulating both glucose and lipid metabolisms. However, the mechanisms by which these distinct signaling arms are differentially regulated remain poorly understood. Using a Cy5-glucagon agonist, we show that the endosomal protein Vps37a uncouples glucose production from lipid usage downstream of Gcgr signaling by altering intracellular receptor localization. Hepatocyte-specific knockdown of Vps37a causes an accumulation of Gcgr in endosomes, resulting in overactivation of the cAMP/PKA/p-Creb signaling pathway to gluconeogenesis without affecting β-oxidation. Shifting the receptor back to the plasma membrane rescues the differential signaling and highlights the importance of the spatiotemporal localization of Gcgr for its metabolic effects. Importantly, since Vps37a knockdown in animals fed with a high-fat diet leads to hyperglycemia, although its overexpression reduces blood glucose levels, these data reveal a contribution of endosomal signaling to metabolic diseases that could be exploited for treatments of type 2 diabetes.
Collapse
|
66
|
Vasileva A, Marx T, Beaudry JL, Stern JH. Glucagon receptor signaling at white adipose tissue does not regulate lipolysis. Am J Physiol Endocrinol Metab 2022; 323:E389-E401. [PMID: 36002172 PMCID: PMC9576180 DOI: 10.1152/ajpendo.00078.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Although the physiological role of glucagon receptor signaling in the liver is well defined, the impact of glucagon receptor (Gcgr) signaling on white adipose tissue (WAT) continues to be debated. Although numerous studies propose that glucagon stimulates WAT lipolysis, we lack evidence that physiological concentrations of glucagon regulate WAT lipolysis. In turn, we performed studies in both wild-type and WAT Gcgr knockout mice to determine if glucagon regulates lipolysis at WAT in the mouse. We assessed the effects of fasting and acute exogenous glucagon administration in wild-type C57BL/6J and GcgrAdipocyte+/+ versus GcgrAdipocyte-/- mice. Using an ex vivo lipolysis protocol, we further examined the direct effects of glucagon on physiologically (fasted) and pharmacologically stimulated lipolysis. We found that adipocyte Gcgr expression did not affect fasting-induced lipolysis or hepatic lipid accumulation in lean or diet-induced obese (DIO) mice. Acute glucagon administration did not affect serum nonesterified fatty acids (NEFA), leptin, or adiponectin concentration, but did increase serum glucose and FGF21, regardless of genotype. Glucagon did not affect ex vivo lipolysis in explants from either GcgrAdipocyte+/+ or GcgrAdipocyte-/- mice. Gcgr expression did not affect fasting-induced or isoproterenol-stimulated lipolysis from WAT explants. Moreover, glucagon receptor signaling at WAT did not affect body weight or glucose homeostasis in lean or DIO mice. Our studies have established that physiological levels of glucagon do not regulate WAT lipolysis, either directly or indirectly. Given that glucagon receptor agonism can improve dyslipidemia and decrease hepatic lipid accumulation, it is critical to understand the tissue-specific effects of glucagon receptor action. Unlike the crucial role of hepatic glucagon receptor signaling in maintaining glucose and lipid homeostasis, we observed no metabolic consequence of WAT glucagon receptor deletion.NEW & NOTEWORTHY It has been postulated that glucagon stimulates lipolysis and fatty acid release from white adipose tissue. We observed no metabolic effects of eliminating or activating glucagon receptor signaling at white adipose tissue.
Collapse
Affiliation(s)
- Anastasiia Vasileva
- Division of Endocrinology, University of Arizona College of Medicine, Tucson, Arizona
| | - Tyler Marx
- Division of Endocrinology, University of Arizona College of Medicine, Tucson, Arizona
| | - Jacqueline L Beaudry
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer H Stern
- Division of Endocrinology, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
67
|
Pharmacokinetics and Endocrine Effects of an Oral Dose of D-Pinitol in Human Fasting Healthy Volunteers. Nutrients 2022; 14:nu14194094. [PMID: 36235746 PMCID: PMC9572189 DOI: 10.3390/nu14194094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
The present study characterizes the oral pharmacokinetics of D-Pinitol, a natural insulin mimetic inositol, in human healthy volunteers (14 males and 11 females). D-Pinitol absorption was studied in (a) subjects receiving a single oral dose of 15 mg/kg (n = 10), or (b) 5 mg/kg pure D-Pinitol (n = 6), and (c) subjects receiving D-Pinitol as part of carbohydrate-containing carob pods-derived syrup with a 3.2% D-Pinitol (Dose of 1600 mg/subject, n = 9). The volunteers received a randomly assigned single dose of either D-Pinitol or carob pod-derived syrup. Blood samples were collected at 0, 15, 30, 45, 60, 90, 120, 180, 240, 360 and 1440 min after intake. Plasma concentration of D-Pinitol was measured and pharmacokinetic parameters obtained. The data indicate that when given alone, the oral absorption of D-Pinitol is dose-dependent and of extended duration, with a Tmax reached after almost 4 h, and a half-life greater than 5 h. When the source of D-Pinitol was a carob pods-derived syrup, Cmax was reduced to 40% of the expected based on the data of D-Pinitol alone, suggesting a reduced absorption probably because of competition with monosaccharide transport. In this group, Tmax was reached before that of D-Pinitol alone, but the estimated half-life remained the same. In the D-Pinitol groups, plasma concentrations of glucose, insulin, glucagon, ghrelin, free fatty acids, and pituitary hormones were additionally measured. A dose of 15 mg/kg of D-Pinitol did not affect glucose levels in healthy volunteers, but reduced insulin and increased glucagon and ghrelin concentrations. D-Pinitol did not increase other hormones known to enhance plasma glucose, such as cortisol or GH, which were surprisingly reduced after the ingestion of this inositol. Other pituitary hormones (gonadotropins, prolactin, and thyroid-stimulating hormone) were not affected after D-Pinitol ingestion. In a conclusion, D-Pinitol is absorbed through the oral route, having an extended half-life and displaying the pharmacological profile of an endocrine pancreas protector, a pharmacological activity of potential interest for the treatment or prevention of insulin resistance-associated conditions.
Collapse
|
68
|
Richter MM, Galsgaard KD, Elmelund E, Knop FK, Suppli MP, Holst JJ, Winther-Sørensen M, Kjeldsen SA, Wewer Albrechtsen NJ. The Liver-α-Cell Axis in Health and in Disease. Diabetes 2022; 71:1852-1861. [PMID: 35657688 PMCID: PMC9862287 DOI: 10.2337/dbi22-0004] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023]
Abstract
Glucagon and insulin are the main regulators of blood glucose. While the actions of insulin are extensively mapped, less is known about glucagon. Besides glucagon's role in glucose homeostasis, there are additional links between the pancreatic α-cells and the hepatocytes, often collectively referred to as the liver-α-cell axis, that may be of importance for health and disease. Thus, glucagon receptor antagonism (pharmacological or genetic), which disrupts the liver-α-cell axis, results not only in lower fasting glucose but also in reduced amino acid turnover and dyslipidemia. Here, we review the actions of glucagon on glucose homeostasis, amino acid catabolism, and lipid metabolism in the context of the liver-α-cell axis. The concept of glucagon resistance is also discussed, and we argue that the various elements of the liver-α-cell axis may be differentially affected in metabolic diseases such as diabetes, obesity, and nonalcoholic fatty liver disease (NAFLD). This conceptual rethinking of glucagon biology may explain why patients with type 2 diabetes have hyperglucagonemia and how NAFLD disrupts the liver-α-cell axis, compromising the normal glucagon-mediated enhancement of substrate-induced amino acid turnover and possibly fatty acid β-oxidation. In contrast to amino acid catabolism, glucagon-induced glucose production may not be affected by NAFLD, explaining the diabetogenic effect of NAFLD-associated hyperglucagonemia. Consideration of the liver-α-cell axis is essential to understanding the complex pathophysiology underlying diabetes and other metabolic diseases.
Collapse
Affiliation(s)
- Michael M. Richter
- Department of Clinical Biochemistry, Diagnostic Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine D. Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Elmelund
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K. Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Malte P. Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Jens J. Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Winther-Sørensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sasha A.S. Kjeldsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J. Wewer Albrechtsen
- Department of Clinical Biochemistry, Diagnostic Center, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital—Bispebjerg and Frederiksberg Hospital, Bispebjerg, Denmark
- Corresponding author: Nicolai J. Wewer Albrechtsen,
| |
Collapse
|
69
|
Signaling pathways in obesity: mechanisms and therapeutic interventions. Signal Transduct Target Ther 2022; 7:298. [PMID: 36031641 PMCID: PMC9420733 DOI: 10.1038/s41392-022-01149-x] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 12/19/2022] Open
Abstract
Obesity is a complex, chronic disease and global public health challenge. Characterized by excessive fat accumulation in the body, obesity sharply increases the risk of several diseases, such as type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease, and is linked to lower life expectancy. Although lifestyle intervention (diet and exercise) has remarkable effects on weight management, achieving long-term success at weight loss is extremely challenging, and the prevalence of obesity continues to rise worldwide. Over the past decades, the pathophysiology of obesity has been extensively investigated, and an increasing number of signal transduction pathways have been implicated in obesity, making it possible to fight obesity in a more effective and precise way. In this review, we summarize recent advances in the pathogenesis of obesity from both experimental and clinical studies, focusing on signaling pathways and their roles in the regulation of food intake, glucose homeostasis, adipogenesis, thermogenesis, and chronic inflammation. We also discuss the current anti-obesity drugs, as well as weight loss compounds in clinical trials, that target these signals. The evolving knowledge of signaling transduction may shed light on the future direction of obesity research, as we move into a new era of precision medicine.
