1
|
Harahap IA, Kuligowski M, Cieslak A, Kołodziejski PA, Suliburska J. Effect of Tempeh and Daidzein on Calcium Status, Calcium Transporters, and Bone Metabolism Biomarkers in Ovariectomized Rats. Nutrients 2024; 16:651. [PMID: 38474779 DOI: 10.3390/nu16050651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Menopause marks a critical life stage characterized by hormonal changes that significantly impact bone health, leading to a heightened susceptibility to bone fractures. This research seeks to elucidate the impact of daidzein and tempeh on calcium status, calcium transporters, and bone metabolism in an ovariectomized rat model. Forty female Wistar rats, aged 3 months, participated in a two-phase experiment. The initial phase involved inducing a calcium deficit, while the second phase comprised dietary interventions across five groups: Sham (S) and Ovariectomy (O) with a standard diet, O with bisphosphonate (OB), O with pure daidzein (OD), and O with tempeh (OT). Multiple parameters, encompassing calcium levels, calcium transporters, bone histopathology, and serum bone metabolism markers, were evaluated. The findings revealed that the OT group showcased heightened levels of bone turnover markers, such as pyridinoline, C-telopeptide of type I collagen, bone alkaline phosphatase, and procollagen type I N-terminal propeptide, in contrast to S and O groups, with statistical significance (p < 0.05). Histopathologically, both the OD and OT groups exhibited effects akin to the OB group, indicating a decrease in the surface area occupied by adipocytes in the femoral bone structure, although statistically non-equivalent, supporting the directionally similar trends. Although TRPV5 and TRPV6 mRNA expression levels in the jejunum and duodenum did not display statistically significant differences (p > 0.05), the OD and OT groups exhibited increased expression compared to the O group. We hypothesized that obtained results may be related to the effect of isoflavones on estrogen pathways because of their structurally similar to endogenous estrogen and weak estrogenic properties. In conclusion, the daily consumption of pure daidzein and tempeh could potentially improve and reinstate calcium status, calcium transport, and bone metabolism in ovariectomized rats. Additionally, isoflavone products demonstrate effects similar to bisphosphonate drugs on these parameters in ovariectomized rats.
Collapse
Affiliation(s)
- Iskandar Azmy Harahap
- Department of Human Nutrition and Dietetics, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, 60-624 Poznan, Poland
| | - Maciej Kuligowski
- Department of Food Technology of Plant Origin, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, 60-624 Poznan, Poland
| | - Adam Cieslak
- Department of Animal Nutrition, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, 60-637 Poznan, Poland
| | - Paweł A Kołodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, 60-637 Poznan, Poland
| | - Joanna Suliburska
- Department of Human Nutrition and Dietetics, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, 60-624 Poznan, Poland
| |
Collapse
|
2
|
Jiménez-Sánchez C, Sinturel F, Mezza T, Loizides-Mangold U, Montoya JP, Li L, Di Giuseppe G, Quero G, Guessous I, Jornayvaz F, Schrauwen P, Stenvers DJ, Alfieri S, Giaccari A, Berishvili E, Compagnon P, Bosco D, Riezman H, Dibner C, Maechler P. Lysophosphatidylinositols Are Upregulated After Human β-Cell Loss and Potentiate Insulin Release. Diabetes 2024; 73:93-107. [PMID: 37862465 DOI: 10.2337/db23-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
In this study, we identified new lipid species associated with the loss of pancreatic β-cells triggering diabetes. We performed lipidomics measurements on serum from prediabetic mice lacking β-cell prohibitin-2 (a model of monogenic diabetes) patients without previous history of diabetes but scheduled for pancreaticoduodenectomy resulting in the acute reduction of their β-cell mass (∼50%), and patients with type 2 diabetes (T2D). We found lysophosphatidylinositols (lysoPIs) were the main circulating lipid species altered in prediabetic mice. The changes were confirmed in the patients with acute reduction of their β-cell mass and in those with T2D. Increased lysoPIs significantly correlated with HbA1c (reflecting glycemic control), fasting glycemia, and disposition index, and did not correlate with insulin resistance or obesity in human patients with T2D. INS-1E β-cells as well as pancreatic islets isolated from nondiabetic mice and human donors exposed to exogenous lysoPIs showed potentiated glucose-stimulated and basal insulin secretion. Finally, addition of exogenous lysoPIs partially rescued impaired glucose-stimulated insulin secretion in islets from mice and humans in the diabetic state. Overall, lysoPIs appear to be lipid species upregulated in the prediabetic stage associated with the loss of β-cells and that support the secretory function of the remaining β-cells. ARTICLE HIGHLIGHTS Circulating lysophosphatidylinositols (lysoPIs) are increased in situations associated with β-cell loss in mice and humans such as (pre-)diabetes, and hemipancreatectomy. Pancreatic islets isolated from nondiabetic mice and human donors, as well as INS-1E β-cells, exposed to exogenous lysoPIs exhibited potentiated glucose-stimulated and basal insulin secretion. Addition of exogenous lysoPIs partially rescued impaired glucose-stimulated insulin secretion in islets from mice and humans in the diabetic state. LysoPIs appear as lipid species being upregulated already in the prediabetic stage associated with the loss of β-cells and supporting the function of the remaining β-cells.
Collapse
Affiliation(s)
- Cecilia Jiménez-Sánchez
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Flore Sinturel
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Teresa Mezza
- Pancreas Unit, Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli, Institute of Hospitalization and Scientific Care (IRCCS), Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ursula Loizides-Mangold
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Jonathan Paz Montoya
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lingzi Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Gianfranco Di Giuseppe
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Giuseppe Quero
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Chirurgia Digestiva, Fondazione Policlinico Universitario Gemelli IRCSS Università Cattolica del Sacro Cuore, Rome, Italy
| | - Idris Guessous
- Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - François Jornayvaz
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Division of Endocrinology, Diabetes, Nutrition and Patient Education, Department of Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, the Netherlands
| | - Sergio Alfieri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Chirurgia Digestiva, Fondazione Policlinico Universitario Gemelli IRCSS Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Giaccari
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Ekaterine Berishvili
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Compagnon
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Domenico Bosco
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
- Cell isolation and Transplantation Center, Geneva University Hospitals, Geneva, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, National Centre of Competence in Research Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Charna Dibner
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
3
|
Stamateris RE, Landa-Galvan HV, Sharma RB, Darko C, Redmond D, Rane SG, Alonso LC. Noncanonical CDK4 signaling rescues diabetes in a mouse model by promoting β cell differentiation. J Clin Invest 2023; 133:e166490. [PMID: 37712417 PMCID: PMC10503800 DOI: 10.1172/jci166490] [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: 10/24/2022] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Abstract
Expanding β cell mass is a critical goal in the fight against diabetes. CDK4, an extensively characterized cell cycle activator, is required to establish and maintain β cell number. β cell failure in the IRS2-deletion mouse type 2 diabetes model is, in part, due to loss of CDK4 regulator cyclin D2. We set out to determine whether replacement of endogenous CDK4 with the inhibitor-resistant mutant CDK4-R24C rescued the loss of β cell mass in IRS2-deficient mice. Surprisingly, not only β cell mass but also β cell dedifferentiation was effectively rescued, despite no improvement in whole body insulin sensitivity. Ex vivo studies in primary islet cells revealed a mechanism in which CDK4 intervened downstream in the insulin signaling pathway to prevent FOXO1-mediated transcriptional repression of critical β cell transcription factor Pdx1. FOXO1 inhibition was not related to E2F1 activity, to FOXO1 phosphorylation, or even to FOXO1 subcellular localization, but rather was related to deacetylation and reduced FOXO1 abundance. Taken together, these results demonstrate a differentiation-promoting activity of the classical cell cycle activator CDK4 and support the concept that β cell mass can be expanded without compromising function.