Collapse
|
70
|
Arad E, Yosefi G, Kolusheva S, Bitton R, Rapaport H, Jelinek R. Native Glucagon Amyloids Catalyze Key Metabolic Reactions. ACS NANO 2022; 16:12889-12899. [PMID: 35866668 DOI: 10.1021/acsnano.2c05166] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Glucagon is a prominent peptide hormone, playing central roles in the regulation of glucose blood-level and lipid metabolism. Formation of glucagon amyloid fibrils has been previously reported, although no biological functions of such fibrils are known. Here, we demonstrate that glucagon amyloid fibrils catalyze biologically important reactions, including esterolysis, lipid hydrolysis, and dephosphorylation. In particular, we found that glucagon fibrils catalyze dephosphorylation of adenosine triphosphate (ATP), a core metabolic reaction in cell biology. Comparative analysis of several glucagon variants allowed mapping the catalytic activity to an enzymatic pocket-like triad formed at the glucagon fibril surface, comprising the histidyl-serine domain at the N-terminus of the peptide. This study may point to previously unknown physiological roles and pathological consequences of glucagon fibrillation and supports the hypothesis that catalytic activities of native amyloid fibrils play functional roles in human physiology and disease.
Collapse
Affiliation(s)
- Elad Arad
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Gal Yosefi
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sofiya Kolusheva
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Ronit Bitton
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Hanna Rapaport
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Raz Jelinek
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
71
|
Navarro JA, Decara J, Medina-Vera D, Tovar R, Lopez-Gambero AJ, Suarez J, Pavón FJ, Serrano A, de Ceglia M, Sanjuan C, Baltasar YA, Baixeras E, Rodríguez de Fonseca F. Endocrine and Metabolic Impact of Oral Ingestion of a Carob-Pod-Derived Natural-Syrup-Containing D-Pinitol: Potential Use as a Novel Sweetener in Diabetes. Pharmaceutics 2022; 14:pharmaceutics14081594. [PMID: 36015220 PMCID: PMC9416495 DOI: 10.3390/pharmaceutics14081594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 02/05/2023] Open
Abstract
The widespread use of added sugars or non-nutritive sweeteners in processed foods is a challenge for addressing the therapeutics of obesity and diabetes. Both types of sweeteners generate health problems, and both are being blamed for multiple complications associated with these prevalent diseases. As an example, fructose is proven to contribute to obesity and liver steatosis, while non-nutritive sweeteners generate gut dysbiosis that complicates the metabolic control exerted by the liver. The present work explores an alternative approach for sweetening through the use of a simple carob-pod-derived syrup. This sweetener consists of a balanced mixture of fructose (47%) and glucose (45%), as sweetening sugars, and a functional natural ingredient (D-Pinitol) at a concentration (3%) capable of producing active metabolic effects. The administration of this syrup to healthy volunteers (50 g of total carbohydrates) resulted in less persistent glucose excursions, a lower insulin response to the hyperglycemia produced by its ingestion, and an enhanced glucagon/insulin ratio, compared to that observed after the ingestion of 50 g of glucose. Daily administration of the syrup to Wistar rats for 10 days lowered fat depots in the liver, reduced liver glycogen, promoted fat oxidation, and was devoid of toxic effects. In addition, this repeated administration of the syrup improved glucose handling after a glucose (2 g/kg) load. Overall, this alternative functional sweetener retains the natural palatability of a glucose/fructose syrup while displaying beneficial metabolic effects that might serve to protect against the progression towards complicated obesity, especially the development of liver steatosis.
Collapse
Affiliation(s)
- Juan A. Navarro
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
- Facultad de Medicina, Campus de Teatinos s/n, Universidad de Málaga, 29010 Málaga, Spain
| | - Juan Decara
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
| | - Dina Medina-Vera
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
- Facultad de Medicina, Campus de Teatinos s/n, Universidad de Málaga, 29010 Málaga, Spain
- Unidad de Gestión del Corazón, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Facultad de Ciencias, Campus de Teatinos s/n, Universidad de Málaga, 29010 Málaga, Spain
| | - Ruben Tovar
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
- Facultad de Medicina, Campus de Teatinos s/n, Universidad de Málaga, 29010 Málaga, Spain
| | - Antonio J. Lopez-Gambero
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
- Facultad de Ciencias, Campus de Teatinos s/n, Universidad de Málaga, 29010 Málaga, Spain
| | - Juan Suarez
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, 29010 Málaga, Spain
| | - Francisco Javier Pavón
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
- Unidad de Gestión del Corazón, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Antonia Serrano
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
| | - Marialuisa de Ceglia
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
| | - Carlos Sanjuan
- Euronutra S.L. Calle Johannes Kepler, 3, 29590 Málaga, Spain; (C.S.); (Y.A.B.)
| | | | - Elena Baixeras
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
- Correspondence: (E.B.); (F.R.d.F.); Tel.: +34-655373093 (E.B.); +34-669426548 (F.R.d.F.)
| | - Fernando Rodríguez de Fonseca
- Laboratorio de Medicina Regenerativa, Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.A.N.); (J.D.); (D.M.-V.); (R.T.); (A.J.L.-G.); (J.S.); (F.J.P.); (A.S.); (M.d.C.)
- Correspondence: (E.B.); (F.R.d.F.); Tel.: +34-655373093 (E.B.); +34-669426548 (F.R.d.F.)
| |
Collapse
|
72
|
Moon JS, Hong JH, Jung YJ, Ferrannini E, Nauck MA, Lim S. SGLT-2 inhibitors and GLP-1 receptor agonists in metabolic dysfunction-associated fatty liver disease. Trends Endocrinol Metab 2022; 33:424-442. [PMID: 35491295 DOI: 10.1016/j.tem.2022.03.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/22/2022] [Accepted: 03/11/2022] [Indexed: 02/07/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a chronic condition that affects nearly one billion people globally, characterized by triacylglycerol accumulation in the liver as a consequence of metabolic abnormalities (obesity and impaired glucose regulation). Low-grade inflammation, oxidative stress, mitochondrial dysfunction, and dysbiosis in gut microbiota are involved in the etiology of MAFLD, and both cardiovascular events and hepatic complications are the long-term consequences. In the absence of approved therapies for this condition, sodium-glucose cotransporter 2 inhibitors (SGLT-2 Is) and glucagon-like peptide 1 receptor agonists (GLP-1 RAs) have the specific advantage of lowering body weight and providing cardiovascular benefits. Here, we discuss potential roles for SGLT-2 Is and GLP-1 RAs in the prevention and treatment of intrahepatic triacylglycerol accumulation and associated inflammation and/or fibrosis.
Collapse
Affiliation(s)
- Jun Sung Moon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Jun Hwa Hong
- Department of Internal Medicine, Eulji University Hospital, School of Medicine, Daejeon, Republic of Korea
| | - Yong Jin Jung
- Department of Internal Medicine, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | - Michael A Nauck
- Diabetes Division, Katholisches Klinikum Bochum, St Josef Hospital (Ruhr-University, Bochum), Bochum, Germany.
| | - Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| |
Collapse
|
73
|
Stinson SE, Jonsson AE, de Retana Alzola IF, Lund MAV, Frithioff-Bøjsøe C, Aas Holm L, Fonvig CE, Pedersen O, Ängquist L, Sørensen TIA, Holst JJ, Christiansen M, Holm JC, Hartmann B, Hansen T. Hyperglucagonemia in Pediatric Adiposity Associates With Cardiometabolic Risk Factors but Not Hyperglycemia. J Clin Endocrinol Metab 2022; 107:1569-1576. [PMID: 35213713 PMCID: PMC9113783 DOI: 10.1210/clinem/dgac108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 01/18/2023]
Abstract
CONTEXT In adults, hyperglucagonemia is associated with type 2 diabetes, impaired glucose tolerance, and obesity. The role of glucagon in pediatric overweight/obesity remains unclear. OBJECTIVE We examined whether fasting concentrations of glucagon are elevated in youth with overweight/obesity and whether this associates with cardiometabolic risk profiles. METHODS Analyses were based on the cross-sectional HOLBAEK study, including children and adolescents 6 to 19 years of age, with overweight/obesity from an obesity clinic group (n = 2154) and with normal weight from a population-based group (n = 1858). Fasting concentrations of plasma glucagon and cardiometabolic risk outcomes were assessed, and multiple linear and logistic regressions models were performed. RESULTS The obesity clinic group had higher glucagon concentrations than the population-based group (P < 0.001). Glucagon positively associated with body mass index (BMI) standard deviation score (SDS), waist, body fat %, liver fat %, alanine transaminase (ALT), high-sensitivity C-reactive protein, homeostasis model assessment of insulin resistance, insulin, C-peptide, LDL-C, triglycerides, SDS of diastolic and systolic blood pressure, and was inversely associated with fasting glucose. The inverse relationship between glucagon and glucose was attenuated in individuals with high BMI SDS and high fasting insulin. Glucagon was associated with a higher prevalence of insulin resistance, increased ALT, dyslipidemia, and hypertension, but not with hyperglycemia. Glucagon was positively associated with fasting total glucagon-like peptide-1. CONCLUSION Compared with normal weight peers, children and adolescents with overweight/obesity had elevated concentrations of fasting glucagon, which corresponded to worsened cardiometabolic risk outcomes, except for hyperglycemia. This suggests hyperglucagonemia in youth may precede impairments in glucose regulation.