Collapse
Affiliation(s)
- Rachel E. Stamateris
- MD/PhD Program, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Huguet V. Landa-Galvan
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| | - Rohit B. Sharma
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| | - Christine Darko
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| | - David Redmond
- Hartman Institute for Therapeutic Regenerative Medicine, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Sushil G. Rane
- Integrative Cellular Metabolism Section, Diabetes, Endocrinology and Obesity Branch, National Institute for Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Laura C. Alonso
- Division of Endocrinology, Diabetes and Metabolism and the Joan and Sanford I. Weill Center for Metabolic Health and
| |
Collapse
|
4
|
Sun Y, Mehmood A, Giampieri F, Battino MA, Chen X. Insights into the cellular, molecular, and epigenetic targets of gamma-aminobutyric acid against diabetes: a comprehensive review on its mechanisms. Crit Rev Food Sci Nutr 2023:1-18. [PMID: 37694998 DOI: 10.1080/10408398.2023.2255666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Diabetes is a metabolic disease due to impaired or defective insulin secretion and is considered one of the most serious chronic diseases worldwide. Gamma-aminobutyric acid (GABA) is a naturally occurring non-protein amino acid commonly present in a wide range of foods. A number of studies documented that GABA has good anti-diabetic potential. This review summarized the available dietary sources of GABA as well as animal and human studies on the anti-diabetic properties of GABA, while also discussing the underlying mechanisms. GABA may modulate diabetes through various pathways such as inhibiting the activities of α-amylase and α-glucosidase, promoting β-cell proliferation, stimulating insulin secretion from β-cells, inhibiting glucagon secretion from α-cells, improving insulin resistance and glucose tolerance, and increasing antioxidant and anti-inflammatory activities. However, further mechanistic studies on animals and human are needed to confirm the therapeutic effects of GABA against diabetes.
Collapse
Affiliation(s)
- Yu Sun
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Institute of Food Physical Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Arshad Mehmood
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Institute of Food Physical Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Francesca Giampieri
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
| | - Maurizio Antonio Battino
- International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander, Spain
- Department of Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Xiumin Chen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- Institute of Food Physical Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
- International Joint Research Laboratory of Intelligent Agriculture and Agri-products Processing, Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| |
Collapse
|
5
|
Cell Replacement Therapy for Type 1 Diabetes Patients: Potential Mechanisms Leading to Stem-Cell-Derived Pancreatic β-Cell Loss upon Transplant. Cells 2023; 12:cells12050698. [PMID: 36899834 PMCID: PMC10000642 DOI: 10.3390/cells12050698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cell replacement therapy using stem-cell-derived insulin-producing β-like cells (sBCs) has been proposed as a practical cure for patients with type one diabetes (T1D). sBCs can correct diabetes in preclinical animal models, demonstrating the promise of this stem cell-based approach. However, in vivo studies have demonstrated that most sBCs, similarly to cadaveric human islets, are lost upon transplantation due to ischemia and other unknown mechanisms. Hence, there is a critical knowledge gap in the current field concerning the fate of sBCs upon engraftment. Here we review, discuss effects, and propose additional potential mechanisms that could contribute toward β-cell loss in vivo. We summarize and highlight some of the literature on phenotypic loss in β-cells under both steady, stressed, and diseased diabetic conditions. Specifically, we focus on β-cell death, dedifferentiation into progenitors, trans-differentiation into other hormone-expressing cells, and/or interconversion into less functional β-cell subtypes as potential mechanisms. While current cell replacement therapy efforts employing sBCs carry great promise as an abundant cell source, addressing the somewhat neglected aspect of β-cell loss in vivo will further accelerate sBC transplantation as a promising therapeutic modality that could significantly enhance the life quality of T1D patients.
Collapse
|
6
|
Ishihara H. Metabolism-secretion coupling in glucose-stimulated insulin secretion. Diabetol Int 2022; 13:463-470. [PMID: 35693987 PMCID: PMC9174369 DOI: 10.1007/s13340-022-00576-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 02/27/2022] [Indexed: 01/09/2023]
Abstract
Pancreatic β-cells in the islets of Langerhans secrete insulin in response to blood glucose levels. Precise control of the amount of insulin secreted is of critical importance for maintaining systemic carbohydrate homeostasis. It is now well established that glucose induced production of ATP from ADP and the KATP channel closure elevate cytosolic Ca2+, triggering insulin exocytosis in β-cells. However, for full activation of insulin secretion by glucose, other mechanisms besides Ca2+ elevation are needed. These mechanisms are the targets of current research and include intracellular metabolic pathways branching from glycolysis. They are metabolic pathways originating from the TCA cycle intermediates, the glycerolipid/free fatty acid cycle and the pentose phosphate pathway. Signaling effects of these pathways including degradation (removal) of protein SUMOylation, modulation of insulin vesicular energetics, and lipid modulation of exocytotic machinery may converge to fulfill insulin secretion, though the precise mechanisms have yet to be elucidated. This mini-review summarize recent advances in research on metabolic coupling mechanisms functioning in insulin secretion.
Collapse
Affiliation(s)
- Hisamitsu Ishihara
- Division of Diabetes and Metabolism, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610 Japan
| |
Collapse
|
7
|
Oberhauser L, Jiménez-Sánchez C, Madsen JGS, Duhamel D, Mandrup S, Brun T, Maechler P. Glucolipotoxicity promotes the capacity of the glycerolipid/NEFA cycle supporting the secretory response of pancreatic beta cells. Diabetologia 2022; 65:705-720. [PMID: 35018486 DOI: 10.1007/s00125-021-05633-x] [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: 08/01/2021] [Accepted: 10/12/2021] [Indexed: 11/03/2022]
Abstract
AIMS/HYPOTHESIS Chronic exposure of pancreatic beta cells to high glucose and fatty acids has been proposed to induce glucolipotoxicity. However, contradictory results suggest adaptations of the beta cells, which might be instrumental for partial preservation of the secretory response. In this context, we delineated the expression pattern of genes related to lipid pathways along with fat storage/mobilisation during glucose-stimulated insulin secretion. METHODS Insulin-secreting cells were cultured for 3 days at different glucose concentrations (5.5, 11.1, 25 mmol/l) without or with BSA-complexed 0.4 mmol/l palmitate and oleate. Then, transcriptomic analyses of lipid pathways were performed in human islets by RNA-Seq and in INS-1E cells and rat islets by quantitative RT-PCR. Storage of fat was assessed in INS-1E cells by electron microscopy and Bodipy staining, which was also used for measuring lipid mobilisation rate. The secretory response was monitored during acute 15 mmol/l glucose stimulation using online luminescence assay for INS-1E cells and by radioimmunoassay for rat islets. RESULTS In human islets, chronic exposure to palmitate and oleate modified expression of a panel of genes involved in lipid handling. Culture at 25 mmol/l glucose upregulated genes encoding for enzymes of the glycerolipid/NEFA cycle and downregulated receptors implicated in fatty acid signalling. Similar results were obtained in INS-1E cells, indicating enhanced capacity of the glycerolipid/NEFA cycle under glucotoxic conditions. Exposure to unsaturated C18:1 fatty acid favoured intracellular lipid accumulation in a glucose-dependent way, an effect also observed with saturated C16:0 fatty acid when combined with the panlipase inhibitor Orlistat. After the glucolipotoxic culture, intracellular fat mobilisation was required for acute glucose-stimulated secretion, particularly in oleate-treated cells under glucotoxic culture conditions. The lipid mobilisation rate was governed chiefly by the levels of stored fat as a direct consequence of the culture conditions rather than energetic demands, except in palmitate-loaded cells. CONCLUSIONS/INTERPRETATION Glucolipotoxic conditions promote the capacity of the glycerolipid/NEFA cycle thereby preserving part of the secretory response. The cycle of fat storage/mobilisation emerges as a mechanism helping the beta cell to cope with glucotoxic conditions.