Collapse
Affiliation(s)
- Sara E Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna E Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ierai Fernández de Retana Alzola
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten A V Lund
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Children’s Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, Holbæk, Denmark
| | - Christine Frithioff-Bøjsøe
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Children’s Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, Holbæk, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Children’s Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, Holbæk, Denmark
| | - Cilius E Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Children’s Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, Holbæk, Denmark
- Department of Pediatrics, Kolding Hospital a part of Lillebælt Hospital, Kolding, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Christiansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Children’s Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Holbæk Hospital, Holbæk, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
74
|
Rodgers RL. Glucagon, cyclic AMP, and hepatic glucose mobilization: A half‐century of uncertainty. Physiol Rep 2022; 10:e15263. [PMID: 35569125 PMCID: PMC9107925 DOI: 10.14814/phy2.15263] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022] Open
Abstract
For at least 50 years, the prevailing view has been that the adenylate cyclase (AC)/cyclic AMP (cAMP)/protein kinase A pathway is the predominant signal mediating the hepatic glucose‐mobilizing actions of glucagon. A wealth of evidence, however, supports the alternative, that the operative signal most of the time is the phospholipase C (PLC)/inositol‐phosphate (IP3)/calcium/calmodulin pathway. The evidence can be summarized as follows: (1) The consensus threshold glucagon concentration for activating AC ex vivo is 100 pM, but the statistical hepatic portal plasma glucagon concentration range, measured by RIA, is between 28 and 60 pM; (2) Within that physiological concentration range, glucagon stimulates the PLC/IP3 pathway and robustly increases glucose output without affecting the AC/cAMP pathway; (3) Activation of a latent, amplified AC/cAMP pathway at concentrations below 60 pM is very unlikely; and (4) Activation of the PLC/IP3 pathway at physiological concentrations produces intracellular effects that are similar to those produced by activation of the AC/cAMP pathway at concentrations above 100 pM, including elevated intracellular calcium and altered activities and expressions of key enzymes involved in glycogenolysis, gluconeogenesis, and glycogen synthesis. Under metabolically stressful conditions, as in the early neonate or exercising adult, plasma glucagon concentrations often exceed 100 pM, recruiting the AC/cAMP pathway and enhancing the activation of PLC/IP3 pathway to boost glucose output, adaptively meeting the elevated systemic glucose demand. Whether the AC/cAMP pathway is consistently activated in starvation or diabetes is not clear. Because the importance of glucagon in the pathogenesis of diabetes is becoming increasingly evident, it is even more urgent now to resolve lingering uncertainties and definitively establish glucagon’s true mechanism of glycemia regulation in health and disease.
Collapse
Affiliation(s)
- Robert L. Rodgers
- Department of Biomedical and Pharmaceutical Sciences College of Pharmacy University of Rhode Island Kingston Rhode Island USA
| |
Collapse
|
75
|
De Block CEM, Dirinck E, Verhaegen A, Van Gaal LF. Efficacy and safety of high-dose glucagon-like peptide-1, glucagon-like peptide-1/glucose-dependent insulinotropic peptide, and glucagon-like peptide-1/glucagon receptor agonists in type 2 diabetes. Diabetes Obes Metab 2022; 24:788-805. [PMID: 34984793 DOI: 10.1111/dom.14640] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/16/2021] [Accepted: 01/01/2022] [Indexed: 12/11/2022]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) have become agents of choice for people with type 2 diabetes (T2D) with established cardiovascular disease or in high-risk individuals. With currently available GLP-1 RAs, 51%-79% of subjects achieve an HbA1c target of less than 7.0% and 4%-27% lose 10% of body weight, illustrating the need for more potent agents. Three databases (PubMed, Cochrane, Web of Science) were searched using the MESH terms 'glucagon-like peptide-1 receptor agonist', 'glucagon receptor agonist', 'glucose-dependent insulinotropic peptide', 'dual or co-agonist', and 'tirzepatide'. Quality of papers was scored using PRISMA guidelines. Risk of bias was evaluated using the Cochrane assessment tool. An HbA1c target of less than 7.0% was attained by up to 80% with high-dose GLP-1 RAs and up to 97% with tirzepatide, with even up to 62% of people with T2D reaching an HbA1c of less than 5.7%. A body weight loss of 10% or greater was obtained by up to 50% and up to 69% with high-dose GLP-1 RAs or tirzepatide, respectively. The glucose- and weight-lowering effects of the GLP-1/glucagon RA cotadutide equal those of liraglutide 1.8 mg. Gastrointestinal side effects of high-dose GLP-1 RAs and co-agonists occurred in 30%-70% of patients, mostly arising within the first 2 weeks of the first dose, being mild or moderate in severity, and transient. The development of high-dose GLP-1 RAs and the dual GLP-1/glucose-dependent insulinotropic peptide RA tirzepatide resulted in increasing numbers of people reaching HbA1c and body weight targets, with up to 62% attaining normoglycaemia with 15-mg tirzepatide. Whether this will also translate to better cardiovascular outcomes and affect treatment guidelines remains to be studied.
Collapse
Affiliation(s)
- Christophe E M De Block
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), University of Antwerp, Wilrijk, Belgium
| | - Eveline Dirinck
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), University of Antwerp, Wilrijk, Belgium
| | - Ann Verhaegen
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), University of Antwerp, Wilrijk, Belgium
| | - Luc F Van Gaal
- Department of Endocrinology, Diabetology & Metabolism, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
- Faculty of Medicine & Health Sciences, Laboratory of Experimental Medicine and Paediatrics (LEMP), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
76
|
Khan D, Moffett RC, Flatt PR, Tarasov AI. Classical and non-classical islet peptides in the control of β-cell function. Peptides 2022; 150:170715. [PMID: 34958851 DOI: 10.1016/j.peptides.2021.170715] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/25/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022]
Abstract
The dual role of the pancreas as both an endocrine and exocrine gland is vital for food digestion and control of nutrient metabolism. The exocrine pancreas secretes enzymes into the small intestine aiding digestion of sugars and fats, whereas the endocrine pancreas secretes a cocktail of hormones into the blood, which is responsible for blood glucose control and regulation of carbohydrate, protein and fat metabolism. Classical islet hormones, insulin, glucagon, pancreatic polypeptide and somatostatin, interact in an autocrine and paracrine manner, to fine-tube the islet function and insulin secretion to the needs of the body. Recently pancreatic islets have been reported to express a number of non-classical peptide hormones involved in metabolic signalling, whose major production site was believed to reside outside pancreas, e.g. in the small intestine. We highlight the key non-classical islet peptides, and consider their involvement, together with established islet hormones, in regulation of stimulus-secretion coupling as well as proliferation, survival and transdifferentiation of β-cells. We furthermore focus on the paracrine interaction between classical and non-classical islet hormones in the maintenance of β-cell function. Understanding the functional relationships between these islet peptides might help to develop novel, more efficient treatments for diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| | - R Charlotte Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Andrei I Tarasov
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
77
|
Kettunen JLT, Rantala E, Dwivedi OP, Isomaa B, Sarelin L, Kokko P, Hakaste L, Miettinen PJ, Groop LC, Tuomi T. A multigenerational study on phenotypic consequences of the most common causal variant of HNF1A-MODY. Diabetologia 2022; 65:632-643. [PMID: 34951657 PMCID: PMC8894160 DOI: 10.1007/s00125-021-05631-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Systematic studies on the phenotypic consequences of variants causal of HNF1A-MODY are rare. Our aim was to assess the phenotype of carriers of a single HNF1A variant and genetic and clinical factors affecting the clinical spectrum. METHODS We conducted a family-based multigenerational study by comparing heterozygous carriers of the HNF1A p.(Gly292fs) variant with the non-carrier relatives irrespective of diabetes status. During more than two decades, 145 carriers and 131 non-carriers from 12 families participated in the study, and 208 underwent an OGTT at least once. We assessed the polygenic risk score for type 2 diabetes, age at onset of diabetes and measures of body composition, as well as plasma glucose, serum insulin, proinsulin, C-peptide, glucagon and NEFA response during the OGTT. RESULTS Half of the carriers remained free of diabetes at 23 years, one-third at 33 years and 13% even at 50 years. The median age at diagnosis was 21 years (IQR 17-35). We could not identify clinical factors affecting the age at conversion; sex, BMI, insulin sensitivity or parental carrier status had no significant effect. However, for 1 SD unit increase of a polygenic risk score for type 2 diabetes, the predicted age at diagnosis decreased by 3.2 years. During the OGTT, the carriers had higher levels of plasma glucose and lower levels of serum insulin and C-peptide than the non-carriers. The carriers were also leaner than the non-carriers (by 5.0 kg, p=0.012, and by 2.1 kg/m2 units of BMI, p=2.2 × 10-4, using the first adult measurements) and, possibly as a result of insulin deficiency, demonstrated higher lipolytic activity (with medians of NEFA at fasting 621 vs 441 μmol/l, p=0.0039; at 120 min during an OGTT 117 vs 64 μmol/l, p=3.1 × 10-5). CONCLUSIONS/INTERPRETATION The most common causal variant of HNF1A-MODY, p.(Gly292fs), presents not only with hyperglycaemia and insulin deficiency, but also with increased lipolysis and markedly lower adult BMI. Serum insulin was more discriminative than C-peptide between carriers and non-carriers. A considerable proportion of carriers develop diabetes after young adulthood. Even among individuals with a monogenic form of diabetes, polygenic risk of diabetes modifies the age at onset of diabetes.
Collapse
Affiliation(s)
- Jarno L T Kettunen
- Folkhälsan Research Center, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | | | - Om P Dwivedi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Bo Isomaa
- Folkhälsan Research Center, Helsinki, Finland
| | | | - Paula Kokko
- Folkhälsan Research Center, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Liisa Hakaste
- Folkhälsan Research Center, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Päivi J Miettinen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Molecular Neurology, and Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
| | - Leif C Groop
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Tiinamaija Tuomi
- Folkhälsan Research Center, Helsinki, Finland.
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.
- Department of Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland.
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland.