Collapse
Affiliation(s)
- Lucie Oberhauser
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Cecilia Jiménez-Sánchez
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Jesper Grud Skat Madsen
- Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Dominique Duhamel
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Susanne Mandrup
- Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Thierry Brun
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland.
- Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland.
| |
Collapse
|
8
|
Bian Y, Hou W, Chen X, Fang J, Xu N, Ruan BH. Glutamate Dehydrogenase as a Promising Target for Hyperinsulinism Hyperammonemia Syndrome Therapy. Curr Med Chem 2021; 29:2652-2672. [PMID: 34525914 DOI: 10.2174/0929867328666210825105342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 11/22/2022]
Abstract
Hyperinsulinism-hyperammonemia syndrome (HHS) is a rare disease characterized by recurrent hypoglycemia and persistent elevation of plasma ammonia, and it can lead to severe epilepsy and permanent brain damage. It has been demonstrated that functional mutations of glutamate dehydrogenase (GDH), an enzyme in the mitochondrial matrix, are responsible for the HHS. Thus, GDH has become a promising target for the small molecule therapeutic intervention of HHS. Several medicinal chemistry studies are currently aimed at GDH, however, to date, none of the compounds reported has been entered clinical trials. This perspective summarizes the progress in the discovery and development of GDH inhibitors, including the pathogenesis of HHS, potential binding sites, screening methods, and research models. Future therapeutic perspectives are offered to provide a reference for discovering potent GDH modulators and encourage additional research that will provide more comprehensive guidance for drug development.
Collapse
Affiliation(s)
- Yunfei Bian
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hantgzhou 310014. China
| | - Wei Hou
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hantgzhou 310014. China
| | - Xinrou Chen
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hantgzhou 310014. China
| | - Jinzhang Fang
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hantgzhou 310014. China
| | - Ning Xu
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hantgzhou 310014. China
| | - Benfang Helen Ruan
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hantgzhou 310014. China
| |
Collapse
|
9
|
Li L, Krznar P, Erban A, Agazzi A, Martin-Levilain J, Supale S, Kopka J, Zamboni N, Maechler P. Metabolomics Identifies a Biomarker Revealing In Vivo Loss of Functional β-Cell Mass Before Diabetes Onset. Diabetes 2019; 68:2272-2286. [PMID: 31537525 DOI: 10.2337/db19-0131] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 09/10/2019] [Indexed: 11/13/2022]
Abstract
Identification of individuals with decreased functional β-cell mass is essential for the prevention of diabetes. However, in vivo detection of early asymptomatic β-cell defect remains unsuccessful. Metabolomics has emerged as a powerful tool in providing readouts of early disease states before clinical manifestation. We aimed at identifying novel plasma biomarkers for loss of functional β-cell mass in the asymptomatic prediabetes stage. Nontargeted and targeted metabolomics were applied in both lean β-Phb2-/- (β-cell-specific prohibitin-2 knockout) mice and obese db/db (leptin receptor mutant) mice, two distinct mouse models requiring neither chemical nor dietary treatments to induce spontaneous decline of functional β-cell mass promoting progressive diabetes development. Nontargeted metabolomics on β-Phb2-/- mice identified 48 and 82 significantly affected metabolites in liver and plasma, respectively. Machine learning analysis pointed to deoxyhexose sugars consistently reduced at the asymptomatic prediabetes stage, including in db/db mice, showing strong correlation with the gradual loss of β-cells. Further targeted metabolomics by gas chromatography-mass spectrometry uncovered the identity of the deoxyhexose, with 1,5-anhydroglucitol displaying the most substantial changes. In conclusion, this study identified 1,5-anhydroglucitol as associated with the loss of functional β-cell mass and uncovered metabolic similarities between liver and plasma, providing insights into the systemic effects caused by early decline in β-cells.
Collapse
Affiliation(s)
- Lingzi Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| | - Petra Krznar
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- PhD Program in Systems Biology, Life Science Zurich Graduate School, Zurich, Switzerland
| | - Alexander Erban
- Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
| | - Andrea Agazzi
- Theoretical Physics Department, University of Geneva, Geneva, Switzerland
| | - Juliette Martin-Levilain
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| | - Sachin Supale
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| | - Joachim Kopka
- Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| |
Collapse
|
10
|
TAKAHASHI H, YOKOI N, SEINO S. Glutamate as intracellular and extracellular signals in pancreatic islet functions. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2019; 95:246-260. [PMID: 31189778 PMCID: PMC6751295 DOI: 10.2183/pjab.95.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/08/2019] [Indexed: 05/25/2023]
Abstract
l-Glutamate is one of the most abundant amino acids in the body and is a constituent of proteins and a substrate in metabolism. It is well known that glutamate serves as a primary excitatory neurotransmitter and a critical neuromodulator in the brain. Recent studies have shown that in addition to its pivotal role in neural functions, glutamate plays many important roles in a variety of cellular functions, including those as intracellular and extracellular signals. In pancreatic islets, glutamate is now known to be required for the normal regulation of insulin secretion, such as incretin-induced insulin secretion. In this review, we primarily discuss the physiological and pathophysiological roles of glutamate as intracellular and extracellular signals in the functions of pancreatic islets.
Collapse
Affiliation(s)
- Harumi TAKAHASHI
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Norihide YOKOI
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Susumu SEINO
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| |
Collapse
|
11
|
Karaca M, Martin-Levilain J, Grimaldi M, Li L, Dizin E, Emre Y, Maechler P. Liver Glutamate Dehydrogenase Controls Whole-Body Energy Partitioning Through Amino Acid-Derived Gluconeogenesis and Ammonia Homeostasis. Diabetes 2018; 67:1949-1961. [PMID: 30002133 DOI: 10.2337/db17-1561] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/01/2018] [Indexed: 11/13/2022]
Abstract
Ammonia detoxification and gluconeogenesis are major hepatic functions mutually connected through amino acid metabolism. The liver is rich in glutamate dehydrogenase (GDH) that catalyzes the reversible oxidative deamination of glutamate to α-ketoglutarate and ammonia, thus bridging amino acid-to-glucose pathways. Here we generated inducible liver-specific GDH-knockout mice (HepGlud1-/- ) to explore the role of hepatic GDH on metabolic homeostasis. Investigation of nitrogen metabolism revealed altered ammonia homeostasis in HepGlud1-/- mice characterized by increased circulating ammonia associated with reduced detoxification process into urea. The abrogation of hepatic GDH also modified energy homeostasis. In the fasting state, HepGlud1-/- mice could barely produce glucose in response to alanine due to impaired liver gluconeogenesis. Compared with control mice, lipid consumption in HepGlud1-/- mice was favored over carbohydrates as a compensatory energy fuel. The changes in energy partitioning induced by the lack of liver GDH modified the circadian rhythm of food intake. Overall, this study demonstrates the central role of hepatic GDH as a major regulator for the maintenance of ammonia and whole-body energy homeostasis.