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
| |
Collapse
|
78
|
Huh KY, Hwang JG, Shin W, Baek S, Choi J, Lee N, Cho YM, Lee H. A double-blind, placebo-controlled, single-ascending dose study to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of HM15136, a novel long-acting glucagon analogue, in healthy subjects. Diabetes Obes Metab 2022; 24:411-420. [PMID: 34726329 DOI: 10.1111/dom.14590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 12/12/2022]
Abstract
AIM To evaluate the safety and tolerability, pharmacokinetics, and pharmacodynamics of HM15136, a novel long-acting glucagon analogue under development, in healthy males and females presenting with no childbearing potential. MATERIALS AND METHODS A randomized, double-blind, placebo-controlled, single-ascending dose study was conducted in 56 subjects who randomly received a single subcutaneous dose of HM15136 or its matching placebo at a ratio of 6:2 at 10, 20, 30, 50, 80, 100, and 120 μg/kg. RESULTS All adverse events were mild and transient. Neither serious adverse events nor discontinuation as a result of adverse events occurred. The most frequent adverse drug reaction was nausea (5.3%, only in the 100- and 120-μg/kg groups). HM15136, particularly at doses of 50 μg/kg or higher, increased fasting blood glucose, with a maximum increase and area under the curve of 1.5 mmol/L at day 10 (P = .006) and 166.3 day·mmol/L (P = .022) at the dose of 80 μg/kg, while suppressing the secretion of endogenous glucagon, which continued until day 17. HM15136 also significantly reduced gluconeogenic and ketogenic amino acids. Compensatory changes in endogenous insulin and incretin hormones by HM15136 were not apparent. HM15136 was slowly but steadily absorbed and reached a peak concentration at 46-68 hours after a single subcutaneous injection. HM15136 was eliminated with a terminal phase half-life of 77.1-101.1 hours. CONCLUSIONS A single subcutaneous dose of HM15136 at 10-120 μg/kg was safe and well tolerated. The long half-life of HM15136, coupled with an increase in blood glucose for ~2 weeks, may warrant a weekly dosing regimen.
Collapse
Affiliation(s)
- Ki Young Huh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Jun Gi Hwang
- Department of Clinical Pharmacology and Therapeutics, Chungbuk National University Hospital, Cheongju-si, South Korea
| | - Wonjung Shin
- Hanmi Pharmaceutical Co., Ltd, Seoul, South Korea
| | | | - JaeDuk Choi
- Hanmi Pharmaceutical Co., Ltd, Seoul, South Korea
| | - Nora Lee
- Hanmi Pharmaceutical Co., Ltd, Seoul, South Korea
| | - Young Min Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Howard Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
- Center for Convergence Approaches in Drug Development, Seoul, South Korea
- Advanced Institutes of Convergence Technology, Suwon, South Korea
| |
Collapse
|
79
|
Wu H, Wang S, Tian Y, Zhou N, Wu C, Li R, Xu W, Xu T, Gu L, Ji F, Xu L, Lu L. Effects of Hydroxylated Lecithin on Growth Performance, Serum Enzyme Activity, Hormone Levels Related to Lipid Metabolism and Meat Quality in Jiangnan White Goslings. Front Vet Sci 2022; 9:829338. [PMID: 35296058 PMCID: PMC8920548 DOI: 10.3389/fvets.2022.829338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
The objective of the present study was to evaluate the effects of hydroxylated lecithin on growth performance, serum enzyme activity, hormone levels related to lipid metabolism and meat quality in Jiangnan White goslings. Six hundred 1-day-old goslings were randomly divided into five treatments with six replicates and 20 for each replicate. The control group (CG) was fed the basal diet, while the experimental group was fed the basal diet with 50, 100, 200 mg/kg hydroxylated lecithin and 100 mg/kg soy lecithin (HLG50, HLG100, HLG200, and LG100, respectively) in the form of powder. Feed and water were provided ad libitum for 32 days. Compared with the CG, (a) the average daily feed intake was higher (P < 0.05) in HLG100, the final body weight and average daily gain were higher (P < 0.05), and the feed conversion ratio was lower in the HLG200; (b) the alanine aminotransferase, malate dehydrogenase, leptin, glucagon, thyroid hormone, Triiodothyronine contents in the HLG200 were lower (P < 0.05); (c) The breast muscle water holding capacity was higher (P < 0.05) in groups with hydroxylated lecithin, the breast muscle shear force and fiber diameter were lower (P < 0.05) in the HLG100; (d) the inositic acid, intramuscular fat, phospholipid contents were higher (P < 0.05), the triglyceride content was lower (P < 0.05) in HLG100 of the breast muscle; (e) the relative expression of sterol regulatory element-binding protein-1 genes were higher (P < 0.05) in the treated groups of muscles, the phosphorylase kinase gamma subunit 1 gene expression was shown an opposite trend. In comparison with LG100, (a) the feed conversion ratio was lower (P < 0.05) in HLG200; (b) the alanine aminotransferase and adiponectin contents were higher (P < 0.05), the malondialdehyde and free fatty acid contents were lower (P < 0.05) in HLG200; (c) the water holding capacity and intramuscular fat contents in the breast and leg muscles were higher (P < 0.05) in HLG200. The hydroxylated lecithin concentration of 200 mg/kg improved the growth performance, serum enzyme activity, hormone levels related to lipid metabolism, and the meat quality of Jiangnan White goslings.
Collapse
Affiliation(s)
- Hongzhi Wu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Sibo Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yong Tian
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Ning Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Chunqin Wu
- Wenzhou Vocational College of Science and Technology, Wenzhou, China
| | - Ruiqing Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Wenwu Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
| | - Tieshan Xu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Lihong Gu
- Institute of Animal Science & Veterinary, Hainan Academy of Agricultural Science, Haikou, China
| | - Fengjie Ji
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Li Xu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
- Li Xu
| | - Lizhi Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Animal Science & Veterinary, Zhejiang Academy of Agricultural Science, Hangzhou, China
- *Correspondence: Lizhi Lu
| |
Collapse
|
80
|
Del Prato S, Gallwitz B, Holst JJ, Meier JJ. The incretin/glucagon system as a target for pharmacotherapy of obesity. Obes Rev 2022; 23:e13372. [PMID: 34713962 PMCID: PMC9286339 DOI: 10.1111/obr.13372] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022]
Abstract
Obesity is a chronic, multifactorial, relapsing disease. Despite multicomponent lifestyle interventions, including pharmacotherapy, maintaining bodyweight loss is challenging for many people. The pathophysiology of obesity is complex, and currently approved pharmacotherapies only target a few of the many pathways involved; thus, single-targeting agents have limited efficacy. Proglucagon-derived peptides, glucagon, and the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), represent attractive targets for managing obesity and metabolic disorders because they may have direct roles in multiple mechanisms including satiety, energy homeostasis, and lipolytic activity. Unimolecular dual and triple agonists targeting glucagon and incretin hormone receptors have been shown to promote bodyweight loss, lower glucose levels, and reduce food intake in animal models of obesity. Multiple dual receptor agonists are in clinical development for the treatment of obesity, including GLP-1/GIP and GLP-1/glucagon receptor agonists. The extent to which glucagon contributes to treatment effects remains to be understood, but it may promote bodyweight loss by reducing food intake, while concomitant GLP-1 receptor agonism ensures normal glucose control. Further research is required to fully understand the molecular mechanisms of action and metabolic effects of both dual and triple receptor agonists.
Collapse
Affiliation(s)
- Stefano Del Prato
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Baptist Gallwitz
- Department of Internal Medicine IVEberhard Karls UniversityTübingenGermany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center MunichUniversity of TübingenTübingenGermany
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Juris J. Meier
- Division of Diabetology, Katholisches Klinikum Bochum, St. Josef HospitalRuhr UniversityBochumGermany
| |
Collapse
|
81
|
Batchuluun B, Pinkosky SL, Steinberg GR. Lipogenesis inhibitors: therapeutic opportunities and challenges. Nat Rev Drug Discov 2022; 21:283-305. [PMID: 35031766 PMCID: PMC8758994 DOI: 10.1038/s41573-021-00367-2] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 12/12/2022]
Abstract
Fatty acids are essential for survival, acting as bioenergetic substrates, structural components and signalling molecules. Given their vital role, cells have evolved mechanisms to generate fatty acids from alternative carbon sources, through a process known as de novo lipogenesis (DNL). Despite the importance of DNL, aberrant upregulation is associated with a wide variety of pathologies. Inhibiting core enzymes of DNL, including citrate/isocitrate carrier (CIC), ATP-citrate lyase (ACLY), acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS), represents an attractive therapeutic strategy. Despite challenges related to efficacy, selectivity and safety, several new classes of synthetic DNL inhibitors have entered clinical-stage development and may become the foundation for a new class of therapeutics. De novo lipogenesis (DNL) is vital for the maintenance of whole-body and cellular homeostasis, but aberrant upregulation of the pathway is associated with a broad range of conditions, including cardiovascular disease, metabolic disorders and cancers. Here, Steinberg and colleagues provide an overview of the physiological and pathological roles of the core DNL enzymes and assess strategies and agents currently in development to therapeutically target them.
Collapse
Affiliation(s)
- Battsetseg Batchuluun
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
82
|
Ko S, Yeom E, Chun YL, Mun H, Howard-McGuire M, Millison NT, Jung J, Lee KP, Lee C, Lee KS, Delaney JR, Yoon JH. Profiling of RNA-binding Proteins Interacting With Glucagon and Adipokinetic Hormone mRNAs. J Lipid Atheroscler 2022; 11:55-72. [PMID: 35118022 PMCID: PMC8792818 DOI: 10.12997/jla.2022.11.1.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/05/2021] [Accepted: 07/20/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Glucagon in mammals and its homolog (adipokinetic hormone [AKH] in Drosophila melanogaster) are peptide hormones which regulate lipid metabolism by breaking down triglycerides. Although regulatory mechanisms of glucagon and AKH expression have been widely studied, post-transcriptional gene expression of glucagon has not been investigated thoroughly. In this study, we aimed to profile proteins binding with Gcg messenger RNA (mRNA) in mouse and Akh mRNA in Drosophila. METHODS Drosophila Schneider 2 (S2) and mouse 3T3-L1 cell lysates were utilized for affinity pull down of Akh and Gcg mRNA respectively using biotinylated anti-sense DNA oligoes against target mRNAs. Mass spectrometry and computational network analysis revealed mRNA-interacting proteins residing in functional proximity. RESULTS We observed that 1) 91 proteins interact with Akh mRNA from S2 cell lysates, 2) 34 proteins interact with Gcg mRNA from 3T3-L1 cell lysates. 3) Akh mRNA interactome revealed clusters of ribosomes and known RNA-binding proteins (RBPs). 4) Gcg mRNA interactome revealed mRNA-binding proteins including Plekha7, zinc finger protein, carboxylase, lipase, histone proteins and a cytochrome, Cyp2c44. 5) Levels of Gcg mRNA and its interacting proteins are elevated in skeletal muscles isolated from old mice compared to ones from young mice. CONCLUSION Akh mRNA in S2 cells are under active translation in a complex of RBPs and ribosomes. Gcg mRNA in mouse precursor adipocyte is in a condition distinct from Akh mRNA due to biochemical interactions with a subset of RBPs and histones. We anticipate that our study contributes to investigating regulatory mechanisms of Gcg and Akh mRNA decay, translation, and localization.
Collapse
Affiliation(s)
- Seungbeom Ko
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Eunbyul Yeom
- Neurophysiology and Metabolism Research Group, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Yoo Lim Chun
- Department of Biomedical Science, Graduation School, Kyung Hee University, Seoul, Korea
| | - Hyejin Mun
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Marina Howard-McGuire
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Nathan T. Millison
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Changhan Lee
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kyu-Sun Lee
- Neurophysiology and Metabolism Research Group, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Joe R. Delaney
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Je-Hyun Yoon
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
83
|
Memaj P, Jornayvaz FR. Non-alcoholic fatty liver disease in type 1 diabetes: Prevalence and pathophysiology. Front Endocrinol (Lausanne) 2022; 13:1031633. [PMID: 36531463 PMCID: PMC9752856 DOI: 10.3389/fendo.2022.1031633] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most frequent chronic liver disease in the general population with a global prevalence of 25%. It is often associated with metabolic syndrome and type 2 diabetes, as insulin resistance and hyperinsulinemia are known to be favoring factors. Recent studies have described growing incidence of NAFLD in type 1 diabetes (T1D) as well. Although increasing prevalence of metabolic syndrome in these patients seems to explain part of this increase in NAFLD, other underlying mechanisms may participate in the emergence of NAFLD. Notably, some genetic factors are more associated with fatty liver disease, but their prevalence in T1D has not been evaluated. Moreover, oxidative stress, poor glucose control and long-lasting hyperglycemia, as well as exogenous insulin administration play an important role in intrahepatic fat homeostasis. The main differential diagnosis of NAFLD in T1D is glycogenic hepatopathy, which needs to be considered mostly in T1D patients with poor glycemic control. This article aims to review the prevalence and pathophysiology of NAFLD in T1D and open perspectives for clinicians taking care of T1D patients with potential hepatopathy.
Collapse
Affiliation(s)
- Plator Memaj
- Division of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - François R. Jornayvaz
- Division of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, Geneva University, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva University, Geneva, Switzerland
- *Correspondence: François R. Jornayvaz,
| |
Collapse
|
84
|
Park BG, Kim GM, Lee HJ, Ryu JH, Kim DH, Seong JY, Kim S, Park ZY, Kim YJ, Lee J, Kim JI. Antiobesity therapeutics with complementary dual-agonist activities at glucagon and glucagon-like peptide 1 receptors. Diabetes Obes Metab 2022; 24:50-60. [PMID: 34491605 DOI: 10.1111/dom.14546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/17/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022]
Abstract
AIM To develop more effective and long-lasting antiobesity and antidiabetic therapeutics by employing novel chemical modifications of glucagon-like peptide-1 receptor (GLP-1R) agonists. METHODS We constructed novel unimolecular dual agonists of GLP-1R and glucagon receptor prepared by linking sEx-4 and native glucagon (GCG) via lysine or triazole [sEx4-GCG(K) and sEx4-GCG(T), respectively] and evaluated their antiobesity and antidiabetic efficacy in the diabetic and obese mouse model. RESULTS Both sEx4-GCG(K) and sEx4-GCG(T) showed the beneficial metabolic effects of GLP-1 and glucagon: they promoted weight loss and ameliorated insulin resistance and hepatic steatosis. They also increased thermogenesis in brown adipose tissue, and lipolysis and β-oxidation in white adipose tissue, with concomitant suppression of lipogenesis. Furthermore, both dual agonists activated the 5'-AMP-activated protein kinase signalling pathway and prevented palmitate-induced oxidative stress in skeletal muscle cells. CONCLUSION Through their complementary dual agonism, sEx4-GCG(T) and sEx4-GCG(K) induce more marked weight loss and metabolic improvements than conventional agonists, and could be developed as novel therapeutic agents for the treatment of obesity and associated metabolic disorders in humans.
Collapse
Affiliation(s)
- Bong Gyu Park
- Department of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Gyeong Min Kim
- Department of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Hye-Jin Lee
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae Ha Ryu
- Pilot Plant, Anygen, Gwangju, Republic of Korea
| | - Dong-Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae-Young Seong
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | - Soojeong Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Zee-Yong Park
- Department of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Young-Joon Kim
- Department of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Jae Il Kim
- Department of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
- Pilot Plant, Anygen, Gwangju, Republic of Korea
| |
Collapse
|
85
|
Sohrabipour S, Sharifi MR, Sharifi M, Talebi A, Soltani N. Combination Therapy with GABA and MgSO 4 Improves Insulin Sensitivity in Type 2 Diabetic Rat. Int J Endocrinol 2022; 2022:2144615. [PMID: 35211170 PMCID: PMC8863457 DOI: 10.1155/2022/2144615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA) and magnesium sulfate (MgSO4) play a crucial role in glycemic control. Therefore, we studied the effect of combination therapy with GABA and MgSO4 to improve insulin sensitivity in diabetes induced by streptozotocin as well as high-fat diet in a diabetic rat model. Design and Methods. Forty randomly selected rats were assigned to four groups: nondiabetic control group was fed the normal diet, insulin-resistant diabetic rat model was induced by streptozotocin and high-fat diet, GABA + MgSO4 group received GABA and MgSO4, and insulin group was treated with insulin. Body weight, abdominal fat, blood glucose, serum insulin, and glucagon concentration were measured. The glucose clamp technique, glucose tolerance test, and insulin tolerance test were performed to study insulin sensitivity. Also, the expressions of glucose 6 phosphatase, glucagon receptor, and phosphoenolpyruvate carboxykinase genes in liver were assessed for the gluconeogenesis pathway. Protein translocation and glucose transporter 4 (Glut4) genes expression in muscle were also assessed. RESULTS Combination of GABA + MgSO4 or insulin therapy enhanced insulin level, glycemic control, glucose and insulin tolerance test, some enzymes expression in the gluconeogenesis pathway, body fat, body weight, and glucagon receptor in diabetic rats. Moreover, an increase was observed in protein and gene expression of Glut4. Insulin sensitivity in combination therapy was more than the insulin group. CONCLUSIONS GABA and MgSO4 enhanced insulin sensitivity via increasing Glut4 and reducing the gluconeogenesis enzyme and glucagon receptor gene expressions.
Collapse
Affiliation(s)
- Shahla Sohrabipour
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Reza Sharifi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ardeshir Talebi
- Department of Clinical Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nepton Soltani
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
86
|
Chen CC, Hsu LW, Chen KD, Chiu KW, Chen CL, Huang KT. Emerging Roles of Calcium Signaling in the Development of Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2021; 23:ijms23010256. [PMID: 35008682 PMCID: PMC8745268 DOI: 10.3390/ijms23010256] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 01/10/2023] Open
Abstract
The liver plays a central role in energy metabolism. Dysregulated hepatic lipid metabolism is a major cause of non-alcoholic fatty liver disease (NAFLD), a chronic liver disorder closely linked to obesity and insulin resistance. NAFLD is rapidly emerging as a global health problem with currently no approved therapy. While early stages of NAFLD are often considered benign, the disease can progress to an advanced stage that involves chronic inflammation, with increased risk for developing end-stage disease including fibrosis and liver cancer. Hence, there is an urgent need to identify potential pharmacological targets. Ca2+ is an essential signaling molecule involved in a myriad of cellular processes. Intracellular Ca2+ is intricately compartmentalized, and the Ca2+ flow is tightly controlled by a network of Ca2+ transport and buffering proteins. Impaired Ca2+ signaling is strongly associated with endoplasmic reticulum stress, mitochondrial dysfunction and autophagic defects, all of which are etiological factors of NAFLD. In this review, we describe the recent advances that underscore the critical role of dysregulated Ca2+ homeostasis in lipid metabolic abnormalities and discuss the feasibility of targeting Ca2+ signaling as a potential therapeutic approach.