Collapse
Affiliation(s)
- Melis Karaca
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical School, Geneva, Switzerland
| | - Juliette Martin-Levilain
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical School, Geneva, Switzerland
| | - Mariagrazia Grimaldi
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical School, Geneva, Switzerland
| | - Lingzi Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical School, Geneva, Switzerland
| | - Eva Dizin
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
| | - Yalin Emre
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Geneva, Switzerland
- Faculty Diabetes Center, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
12
|
Gaspar C, Silva-Marrero JI, Fàbregas A, Miñarro M, Ticó JR, Baanante IV, Metón I. Administration of chitosan-tripolyphosphate-DNA nanoparticles to knockdown glutamate dehydrogenase expression impairs transdeamination and gluconeogenesis in the liver. J Biotechnol 2018; 286:5-13. [PMID: 30195924 DOI: 10.1016/j.jbiotec.2018.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/02/2018] [Accepted: 09/04/2018] [Indexed: 11/17/2022]
Abstract
Glutamate dehydrogenase (GDH) plays a major role in amino acid catabolism. To increase the current knowledge of GDH function, we analysed the effect of GDH silencing on liver intermediary metabolism from gilthead sea bream (Sparus aurata). Sequencing of GDH cDNA from S. aurata revealed high homology with its vertebrate orthologues and allowed us to design short hairpin RNAs (shRNAs) to knockdown GDH expression. Following validation of shRNA-dependent downregulation of S. aurata GDH in vitro, chitosan-tripolyphosphate (TPP) nanoparticles complexed with a plasmid encoding a selected shRNA (pCpG-sh2GDH) were produced to address the effect of GDH silencing on S. aurata liver metabolism. Seventy-two hours following intraperitoneal administration of chitosan-TPP-pCpG-sh2GDH, GDH mRNA levels and immunodetectable protein decreased in the liver, leading to reduced GDH activity in both oxidative and reductive reactions to about 53-55 % of control values. GDH silencing decreased glutamate, glutamine and aspartate aminotransferase activity, while increased 2-oxoglutarate content, 2-oxoglutarate dehydrogenase activity and 6-phosphofructo-1-kinase/fructose-1,6-bisphosphatase activity ratio. Our findings show for the first time that GDH silencing reduces transdeamination and gluconeogenesis in the liver, hindering the use of amino acids as gluconeogenic substrates and enabling protein sparing and metabolisation of dietary carbohydrates, which would reduce environmental impact and production costs of aquaculture.
Collapse
Affiliation(s)
- Carlos Gaspar
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Jonás I Silva-Marrero
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Anna Fàbregas
- Departament de Farmàcia i Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Montserrat Miñarro
- Departament de Farmàcia i Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Josep R Ticó
- Departament de Farmàcia i Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Isabel V Baanante
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Isidoro Metón
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
13
|
Mathioudakis L, Bourbouli M, Daklada E, Kargatzi S, Michaelidou K, Zaganas I. Localization of Human Glutamate Dehydrogenases Provides Insights into Their Metabolic Role and Their Involvement in Disease Processes. Neurochem Res 2018; 44:170-187. [PMID: 29943084 DOI: 10.1007/s11064-018-2575-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 12/21/2022]
Abstract
Glutamate dehydrogenase (GDH) catalyzes the reversible deamination of L-glutamate to α-ketoglutarate and ammonia. In mammals, GDH contributes to important processes such as amino acid and carbohydrate metabolism, energy production, ammonia management, neurotransmitter recycling and insulin secretion. In humans, two isoforms of GDH are found, namely hGDH1 and hGDH2, with the former being ubiquitously expressed and the latter found mainly in brain, testis and kidney. These two iso-enzymes display highly divergent allosteric properties, especially concerning their basal activity, ADP activation and GTP inhibition. On the other hand, both enzymes are thought to predominantly localize in the mitochondrial matrix, even though alternative localizations have been proposed. To further study the subcellular localization of the two human iso-enzymes, we created HEK293 cell lines stably over-expressing hGDH1 and hGDH2. In these cell lines, immunofluorescence and enzymatic analyses verified the overexpression of both hGDH1 and hGDH2 iso-enzymes, whereas subcellular fractionation followed by immunoblotting showed their predominantly mitochondrial localization. Given that previous studies have only indirectly compared the subcellular localization of the two iso-enzymes, we co-expressed them tagged with different fluorescent dyes (green and red fluorescent protein for hGDH1 and hGDH2, respectively) and found them to co-localize. Despite the wealth of information related to the functional properties of hGDH1 and hGDH2 and the availability of the hGDH1 structure, there is still an ongoing debate concerning their metabolic role and their involvement in disease processes. Data on the localization of hGDHs, as the ones presented here, could contribute to better understanding of the function of these important human enzymes.
Collapse
Affiliation(s)
- Lambros Mathioudakis
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Mara Bourbouli
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Elisavet Daklada
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Sofia Kargatzi
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Kleita Michaelidou
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Ioannis Zaganas
- Neurology Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece. .,Department of Neurology, University Hospital of Heraklion, Heraklion, Crete, Greece.
| |
Collapse
|
14
|
Bruni A, Bornstein S, Linkermann A, Shapiro AMJ. Regulated Cell Death Seen through the Lens of Islet Transplantation. Cell Transplant 2018; 27:890-901. [PMID: 29845882 PMCID: PMC6050903 DOI: 10.1177/0963689718766323] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Clinical islet transplantation effectively restores euglycemia and corrects glycosylated
hemoglobin in labile type 1 diabetes mellitus (T1DM). Despite marked improvements in islet
transplantation outcomes, acute islet cell death remains a substantial obstacle that
compromises long-term engraftment outcomes. Multiple organ donors are routinely required
to achieve insulin independence. Therapeutic agents that ameliorate cell death and/or
control injury-related inflammatory cascades offer potential to improve islet transplant
success. Apoptotic cell death has been identified as a major contributor to cellular
demise and therapeutic strategies that subvert initiation and consequences of apoptotic
cell death have shown promise in pre-clinical models. Indeed, in numerous pathologies and
diseases apoptosis has been the most extensively described form of regulated cell death.
However, recent identification of novel, alternative regulated cell death pathways in
other disease states and solid organ transplantation suggest that these additional
pathways may also have substantial relevance in islet transplantation. These regulated,
non-apoptotic cell death pathways exhibit distinct biochemical characteristics but have
yet to be fully characterized within islet transplantation. We review herein the various
regulated cell death pathways and highlight their relative potential contributions to
islet viability, engraftment failure and islet dysfunction.
Collapse
Affiliation(s)
- Antonio Bruni
- 1 Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,2 Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Stefan Bornstein
- 3 Division of Nephrology, Medical Clinic 3, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Andreas Linkermann
- 3 Division of Nephrology, Medical Clinic 3, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - A M James Shapiro
- 1 Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,2 Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Bruni A, Pepper AR, Pawlick RL, Gala-Lopez B, Gamble AF, Kin T, Seeberger K, Korbutt GS, Bornstein SR, Linkermann A, Shapiro AMJ. Ferroptosis-inducing agents compromise in vitro human islet viability and function. Cell Death Dis 2018; 9:595. [PMID: 29789532 PMCID: PMC5964226 DOI: 10.1038/s41419-018-0506-0] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 02/07/2023]
Abstract
Human islet transplantation has been hampered by donor cell death associated with the islet preparation procedure before transplantation. Regulated necrosis pathways are biochemically and morphologically distinct from apoptosis. Recently, ferroptosis was identified as a non-apoptotic form of iron-dependent regulated necrosis implicated in various pathological conditions. Mediators of islet oxidative stress, including glutathione peroxidase-4 (GPX4), have been identified as inhibitors of ferroptosis, and mechanisms that affect GPX4 function can impact islet function and viability. Ferroptosis has not been investigated directly in human islets, and its relevance in islet transplantation remains unknown. Herein, we sought to determine whether in vitro human islet viability and function is compromised in the presence of two distinct ferroptosis-inducing agents (FIA), erastin or RSL3, and whether these effects could be rescued with ferroptosis inhibitors, ferrostatin-1 (Fer-1), or desferrioxamine (DFO). Viability, as assessed by lactate dehydrogenase (LDH) release, revealed significant death in erastin- and RSL3-treated islets, 20.3% ± 3.8 and 24.4% ± 2.5, 24 h post culture, respectively. These effects were ameliorated in islets pre-treated with Fer-1 or the iron chelator, desferrioxamine (DFO). Stimulation index, a marker of islet function revealed a significant reduction in function in erastin-treated islets (control 1.97 ± 0.13 vs. 50 μM erastin 1.32 ± 0.1) (p < 0.05). Fer-1 and DFO pre-treatment alone did not augment islet viability or function. Pre-treatment of islets with erastin or Fer-1 did not impact in vivo engraftment in an immunodeficient mouse transplant model. Our data reveal that islets are indeed susceptible to ferroptosis in vitro, and induction of this novel cell death modality leads to compromised islet function, which can be recoverable in the presence of the ferroptosis inhibitors. The in vivo impact of this pathway in islet transplantation remains elusive given the constraints of our study, but warrants continued investigation.