Collapse
Affiliation(s)
- Chien-Chih Chen
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Li-Wen Hsu
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
| | - Kuang-Den Chen
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - King-Wah Chiu
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chao-Long Chen
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
| | - Kuang-Tzu Huang
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Correspondence: ; Tel.: +886-7-731-7123 (ext. 8193)
| |
Collapse
|
87
|
O'Connell SM, O'Toole NMA, Cronin CN, Saat-Murphy C, McElduff P, King BR, Smart CE, Shafat A. Does dietary fat cause a dose dependent glycemic response in youth with type 1 diabetes? Pediatr Diabetes 2021; 22:1108-1114. [PMID: 34719089 DOI: 10.1111/pedi.13273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/28/2021] [Accepted: 10/14/2021] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE To determine the glycemic impact of dietary fat alone consumed without prandial insulin in individuals with T1D. RESEARCH DESIGN AND METHODS Thirty participants with T1D (aged 8-18 years) consumed a test drink with either 20 g glucose or 1, 13, 26, 39, 51 g of fat with negligible carbohydrate/protein on 6 consecutive evenings, in a randomized order without insulin. Continuous glucose monitoring was used to measure glucose levels for 8 h postprandially. Primary outcome was mean glycemic excursion at each 30 min interval for each test condition. Generalized linear mixed models with a random effect for people with diabetes were used to test for an increase in blood glucose excursion with increasing quantity of fat. RESULTS Glycemic excursions after 20 g glucose were higher than after fat drinks over the first 2 h (p < 0.05). Glycemic excursion for the fat drinks demonstrated a dose response, statistically significant from 4 h (p = 0.026), such that increasing loads of fat caused a proportionally larger increase in glycemic excursion, remaining statistically significant until 8 h (p < 0.05). Overall, for every 10 g fat added to the drink, glucose concentrations rose by a mean of 0.28 mmol L-1 from 330 min (95% CI 0.15 to 0.39, p < 0.001). CONCLUSIONS Fat ingested without other macronutrients increases glucose excursions from 4 to 8 h after ingestion, in a dose dependent manner. These observations may impact on insulin dosing for high-fat foods in individuals with T1D.
Collapse
Affiliation(s)
- Susan M O'Connell
- Paediatrics and Child Health, Cork University Hospital, Cork, Ireland.,Diabetes and Endocrinology, Children's Health Ireland at Crumlin, Dublin, Ireland.,Paediatrics, Royal College of Surgeons of Ireland, Dublin, Ireland
| | - Nora M A O'Toole
- Paediatrics and Child Health, Cork University Hospital, Cork, Ireland
| | - Conor N Cronin
- Paediatrics and Child Health, Cork University Hospital, Cork, Ireland
| | - Chen Saat-Murphy
- Physiology, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Patrick McElduff
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - Bruce R King
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia.,Department of Diabetes and Endocrinology, John Hunter Children's Hospital, Newcastle, Australia
| | - Carmel E Smart
- Department of Diabetes and Endocrinology, John Hunter Children's Hospital, Newcastle, Australia
| | - Amir Shafat
- Physiology, School of Medicine, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
88
|
Mertens J, De Block C, Spinhoven M, Driessen A, Francque SM, Kwanten WJ. Hepatopathy Associated With Type 1 Diabetes: Distinguishing Non-alcoholic Fatty Liver Disease From Glycogenic Hepatopathy. Front Pharmacol 2021; 12:768576. [PMID: 34759828 PMCID: PMC8573337 DOI: 10.3389/fphar.2021.768576] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune destruction of pancreatic β-cells results in the permanent loss of insulin production in type 1 diabetes (T1D). The daily necessity to inject exogenous insulin to treat hyperglycemia leads to a relative portal vein insulin deficiency and potentiates hypoglycemia which can induce weight gain, while daily fluctuations of blood sugar levels affect the hepatic glycogen storage and overall metabolic control. These, among others, fundamental characteristics of T1D are associated with the development of two distinct, but in part clinically similar hepatopathies, namely non-alcoholic fatty liver disease (NAFLD) and glycogen hepatopathy (GlyH). Recent studies suggest that NAFLD may be increasingly common in T1D because more people with T1D present with overweight and/or obesity, linked to the metabolic syndrome. GlyH is a rare but underdiagnosed complication hallmarked by extremely brittle metabolic control in, often young, individuals with T1D. Both hepatopathies share clinical similarities, troubling both diagnosis and differentiation. Since NAFLD is increasingly associated with cardiovascular and chronic kidney disease, whereas GlyH is considered self-limiting, awareness and differentiation between both condition is important in clinical care. The exact pathogenesis of both hepatopathies remains obscure, hence licensed pharmaceutical therapy is lacking and general awareness amongst physicians is low. This article aims to review the factors potentially contributing to fatty liver disease or glycogen storage disruption in T1D. It ends with a proposal for clinicians to approach patients with T1D and potential hepatopathy.
Collapse
Affiliation(s)
- Jonathan Mertens
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium.,Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Wilrijk, Belgium
| | - Christophe De Block
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Wilrijk, Belgium
| | - Maarten Spinhoven
- Department of Radiology, Antwerp University Hospital, Edegem, Belgium
| | - Ann Driessen
- Department of Pathology, Antwerp University Hospital, Antwerp, Belgium.,CORE, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sven M Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Wilrijk, Belgium
| | - Wilhelmus J Kwanten
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium.,Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
89
|
Insects as a New Complex Model in Hormonal Basis of Obesity. Int J Mol Sci 2021; 22:ijms222011066. [PMID: 34681728 PMCID: PMC8540125 DOI: 10.3390/ijms222011066] [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: 09/06/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 11/30/2022] Open
Abstract
Nowadays, one of the biggest problems in healthcare is an obesity epidemic. Consumption of cheap and low-quality energy-rich diets, low physical activity, and sedentary work favor an increase in the number of obesity cases within many populations/nations. This is a burden on society, public health, and the economy with many deleterious consequences. Thus, studies concerning this disorder are extremely needed, including searching for new, effective, and fitting models. Obesity may be related, among other factors, to disrupting adipocytes activity, disturbance of metabolic homeostasis, dysregulation of hormonal balance, cardiovascular problems, or disorders in nutrition which may lead to death. Because of the high complexity of obesity, it is not easy to find an ideal model for its studies which will be suitable for genetic and physiological analysis including specification of different compounds’ (hormones, neuropeptides) functions, as well as for signaling pathways analysis. In recent times, in search of new models for human diseases there has been more and more attention paid to insects, especially in neuro-endocrine regulation. It seems that this group of animals might also be a new model for human obesity. There are many arguments that insects are a good, multidirectional, and complex model for this disease. For example, insect models can have similar conservative signaling pathways (e.g., JAK-STAT signaling pathway), the presence of similar hormonal axis (e.g., brain–gut axis), or occurrence of structural and functional homologues between neuropeptides (e.g., neuropeptide F and human neuropeptide Y, insulin-like peptides, and human insulin) compared to humans. Here we give a hint to use insects as a model for obesity that can be used in multiple ways: as a source of genetic and peptidomic data about etiology and development correlated with obesity occurrence as well as a model for novel hormonal-based drug activity and their impact on mechanism of disease occurrence.
Collapse
|
90
|
Hope DCD, Vincent ML, Tan TMM. Striking the Balance: GLP-1/Glucagon Co-Agonism as a Treatment Strategy for Obesity. Front Endocrinol (Lausanne) 2021; 12:735019. [PMID: 34566894 PMCID: PMC8457634 DOI: 10.3389/fendo.2021.735019] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/18/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity and Type 2 diabetes represent global health challenges, and there is an unmet need for long-lasting and effective pharmacotherapies. Although long-acting glucagon-like peptide-1 (GLP-1) analogues are now in routine use for diabetes and are now being utilised for obesity per se, the need for ever better treatments has driven the development of co-agonists, with the theoretical advantages of improved efficacy by targeting multiple pathways and reduced adverse effects. In this review, we highlight the past and present progress in our understanding and development of treatments based on GLP-1/glucagon co-agonism. We also reflect on the divergent effects of varying the GLP-1:glucagon activity and ratio in the context of pre-clinical and human clinical trial findings. In particular, the multiple metabolic actions of glucagon highlight the importance of understanding the contributions of individual hormone action to inform the safe, effective and tailored use of GLP-1/glucagon co-agonists to target weight loss and metabolic disease in the future.
Collapse
Affiliation(s)
| | | | - Tricia M. M. Tan
- Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
91
|
Free fatty acids, glicentin and glucose-dependent insulinotropic polypeptide as potential major determinants of fasting substrate oxidation. Sci Rep 2021; 11:16642. [PMID: 34404813 PMCID: PMC8371005 DOI: 10.1038/s41598-021-95750-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/21/2021] [Indexed: 01/07/2023] Open
Abstract
The selection of carbohydrates or fat to generate intracellular energy is thought to be crucial for long-term metabolic health. While most studies assess fuel selection after a metabolic challenge, the determinants of substrate oxidation in the fasted state remain largely unexplored. We therefore assessed the respiratory quotient by indirect calorimetry as a read-out for substrate oxidation following an overnight fast. This cross-sectional analysis consisted of 192 (92 women, 100 men) either lean or obese participants. Following an overnight fast, the respiratory quotient (RQ) was assessed, after which a 5-point 75-g oral glucose tolerance test was performed. Unlike glucose and insulin, fasting free fatty acids (FFA) correlated negatively with fasting RQ (p < 0.0001). Participants with high levels of the ketone body β-hydroxybutyric acid had significantly lower RQ values. Fasting levels of glucose-dependent insulinotropic polypeptide (GIP) and glicentin were positively associated with fasting RQ (all p ≤ 0.03), whereas GLP-1 showed no significant association. Neither BMI, nor total body fat, nor body fat distribution correlated with fasting RQ. No relationship between the RQ and diabetes or the metabolic syndrome could be observed. In the fasting state, FFA concentrations were strongly linked to the preferentially oxidized substrate. Our data did not indicate any relationship between fasting substrate oxidation and metabolic diseases, including obesity, diabetes, and the metabolic syndrome. Since glicentin and GIP are linked to fuel selection in the fasting state, novel therapeutic approaches that target these hormones may have the potential to modulate substrate oxidation.