Collapse
Affiliation(s)
- Antonio Bruni
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Andrew R Pepper
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Rena L Pawlick
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Boris Gala-Lopez
- QEII Health Science Centre, Dalhousie University, Halifax, NS, Canada
| | - Anissa F Gamble
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Tatsuya Kin
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Karen Seeberger
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | | | - Stefan R Bornstein
- Clinic for Internal Medicine 3, Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Andreas Linkermann
- Division of Nephrology, Clinic for Internal Medicine 3, Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - A M James Shapiro
- Clinical Islet Transplant Program, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
16
|
Gaspar C, Silva-Marrero JI, Salgado MC, Baanante IV, Metón I. Role of upstream stimulatory factor 2 in glutamate dehydrogenase gene transcription. J Mol Endocrinol 2018; 60:247-259. [PMID: 29438976 DOI: 10.1530/jme-17-0142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 02/08/2018] [Indexed: 12/18/2022]
Abstract
Glutamate dehydrogenase (Gdh) plays a central role in ammonia detoxification by catalysing reversible oxidative deamination of l-glutamate into α-ketoglutarate using NAD+ or NADP+ as cofactor. To gain insight into transcriptional regulation of glud, the gene that codes for Gdh, we isolated and characterised the 5' flanking region of glud from gilthead sea bream (Sparus aurata). In addition, tissue distribution, the effect of starvation as well as short- and long-term refeeding on Gdh mRNA levels in the liver of S. aurata were also addressed. 5'-Deletion analysis of glud promoter in transiently transfected HepG2 cells, electrophoretic mobility shift assays, chromatin immunoprecipitation (ChIP) and site-directed mutagenesis allowed us to identify upstream stimulatory factor 2 (Usf2) as a novel factor involved in the transcriptional regulation of glud Analysis of tissue distribution of Gdh and Usf2 mRNA levels by reverse transcriptase-coupled quantitative real-time PCR (RT-qPCR) showed that Gdh is mainly expressed in the liver of S. aurata, while Usf2 displayed ubiquitous distribution. RT-qPCR and ChIP assays revealed that long-term starvation down-regulated the hepatic expression of Gdh and Usf2 to similar levels and reduced Usf2 binding to glud promoter, while refeeding resulted in a slow but gradual restoration of both Gdh and Usf2 mRNA abundance. Herein, we demonstrate that Usf2 transactivates S. aurata glud by binding to an E-box located in the proximal region of glud promoter. In addition, our findings provide evidence for a new regulatory mechanism involving Usf2 as a key factor in the nutritional regulation of glud transcription in the fish liver.
Collapse
Affiliation(s)
- Carlos Gaspar
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Jonás I Silva-Marrero
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - María C Salgado
- Servei de Bioquímica Clínica, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Isabel V Baanante
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Isidoro Metón
- Secció de Bioquímica i Biologia Molecular, Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Andersson LE, Shcherbina L, Al-Majdoub M, Vishnu N, Arroyo CB, Aste Carrara J, Wollheim CB, Fex M, Mulder H, Wierup N, Spégel P. Glutamine-Elicited Secretion of Glucagon-Like Peptide 1 Is Governed by an Activated Glutamate Dehydrogenase. Diabetes 2018; 67:372-384. [PMID: 29229616 DOI: 10.2337/db16-1441] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/07/2017] [Indexed: 11/13/2022]
Abstract
Glucagon-like peptide 1 (GLP-1), secreted from intestinal L cells, glucose dependently stimulates insulin secretion from β-cells. This glucose dependence prevents hypoglycemia, rendering GLP-1 analogs a useful and safe treatment modality in type 2 diabetes. Although the amino acid glutamine is a potent elicitor of GLP-1 secretion, the responsible mechanism remains unclear. We investigated how GLP-1 secretion is metabolically coupled in L cells (GLUTag) and in vivo in mice using the insulin-secreting cell line INS-1 832/13 as reference. A membrane-permeable glutamate analog (dimethylglutamate [DMG]), acting downstream of electrogenic transporters, elicited similar alterations in metabolism as glutamine in both cell lines. Both DMG and glutamine alone elicited GLP-1 secretion in GLUTag cells and in vivo, whereas activation of glutamate dehydrogenase (GDH) was required to stimulate insulin secretion from INS-1 832/13 cells. Pharmacological inhibition in vivo of GDH blocked secretion of GLP-1 in response to DMG. In conclusion, our results suggest that nonelectrogenic nutrient uptake and metabolism play an important role in L cell stimulus-secretion coupling. Metabolism of glutamine and related analogs by GDH in the L cell may explain why GLP-1 secretion, but not that of insulin, is activated by these secretagogues in vivo.
Collapse
Affiliation(s)
- Lotta E Andersson
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Liliya Shcherbina
- Neuroendocrine Cell Biology, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Mahmoud Al-Majdoub
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Neelanjan Vishnu
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | | | - Jonathan Aste Carrara
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Claes B Wollheim
- Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
- Department of Cell Physiology and Metabolism, University Medical Centre, Geneva, Switzerland
| | - Malin Fex
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Hindrik Mulder
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Nils Wierup
- Neuroendocrine Cell Biology, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Peter Spégel
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| |
Collapse
|
18
|
Grimaldi M, Karaca M, Latini L, Brioudes E, Schalch T, Maechler P. Identification of the molecular dysfunction caused by glutamate dehydrogenase S445L mutation responsible for hyperinsulinism/hyperammonemia. Hum Mol Genet 2018; 26:3453-3465. [PMID: 28911206 DOI: 10.1093/hmg/ddx213] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/01/2017] [Indexed: 01/14/2023] Open
Abstract
Congenital hyperinsulinism/hyperammonemia (HI/HA) syndrome gives rise to unregulated protein-induced insulin secretion from pancreatic beta-cells, fasting hypoglycemia and elevated plasma ammonia levels. Mutations associated with HI/HA were identified in the Glud1 gene, encoding for glutamate dehydrogenase (GDH). We aimed at identifying the molecular causes of dysregulation in insulin secretion and ammonia production conferred by the most frequent HI/HA mutation Ser445Leu. Following transduction with adenoviruses carrying the human GDH-wild type or GDH-S445L-mutant gene, immunoblotting showed efficient expression of the transgenes in all the investigated cell types. Enzymatic activity tested in INS-1E beta-cells revealed that the mutant was much more sensitive to the allosteric activator ADP, rendering it highly responsive to substrates. INS-1E cells expressing either the wild type or mutant GDH responded similarly to glucose stimulation regarding mitochondrial activation and insulin secretion. However, at basal glucose glutamine stimulation increased mitochondrial activity and insulin release only in the mutant cells. In mouse and human islets, expression of mutant GDH resulted in robust elevation of insulin secretion upon glutamine stimulation, not observed in control islets. Hepatocytes expressing either the wild type or mutant GDH produced similar levels of ammonia when exposed to glutamine, although alanine response was strongly elevated with the mutant form. In conclusion, the GDH-S445L mutation confers hyperactivity to this enzyme due to higher sensitivity to ADP allosteric activation. This renders beta-cells responsive to amino acid stimulation, explaining protein-induced hypoglycemia secondary to non-physiological insulin release. Hepatocytes carrying mutant GDH produced more ammonia upon alanine exposure, which underscores hyperammonemia developed by the patients.
Collapse
Affiliation(s)
- Mariagrazia Grimaldi
- Department of Cell Physiology and Metabolism.,Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland
| | - Melis Karaca
- Department of Cell Physiology and Metabolism.,Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland
| | - Livia Latini
- Department of Cell Physiology and Metabolism.,Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland
| | - Estelle Brioudes
- Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland.,Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, Geneva, Switzerland
| | - Thomas Schalch
- Department of Molecular Biology, Faculty of Science, Institute of Genetics and Genomics of Geneva (iGE3), Sciences III, University of Geneva, 1211 Geneva 4, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism.,Faculty Diabetes Center, University of Geneva Medical Center, 1206 Geneva, Switzerland
| |
Collapse
|
19
|
Brun J, Berthou F, Trajkovski M, Maechler P, Foti M, Bonnet N. Bone Regulates Browning and Energy Metabolism Through Mature Osteoblast/Osteocyte PPARγ Expression. Diabetes 2017; 66:2541-2554. [PMID: 28687706 DOI: 10.2337/db17-0116] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/29/2017] [Indexed: 11/13/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a master regulator of energy metabolism. In bone, it is known to regulate osteoblast differentiation and osteoclast activity. Whether PPARγ expression in bone cells, particularly osteocytes, regulates energy metabolism remains unknown. Here, we show that mature osteoblast/osteocyte-specific ablation of PPARγ in mice (Ocy-PPARγ-/-) alters body composition with age, namely, to produce less fat and more lean mass, and enhances insulin sensitivity and energy expenditure compared with wild-type mice. In addition, Ocy-PPARγ-/- mice exhibit more bone density, structure, and strength by uncoupling bone formation from resorption. When challenged with a high-fat diet, Ocy-PPARγ-/- mice retain glycemic control, with increased browning of the adipose tissue, decreased gluconeogenesis, and less hepatic steatosis. Moreover, these metabolic effects, particularly an increase in fatty acid oxidation, cannot be explained by decarboxylated osteocalcin changes, suggesting existence of other osteokines that are under the control of PPARγ. We further identify bone morphogenetic protein 7 as one of them. Hence, osteocytes coregulate bone and glucose homeostasis through a PPARγ regulatory pathway, and its inhibition could be clinically relevant for the prevention of glucose metabolic disorders.
Collapse
Affiliation(s)
- Julia Brun
- Division of Bone Diseases, Department of Internal Medicine Specialties, Faculty of Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Flavien Berthou
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Mirko Trajkovski
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Michanlegelo Foti
- Department of Cell Physiology and Metabolism, University of Geneva, and Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Faculty of Medicine, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
20
|
Niaz K, Hassan FI, Mabqool F, Khan F, Momtaz S, Baeeri M, Navaei-Nigjeh M, Rahimifard M, Abdollahi M. Effect of styrene exposure on plasma parameters, molecular mechanisms and gene expression in rat model islet cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 54:62-73. [PMID: 28688303 DOI: 10.1016/j.etap.2017.06.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 06/07/2023]
Abstract
Styrene is an aromatic hydrocarbon compound present in the environment and have primary exposure through plastic industry. The current study was designed to evaluate styrene-induced toxicity parameters in rat plasma fasting blood glucose (FBG) level, oral glucose tolerance, insulin secretion, oxidative stress, and inflammatory cytokines in cellular and molecular levels. Styrene was dissolved in corn oil and administered at different doses (250, 500, 1000, 1500, 2000mg/kg/day and control) to each rat, for 42days. In treated groups, styrene significantly increased fasting blood glucose, plasma insulin (p<0.001) and glucose tolerance. Glucose tolerance, insulin resistance and hyperglycemia were found to be the main consequences correlating gene expression of islet cells. Styrene caused a significant enhancement of oxidative stress markers (p<0.001) and inflammatory cytokines in a dose and concentration-dependent manner in plasma (p<0.001). Moreover, the activities of caspase-3 and -9 of the islet cells were significantly up-regulated by this compound at 1500 and 2000mg/kg/day styrene administrated groups (p<0.001). The relative fold change of GLUD1 was downregulated (p<0.05) and upregulated at 1500 and 2000mg/kg, respectively (p<0.01). The relative fold changes of GLUT2 were down regulated at 250 and 1000mg/kg and up regulated in 500, 1500 and 2000mg/kg doses of styrene (p<0.01). The expression level of GCK indicated a significant upregulation at 250mg/kg and downregulation of relative fold changes in the remaining doses of styrene, except for no change at 2000mg/kg of styrene for GCK. Targeting genes (GLUD1, GLUT2 and GCK) of the pancreatic islet cells in styrene exposed groups, disrupted gluconeogenesis, glycogenolysis pathways and insulin secretory functions. The present study illustrated that fasting blood glucose, insulin pathway, oxidative balance, inflammatory cytokines, cell viability and responsible genes of glucose metabolism are susceptible to styrene, which consequently lead to other abnormalities in various organs.
Collapse
Affiliation(s)
- Kamal Niaz
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatima Ismail Hassan
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Faheem Mabqool
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fazlullah Khan
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Baeeri
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Navaei-Nigjeh
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahban Rahimifard
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Maechler P. Glutamate pathways of the beta-cell and the control of insulin secretion. Diabetes Res Clin Pract 2017; 131:149-153. [PMID: 28743063 DOI: 10.1016/j.diabres.2017.07.009] [Citation(s) in RCA: 23] [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: 06/20/2017] [Accepted: 07/04/2017] [Indexed: 11/29/2022]
Abstract
Pancreatic beta-cells secrete insulin in response to circulating glucose, thereby maintaining euglycemia. Inside the beta-cell, glucose is transformed into intracellular signals stimulating exocytosis. While calcium is an obligatory messenger, this ion is not sufficient to promote the full secretory response. Accordingly, glucose metabolism produces the additive factor glutamate that participates to an amplifying pathway of the calcium signal. Although intracellular glutamate potentiates insulin secretion, extracellular glutamate may activate ionotropic receptors. As a consequence of such activation, insulin exocytosis is slowed down. Therefore, for the beta-cell glutamate is a double-edged sword, an amplifying pathway and a negative feedback, illustrating the principle of homeostasis.
Collapse
Affiliation(s)
- Pierre Maechler
- Department of Cell Physiology and Metabolism & Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland.
| |
Collapse
|
22
|
Upregulation of UCP2 in beta-cells confers partial protection against both oxidative stress and glucotoxicity. Redox Biol 2017; 13:541-549. [PMID: 28755631 PMCID: PMC5537434 DOI: 10.1016/j.redox.2017.07.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 01/09/2023] Open
Abstract
Deterioration of pancreatic beta-cells plays a critical role in the development of type 2 diabetes. Among the various stressors contributing to these deleterious effects, glucotoxicity and superoxides have been proposed as major players. In this context, the mitochondrial uncoupling protein UCP2 is regularly associated with the stress response. In the present study, we tested the effects of UCP2 upregulation in mouse islets with beta-cell specific overexpression of UCP2 (RIP-UCP2). Islets were subjected to both chronic glucotoxicity (7 days at 30 mM glucose) and acute oxidative stress (200 µM H2O2 for 10 min). Increased UCP2 expression did not alter mitochondrial potential and ATP generation but protected against glucotoxic effects. Glucose-stimulated insulin secretion was altered by both glucotoxicity and oxidative stress, in particular through higher basal insulin release at non-stimulatory glucose concentrations. The secretory response to glucose stimulation was partially preserved in beta-cells overexpressing UCP2. The higher rate of cell death induced by chronic high glucose exposure was lower in RIP-UCP2 islets. Finally, superoxide production was reduced by high glucose, both under acute and chronic conditions, and not modified by UCP2 overexpression. In conclusion, upregulation of UCP2 conferred protective effects to the stressed beta-cell through mechanisms not directly associated with superoxide production. UCP2 upregulation protects pancreatic ß-cells against glucotoxicity. High glucose reduces superoxide production in pancreatic islets. UCP2 upregulation does not change superoxide production. UCP2 upregulation protects ß-cells against oxidative stress.