Collapse
|
92
|
He J, Liu R, Zheng W, Guo H, Yang Y, Zhao R, Yao W. High ambient temperature exposure during late gestation disrupts glycolipid metabolism and hepatic mitochondrial function tightly related to gut microbial dysbiosis in pregnant mice. Microb Biotechnol 2021; 14:2116-2129. [PMID: 34272826 PMCID: PMC8449678 DOI: 10.1111/1751-7915.13893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/03/2021] [Indexed: 12/11/2022] Open
Abstract
As global warming intensifies, emerging evidence has demonstrated high ambient temperature during pregnancy negatively affects maternal physiology with compromised pregnant outcomes; however, little is known about the roles of gut microbiota and its underlying mechanisms in this process. Here, for the first time, we explored the potential mechanisms of gut microbiota involved in the disrupted glycolipid metabolism via hepatic mitochondrial function. Our results indicate heat stress (HS) reduces fat and protein contents and serum levels of insulin and triglyceride (TG), while increases that of non-esterified fatty acid (NEFA), β-hydroxybutyric acid (B-HBA), creatinine and blood urea nitrogen (BUN) (P < 0.05). Additionally, HS downregulates both mitochondrial genes (mtDNA) and nuclear encoding mitochondrial functional genes with increasing serum levels of malondialdehyde (MDA) and 8-hydroxydeoxyguanosine (8-OHdG) (P < 0.05). Regarding microbial response, HS boosts serum levels of lipopolysaccharide (LPS) (P < 0.05) and alters β-diversity (ANOSIM, P < 0.01), increasing the proportions of Escherichia-Shigella, Acinetobacter and Klebsiella (q < 0.05), while reducing that of Ruminiclostridium, Blautia, Lachnospiraceae_NK4A136_group, Clostridium VadinBB60 and Muribaculaceae (q < 0.05). PICRUSt analysis predicts that HS upregulates 11 KEGG pathways, mainly including bile secretion and bacterial invasion of epithelial cells. The collective results suggest that microbial dysbiosis due to late gestational HS has strong associations with damaged hepatic mitochondrial function and disrupted metabolic profiles.
Collapse
Affiliation(s)
- Jianwen He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.,Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Riliang Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weijiang Zheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Huiduo Guo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yunnan Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruqian Zhao
- Key Lab of Animal Physiology and Biochemistry, Nanjing Agricultural University, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing, 210095, China
| | - Wen Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.,Key Lab of Animal Physiology and Biochemistry, Nanjing Agricultural University, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing, 210095, China
| |
Collapse
|
93
|
Vega RB, Whytock KL, Gassenhuber J, Goebel B, Tillner J, Agueusop I, Truax AD, Yu G, Carnero E, Kapoor N, Gardell S, Sparks LM, Smith SR. A Metabolomic Signature of Glucagon Action in Healthy Individuals With Overweight/Obesity. J Endocr Soc 2021; 5:bvab118. [PMID: 34337278 PMCID: PMC8317630 DOI: 10.1210/jendso/bvab118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Indexed: 11/19/2022] Open
Abstract
Context Glucagon is produced and released from the pancreatic alpha-cell to regulate glucose levels during periods of fasting. The main target for glucagon action is the liver, where it activates gluconeogenesis and glycogen breakdown; however, glucagon is postulated to have other roles within the body. Objective We sought to identify the circulating metabolites that would serve as markers of glucagon action in humans. Methods In this study (NCT03139305), we performed a continuous 72-hour glucagon infusion in healthy individuals with overweight/obesity. Participants were randomized to receive glucagon 12.5 ng/kg/min (GCG 12.5), glucagon 25 ng/kg/min (GCG 25), or a placebo control. A comprehensive metabolomics analysis was then performed from plasma isolated at several time points during the infusion to identify markers of glucagon activity. Results Glucagon (GCG 12.5 and GCG 25) resulted in significant changes in the plasma metabolome as soon as 4 hours following infusion. Pathways involved in amino acid metabolism were among the most affected. Rapid and sustained reduction of a broad panel of amino acids was observed. Additionally, time-dependent changes in free fatty acids and diacylglycerol and triglyceride species were observed. Conclusion These results define a distinct signature of glucagon action that is broader than the known changes in glucose levels. In particular, the robust changes in amino acid levels may prove useful to monitor changes induced by glucagon in the context of additional glucagon-like peptide-1 or gastric inhibitory polypeptide treatment, as these agents also elicit changes in glucose levels.
Collapse
Affiliation(s)
- Rick B Vega
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | | | | | | | | | | | - Gongxin Yu
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Elvis Carnero
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Nidhi Kapoor
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Stephen Gardell
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, FL 32804, USA
| |
Collapse
|
94
|
Nahra R, Wang T, Gadde KM, Oscarsson J, Stumvoll M, Jermutus L, Hirshberg B, Ambery P. Effects of Cotadutide on Metabolic and Hepatic Parameters in Adults With Overweight or Obesity and Type 2 Diabetes: A 54-Week Randomized Phase 2b Study. Diabetes Care 2021; 44:1433-1442. [PMID: 34016612 PMCID: PMC8247525 DOI: 10.2337/dc20-2151] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/23/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Cotadutide, a dual GLP-1 and glucagon receptor agonist, is under development for nonalcoholic steatohepatitis (NASH) and chronic kidney disease with type 2 diabetes. The effects of cotadutide on hepatic and metabolic parameters were evaluated in participants with overweight/obesity and type 2 diabetes. RESEARCH DESIGN AND METHODS In this phase 2b study, 834 adults with BMI ≥25 kg/m2 and type 2 diabetes inadequately controlled with metformin (glycated hemoglobin A1c [HbA1c] of 7.0%-10.5% [53-91 mmol/mol]) were randomized to double-blind cotadutide 100 μg (n = 100), 200 μg (n = 256), or 300 μg (n = 256); placebo (n = 110); or open-label liraglutide 1.8 mg (n = 110)-all administered subcutaneously. Coprimary end points were changes in HbA1c and body weight at week 14. The originally randomized interventions were continued to week 54. Liver damage biomarkers and liver fibrosis algorithms were assessed. RESULTS Cotadutide significantly decreased HbA1c and body weight at weeks 14 and 54 versus placebo (all P < 0.001). Improvements in lipid profile, AST and ALT levels, propeptide of type III collagen level, fibrosis-4 index, and nonalcoholic fatty liver disease fibrosis score were observed with cotadutide 300 μg versus placebo, but not with liraglutide. Weight loss with cotadutide 200 μg was similar to that with liraglutide 1.8 mg and greater with cotadutide 300 μg versus liraglutide 1.8 mg. The most common adverse events with cotadutide (nausea, 35%; vomiting, 17%) decreased over time. CONCLUSIONS Cotadutide treatment for 54 weeks improved glycemic control and weight loss in participants with overweight/obesity and type 2 diabetes. Ad hoc analyses demonstrated improvements in hepatic parameters and support further evaluation of cotadutide in NASH.
Collapse
Affiliation(s)
- Rajaa Nahra
- Clinical Metabolism, Late-stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Tao Wang
- Clinical Biostatistics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Kishore M Gadde
- Clinical Trials Unit, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Jan Oscarsson
- Clinical Metabolism, Late-stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Michael Stumvoll
- Division of Endocrinology, University Hospital Leipzig, Leipzig, Germany
| | - Lutz Jermutus
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, U.K
| | - Boaz Hirshberg
- Research and Early Development, Early CVRM, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Philip Ambery
- Clinical Metabolism, Late-stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
95
|
Metabolic Disturbances in Rat Sublines with Constitutionally Altered Serotonin Homeostasis. Int J Mol Sci 2021; 22:ijms22105400. [PMID: 34065591 PMCID: PMC8161318 DOI: 10.3390/ijms22105400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 01/31/2023] Open
Abstract
Central and peripheral serotonin (5HT) have opposing functions in the regulation of energy homeostasis. Both increasing 5HT signaling in the brain and decreasing 5HT signaling in the periphery have been proposed as potential treatments for obesity. This study investigates the relationship between constitutionally high or low 5HT activity and systemic net energy balance. Two sublines of rats with high and low whole-body 5HT tone, obtained by selective breeding for platelet 5HT parameters, were examined for fat accumulation in different white adipose tissue (WAT) depots, glucose/insulin tolerance, blood metabolic parameters, and expression of various metabolic genes. High-5HT animals, unlike their low-5HT counterparts, developed widespread intra-abdominal obesity associated with glucose and insulin intolerance, which worsened with age. They also had elevated blood glucose and lipid parameters but showed no significant changes in circulating leptin, resistin, and adipsin levels. Surprisingly, adiponectin levels were increased in plasma but reduced in the WAT of high-5HT rats. A limited number of metabolic genes belonging to different functional classes showed differential expression in WAT of high-5HT compared to low-5HT rats. Overall, a constitutive increase in 5HT tone is associated with a positive energy balance acting through subtle dysregulation of a broad spectrum of metabolic pathways.