Collapse
|
23
|
Mulder H. Transcribing β-cell mitochondria in health and disease. Mol Metab 2017; 6:1040-1051. [PMID: 28951827 PMCID: PMC5605719 DOI: 10.1016/j.molmet.2017.05.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/13/2017] [Accepted: 05/22/2017] [Indexed: 12/17/2022] Open
Abstract
Background The recent genome-wide association studies (GWAS) of Type 2 Diabetes (T2D) have identified the pancreatic β-cell as the culprit in the pathogenesis of the disease. Mitochondrial metabolism plays a crucial role in the processes controlling release of insulin and β-cell mass. This notion implies that mechanisms controlling mitochondrial function have the potential to play a decisive pathogenetic role in T2D. Scope of the review This article reviews studies demonstrating that there is indeed mitochondrial dysfunction in islets in T2D, and that GWAS have identified a variant in the gene encoding transcription factor B1 mitochondrial (TFB1M), predisposing to T2D due to mitochondrial dysfunction and impaired insulin secretion. Mechanistic studies of the nature of this pathogenetic link, as well as of other mitochondrial transcription factors, are described. Major conclusions Based on this, it is argued that transcription and translation in mitochondria are critical processes determining mitochondrial function in β-cells in health and disease.
Collapse
Key Words
- AMPK, AMP-dependent protein kinase
- ATGL, adipocyte triglyceride lipase
- COX, Cytochrome c oxidase
- CYTB, Cytochrome b
- ERR-α, Estrogen-related receptor-α
- Expression quantitative trait locus (eQTL)
- GDH, Glutamate dehydrogenase
- GSIS, Glucose-stimulated insulin secretion
- GWAS, Genome-wide association study
- Genome-wide association study (GWAS)
- HSL, Hormone-sensitive lipase
- ICDc, Cytosolic isocitrate dehydrogenase
- Insulin secretion
- Islets
- KATP, ATP-dependent K+-channel
- MTERF, Mitochondrial transcription termination factor
- Mitochondria
- ND, NADH dehydrogenase
- NRF, Nuclear respiratory factor
- NSUN4, NOP2/Sun RNA methyltransferase family member 4
- OXPHOS, Oxidative phosphorylation
- PC, Pyruvate carboxylase
- PDH, pyruvate dehydrogenase
- PGC, Peroxisome proliferator-activated receptor-γ co-activator
- POLRMT, Mitochondrial RNA polymerase
- POLγ, DNA polymerase-γ
- PPARγ, Peroxisome proliferator-activated receptor-γ
- PRC, PGC1-related coactivator
- SENP1, Sentrin/SUMO-specific protease-1
- SNP, Single Nucleotide Polymorphism
- SUR1, Sulphonylurea receptor-1
- T2D, Type 2 Diabetes
- TCA, Tricarboxylic acid
- TEFM, Mitochondrial transcription elongation factor
- TFAM, Transcription factor A mitochondrial
- TFB1M, Transcription factor B1 mitochondrial
- TFB2M, Transcription factor B2 mitochondrial
- eQTL, Expression quantitative trait locus
- β-Cell
Collapse
Affiliation(s)
- Hindrik Mulder
- Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmö, Sweden
| |
Collapse
|
24
|
The Glutamate Dehydrogenase Pathway and Its Roles in Cell and Tissue Biology in Health and Disease. BIOLOGY 2017; 6:biology6010011. [PMID: 28208702 PMCID: PMC5372004 DOI: 10.3390/biology6010011] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 02/07/2023]
Abstract
Glutamate dehydrogenase (GDH) is a hexameric enzyme that catalyzes the reversible conversion of glutamate to α-ketoglutarate and ammonia while reducing NAD(P)⁺ to NAD(P)H. It is found in all living organisms serving both catabolic and anabolic reactions. In mammalian tissues, oxidative deamination of glutamate via GDH generates α-ketoglutarate, which is metabolized by the Krebs cycle, leading to the synthesis of ATP. In addition, the GDH pathway is linked to diverse cellular processes, including ammonia metabolism, acid-base equilibrium, redox homeostasis (via formation of fumarate), lipid biosynthesis (via oxidative generation of citrate), and lactate production. While most mammals possess a single GDH1 protein (hGDH1 in the human) that is highly expressed in the liver, humans and other primates have acquired, via duplication, an hGDH2 isoenzyme with distinct functional properties and tissue expression profile. The novel hGDH2 underwent rapid evolutionary adaptation, acquiring unique properties that enable enhanced enzyme function under conditions inhibitory to its ancestor hGDH1. These are thought to provide a biological advantage to humans with hGDH2 evolution occurring concomitantly with human brain development. hGDH2 is co-expressed with hGDH1 in human brain, kidney, testis and steroidogenic organs, but not in the liver. In human cerebral cortex, hGDH1 and hGDH2 are expressed in astrocytes, the cells responsible for removing and metabolizing transmitter glutamate, and for supplying neurons with glutamine and lactate. In human testis, hGDH2 (but not hGDH1) is densely expressed in the Sertoli cells, known to provide the spermatids with lactate and other nutrients. In steroid producing cells, hGDH1/2 is thought to generate reducing equivalents (NADPH) in the mitochondria for the biosynthesis of steroidal hormones. Lastly, up-regulation of hGDH1/2 expression occurs in cancer, permitting neoplastic cells to utilize glutamine/glutamate for their growth. In addition, deregulation of hGDH1/2 is implicated in the pathogenesis of several human disorders.
Collapse
|
25
|
Pournourmohammadi S, Grimaldi M, Stridh MH, Lavallard V, Waagepetersen HS, Wollheim CB, Maechler P. Epigallocatechin-3-gallate (EGCG) activates AMPK through the inhibition of glutamate dehydrogenase in muscle and pancreatic ß-cells: A potential beneficial effect in the pre-diabetic state? Int J Biochem Cell Biol 2017; 88:220-225. [PMID: 28137482 DOI: 10.1016/j.biocel.2017.01.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 01/19/2017] [Accepted: 01/25/2017] [Indexed: 12/27/2022]
Abstract
Glucose homeostasis is determined by insulin secretion from the ß-cells in pancreatic islets and by glucose uptake in skeletal muscle and other insulin target tissues. While glutamate dehydrogenase (GDH) senses mitochondrial energy supply and regulates insulin secretion, its role in the muscle has not been elucidated. Here we investigated the possible interplay between GDH and the cytosolic energy sensing enzyme 5'-AMP kinase (AMPK), in both isolated islets and myotubes from mice and humans. The green tea polyphenol epigallocatechin-3-gallate (EGCG) was used to inhibit GDH. Insulin secretion was reduced by EGCG upon glucose stimulation and blocked in response to glutamine combined with the allosteric GDH activator BCH (2-aminobicyclo-[2,2,1] heptane-2-carboxylic acid). Insulin secretion was similarly decreased in islets of mice with ß-cell-targeted deletion of GDH (ßGlud1-/-). EGCG did not further reduce insulin secretion in the mutant islets, validating its specificity. In human islets, EGCG attenuated both basal and nutrient-stimulated insulin secretion. Glutamine/BCH-induced lowering of AMPK phosphorylation did not operate in ßGlud1-/- islets and was similarly prevented by EGCG in control islets, while high glucose systematically inactivated AMPK. In mouse C2C12 myotubes, like in islets, the inhibition of AMPK following GDH activation with glutamine/BCH was reversed by EGCG. Stimulation of GDH in primary human myotubes caused lowering of insulin-induced 2-deoxy-glucose uptake, partially counteracted by EGCG. Thus, mitochondrial energy provision through anaplerotic input via GDH influences the activity of the cytosolic energy sensor AMPK. EGCG may be useful in obesity by resensitizing insulin-resistant muscle while blunting hypersecretion of insulin in hypermetabolic states.