Collapse
|
96
|
Zeigerer A, Sekar R, Kleinert M, Nason S, Habegger KM, Müller TD. Glucagon's Metabolic Action in Health and Disease. Compr Physiol 2021; 11:1759-1783. [PMID: 33792899 PMCID: PMC8513137 DOI: 10.1002/cphy.c200013] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Discovered almost simultaneously with insulin, glucagon is a pleiotropic hormone with metabolic action that goes far beyond its classical role to increase blood glucose. Albeit best known for its ability to directly act on the liver to increase de novo glucose production and to inhibit glycogen breakdown, glucagon lowers body weight by decreasing food intake and by increasing metabolic rate. Glucagon further promotes lipolysis and lipid oxidation and has positive chronotropic and inotropic effects in the heart. Interestingly, recent decades have witnessed a remarkable renaissance of glucagon's biology with the acknowledgment that glucagon has pharmacological value beyond its classical use as rescue medication to treat severe hypoglycemia. In this article, we summarize the multifaceted nature of glucagon with a special focus on its hepatic action and discuss the pharmacological potential of either agonizing or antagonizing the glucagon receptor for health and disease. © 2021 American Physiological Society. Compr Physiol 11:1759-1783, 2021.
Collapse
Affiliation(s)
- Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Revathi Sekar
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Maximilian Kleinert
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Shelly Nason
- Comprehensive Diabetes Center, Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirk M. Habegger
- Comprehensive Diabetes Center, Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Timo D. Müller
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomics, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| |
Collapse
|
97
|
Barella LF, Jain S, Kimura T, Pydi SP. Metabolic roles of G protein-coupled receptor signaling in obesity and type 2 diabetes. FEBS J 2021; 288:2622-2644. [PMID: 33682344 DOI: 10.1111/febs.15800] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/31/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022]
Abstract
The incidence of obesity and type 2 diabetes (T2D) has been increasing steadily worldwide. It is estimated that by 2045 more than 800 million people will be suffering from diabetes. Despite the advancements in modern medicine, more effective therapies for treating obesity and T2D are needed. G protein-coupled receptors (GPCRs) have emerged as important drug targets for various chronic diseases, including obesity, T2D, and liver diseases. During the past two decades, many laboratories worldwide focused on understanding the role of GPCR signaling in regulating glucose metabolism and energy homeostasis. The information gained from these studies can guide the development of novel therapeutic agents. In this review, we summarize recent studies providing insights into the role of GPCR signaling in peripheral, metabolically important tissues such as pancreas, liver, skeletal muscle, and adipose tissue, focusing primarily on the use of mutant animal models and human data.
Collapse
Affiliation(s)
- Luiz F Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.,Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Takefumi Kimura
- 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.,Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, India
| |
Collapse
|
98
|
Nason SR, Antipenko J, Presedo N, Cunningham SE, Pierre TH, Kim T, Paul JR, Holleman C, Young ME, Gamble KL, Finan B, DiMarchi R, Hunter CS, Kharitonenkov A, Habegger KM. Glucagon receptor signaling regulates weight loss via central KLB receptor complexes. JCI Insight 2021; 6:141323. [PMID: 33411693 PMCID: PMC7934938 DOI: 10.1172/jci.insight.141323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 12/29/2020] [Indexed: 01/15/2023] Open
Abstract
Glucagon regulates glucose and lipid metabolism and promotes weight loss. Thus, therapeutics stimulating glucagon receptor (GCGR) signaling are promising for obesity treatment; however, the underlying mechanism(s) have yet to be fully elucidated. We previously identified that hepatic GCGR signaling increases circulating fibroblast growth factor 21 (FGF21), a potent regulator of energy balance. We reported that mice deficient for liver Fgf21 are partially resistant to GCGR-mediated weight loss, implicating FGF21 as a regulator of glucagon’s weight loss effects. FGF21 signaling requires an obligate coreceptor (β-Klotho, KLB), with expression limited to adipose tissue, liver, pancreas, and brain. We hypothesized that the GCGR-FGF21 system mediates weight loss through a central mechanism. Mice deficient for neuronal Klb exhibited a partial reduction in body weight with chronic GCGR agonism (via IUB288) compared with controls, supporting a role for central FGF21 signaling in GCGR-mediated weight loss. Substantiating these results, mice with central KLB inhibition via a pharmacological KLB antagonist, 1153, also displayed partial weight loss. Central KLB, however, is dispensable for GCGR-mediated improvements in plasma cholesterol and liver triglycerides. Together, these data suggest GCGR agonism mediates part of its weight loss properties through central KLB and has implications for future treatments of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Shelly R Nason
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Jessica Antipenko
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Natalie Presedo
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Stephen E Cunningham
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Tanya H Pierre
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Teayoun Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Jodi R Paul
- Department of Psychiatry and Behavioral Neurobiology, and
| | - Cassie Holleman
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, and
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Inc., Indianapolis, Indiana, USA
| | - Richard DiMarchi
- Novo Nordisk Research Center Indianapolis, Inc., Indianapolis, Indiana, USA.,Department of Chemistry, College of Arts and Sciences, Indiana University, Bloomington, Indiana, USA
| | - Chad S Hunter
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| | | | - Kirk M Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine
| |
Collapse
|
99
|
Abbas E, Ahmed Siddiqui I, Khan MS, Perveen K, Butt A, Fawwad A. Fasting Glucagon Level in Type 2 Diabetes and Impaired Glucose Tolerance and Its Association With Diabetes-Associated Clinical Parameters: A Study From Karachi, Pakistan. Cureus 2021; 13:e13430. [PMID: 33758717 PMCID: PMC7978393 DOI: 10.7759/cureus.13430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Aim and objective The study aims to analyze fasting glucagon in patients with type 2 diabetes and impaired glucose tolerance and correlate it with anthropometric and biochemical parameters in a large proportion of Pakistani people with diabetes. Methodology The participants of the study were categorized into three groups based on oral glucose tolerance test, as per American Diabetes Association guidelines. Group A consisted of normal glucose tolerance subjects (n=30), Group B consisted of subjects with impaired glucose tolerance (n=30), and Group C had full-blown subjects with type 2 diabetes (n=30). Biochemical parameters, such as fasting glucagon, fasting plasma and 2-hour glucose, glycated hemoglobin, and lipid profile, and anthropometric parameters, such as body mass index (BMI), waist and hip circumference, waist-to-hip ratio, and systolic and diastolic blood pressure, were measured. Results The mean values of fasting glucagon level in Group A, Group B, and Group C were 39.24±4.5, 44.5±8.25, and 49.02±9.15 pg/ml, respectively. Statistically significant difference was not found in fasting glucagon level among these groups (p-value 0.614). Fasting glucagon was positively and independently correlated with 2-hour plasma glucose, systolic blood pressure, diastolic blood pressure, BMI, hip and waist circumference, and hip-to-waist ratio in Group C. In Group B, fasting glucagon was positively correlated with 2-hour plasma glucose, BMI, and hip circumference, while it was not correlated with fasting plasma glucose in both groups. In Group A, fasting glucagon found positively correlated with systolic blood pressure and hip circumference. Conclusion Our observation suggests that fasting plasma glucose is not concomitant with glucagon levels; however, glucagon suppression, after glucose intake, was dysregulated in type 2 diabetes and impaired glucose tolerance. Moreover, glucagon is associated with central obesity in type 2 diabetic patients. .
Collapse
Affiliation(s)
- Eraj Abbas
- Biochemistry, Baqai Medical University, Karachi, PAK
| | | | | | | | - Anum Butt
- Research, Baqai Institute of Diabetology and Endocrinology, Baqai Medical University, Karachi, PAK
| | - Asher Fawwad
- Research, Baqai Institute of Diabetology and Endocrinology, Baqai Medical University, Karachi, PAK
| |
Collapse
|
100
|
Bai X, Jia J, Kang Q, Fu Y, Zhou Y, Zhong Y, Zhang C, Li M. Integrated Metabolomics and Lipidomics Analysis Reveal Remodeling of Lipid Metabolism and Amino Acid Metabolism in Glucagon Receptor-Deficient Zebrafish. Front Cell Dev Biol 2021; 8:605979. [PMID: 33520988 PMCID: PMC7841139 DOI: 10.3389/fcell.2020.605979] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
The glucagon receptor (GCGR) is activated by glucagon and is essential for glucose, amino acid, and lipid metabolism of animals. GCGR blockade has been demonstrated to induce hypoglycemia, hyperaminoacidemia, hyperglucagonemia, decreased adiposity, hepatosteatosis, and pancreatic α cells hyperplasia in organisms. However, the mechanism of how GCGR regulates these physiological functions is not yet very clear. In our previous study, we revealed that GCGR regulated metabolic network at transcriptional level by RNA-seq using GCGR mutant zebrafish (gcgr -/-). Here, we further performed whole-organism metabolomics and lipidomics profiling on wild-type and gcgr -/- zebrafish to study the changes of metabolites. We found 107 significantly different metabolites from metabolomics analysis and 87 significantly different lipids from lipidomics analysis. Chemical substance classification and pathway analysis integrated with transcriptomics data both revealed that amino acid metabolism and lipid metabolism were remodeled in gcgr-deficient zebrafish. Similar to other studies, our study showed that gcgr -/- zebrafish exhibited decreased ureagenesis and impaired cholesterol metabolism. More interestingly, we found that the glycerophospholipid metabolism was disrupted, the arachidonic acid metabolism was up-regulated, and the tryptophan metabolism pathway was down-regulated in gcgr -/- zebrafish. Based on the omics data, we further validated our findings by revealing that gcgr -/- zebrafish exhibited dampened melatonin diel rhythmicity and increased locomotor activity. These global omics data provide us a better understanding about the role of GCGR in regulating metabolic network and new insight into GCGR physiological functions.
Collapse
Affiliation(s)
- Xuanxuan Bai
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianxin Jia
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qi Kang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yadong Fu
- Center for Circadian Clocks, Soochow University, Suzhou, China.,School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Yingbin Zhong
- Center for Circadian Clocks, Soochow University, Suzhou, China.,School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| |
Collapse
|