Collapse
Affiliation(s)
- Shirin Pournourmohammadi
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland; Faculty Diabetes Center, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland
| | - Mariagrazia Grimaldi
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland; Faculty Diabetes Center, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland
| | - Malin H Stridh
- Department of Drug Design and Pharmacotherapy, Faculty of Health and Medical Sciences, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| | - Vanessa Lavallard
- Faculty Diabetes Center, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland; Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospital, Geneva, Switzerland
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacotherapy, Faculty of Health and Medical Sciences, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| | - Claes B Wollheim
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland; Faculty Diabetes Center, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland; Faculty Diabetes Center, University of Geneva Medical Center, 1 rue Michel-Servet, 1206 Geneva, Switzerland.
| |
Collapse
|
26
|
Protein malnutrition potentiates the amplifying pathway of insulin secretion in adult obese mice. Sci Rep 2016; 6:33464. [PMID: 27633083 PMCID: PMC5025848 DOI: 10.1038/srep33464] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 08/30/2016] [Indexed: 12/28/2022] Open
Abstract
Pancreatic beta cell (β) dysfunction is an outcome of malnutrition. We assessed the role of the amplifying pathway (AMP PATH) in β cells in malnourished obese mice. C57Bl-6 mice were fed a control (C) or a low-protein diet (R). The groups were then fed a high-fat diet (CH and RH). AMP PATH contribution to insulin secretion was assessed upon incubating islets with diazoxide and KCl. CH and RH displayed increased glucose intolerance, insulin resistance and glucose-stimulated insulin secretion. Only RH showed a higher contribution of the AMP PATH. The mitochondrial membrane potential of RH was decreased, and ATP flux was unaltered. In RH islets, glutamate dehydrogenase (GDH) protein content and activity increased, and the AMP PATH contribution was reestablished when GDH was blunted. Thus, protein malnutrition induces mitochondrial dysfunction in β cells, leading to an increased contribution of the AMP PATH to insulin secretion through the enhancement of GDH content and activity.
Collapse
|
27
|
Vetterli L, Carobbio S, Frigerio F, Karaca M, Maechler P. The Amplifying Pathway of the β-Cell Contributes to Diet-induced Obesity. J Biol Chem 2016; 291:13063-75. [PMID: 27137930 DOI: 10.1074/jbc.m115.707448] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Indexed: 12/24/2022] Open
Abstract
Efficient energy storage in adipose tissues requires optimal function of the insulin-producing β-cell, whereas its dysfunction promotes diabetes. The associated paradox related to β-cell efficiency is that excessive accumulation of fat in adipose tissue predisposes for type 2 diabetes. Insulin exocytosis is regulated by intracellular metabolic signal transduction, with glutamate dehydrogenase playing a key role in the amplification of the secretory response. Here, we used mice with β-cell-selective glutamate dehydrogenase deletion (βGlud1(-/-)), lacking an amplifying pathway of insulin secretion. As opposed to control mice, βGlud1(-/-) animals fed a high calorie diet maintained glucose tolerance and did not develop diet-induced obesity. Islets of βGlud1(-/-) mice did not increase their secretory response upon high calorie feeding, as did islets of control mice. Inhibited adipose tissue expansion observed in knock-out mice correlated with lower expression of genes responsible for adipogenesis. Rather than being efficiently stored, lipids were consumed at a higher rate in βGlud1(-/-) mice compared with controls, in particular during food intake periods. These results show that reduced β-cell function prior to high calorie feeding prevented diet-induced obesity.
Collapse
Affiliation(s)
- Laurène Vetterli
- From the Department of Cell Physiology and Metabolism and Faculty Diabetes Center, Geneva University Medical Centre, 1211 Geneva 4, Switzerland
| | - Stefania Carobbio
- From the Department of Cell Physiology and Metabolism and Faculty Diabetes Center, Geneva University Medical Centre, 1211 Geneva 4, Switzerland
| | - Francesca Frigerio
- From the Department of Cell Physiology and Metabolism and Faculty Diabetes Center, Geneva University Medical Centre, 1211 Geneva 4, Switzerland
| | - Melis Karaca
- From the Department of Cell Physiology and Metabolism and Faculty Diabetes Center, Geneva University Medical Centre, 1211 Geneva 4, Switzerland
| | - Pierre Maechler
- From the Department of Cell Physiology and Metabolism and Faculty Diabetes Center, Geneva University Medical Centre, 1211 Geneva 4, Switzerland
| |
Collapse
|
28
|
Auro K, Joensuu A, Fischer K, Kettunen J, Salo P, Mattsson H, Niironen M, Kaprio J, Eriksson JG, Lehtimäki T, Raitakari O, Jula A, Tiitinen A, Jauhiainen M, Soininen P, Kangas AJ, Kähönen M, Havulinna AS, Ala-Korpela M, Salomaa V, Metspalu A, Perola M. A metabolic view on menopause and ageing. Nat Commun 2014; 5:4708. [PMID: 25144627 DOI: 10.1038/ncomms5708] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 07/16/2014] [Indexed: 12/12/2022] Open
Abstract
The ageing of the global population calls for a better understanding of age-related metabolic consequences. Here we report the effects of age, sex and menopause on serum metabolites in 26,065 individuals of Northern European ancestry. Age-specific metabolic fingerprints differ significantly by gender and, in females, a substantial atherogenic shift overlapping the time of menopausal transition is observed. In meta-analysis of 10,083 women, menopause status associates with amino acids glutamine, tyrosine and isoleucine, along with serum cholesterol measures and atherogenic lipoproteins. Among 3,204 women aged 40-55 years, menopause status associates additionally with glycine and total, monounsaturated, and omega-7 and -9 fatty acids. Our findings suggest that, in addition to lipid alterations, menopause may contribute to future metabolic and cardiovascular risk via influencing amino-acid concentrations, adding to the growing evidence of the importance of amino acids in metabolic disease progression. These observations shed light on the metabolic consequences of ageing, gender and menopause at the population level.
Collapse
Affiliation(s)
- Kirsi Auro
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3] Department of Obstetrics and Gynecology, Helsinki University Central Hospital and University of Helsinki, Haartmaninkatu 2, Helsinki 00290, Finland [4]
| | - Anni Joensuu
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3]
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Riia 23b, Tartu 51010, Estonia
| | - Johannes Kettunen
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland
| | - Perttu Salo
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Hannele Mattsson
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Marjo Niironen
- Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland
| | - Jaakko Kaprio
- 1] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [2] Departmentof Public Health, Hjelt Institute, University of Helsinki, PO Box 41 Mannerheimintie 172, Helsinki 00014, Finland [3] Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, PO Box 30 (Mannerheimintie 166), Helsinki 00300, Finland
| | - Johan G Eriksson
- 1] Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland [2] Department of General Practice and Primary Health Care, University of Helsinki, PL 20, Tukholmankatu 8B, Helsinki 00029, Finland [3] Vasa Central Hospital, Sandviksgatan 2-4, Vasa 65130, Finland [4] Folkhälsan Research Centre, Helsingfors Universitet, PB 63, Helsinki 00014, Finland [5] Unit of General Practice, Helsinki University Central Hospital, Haartmaninkatu 4, Helsinki 00290, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere University, Kalevantie 4, Tampere 33014, Finland
| | - Olli Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, Turku 20521, Finland
| | - Antti Jula
- Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland
| | - Aila Tiitinen
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital and University of Helsinki, Haartmaninkatu 2, Helsinki 00290, Finland
| | - Matti Jauhiainen
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Pasi Soininen
- 1] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland [2] NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 PL 1627, Kuopio 70211, Finland
| | - Antti J Kangas
- 1] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland [2] NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 PL 1627, Kuopio 70211, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere University, Kalevantie 4, Tampere 33014, Finland
| | - Aki S Havulinna
- Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland
| | - Mika Ala-Korpela
- 1] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland [2] NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 PL 1627, Kuopio 70211, Finland [3] Oulu University Hospital, Kajaanintie 50, Oulu 90220, Finland [4] Computational Medicine, School of Social and Community Medicine and Medical Research Council Integrative Epidemiology Unit, University of Bristol, Senate House, Tyndall Avenue, Bristol, City of Bristol BS8 1TH, UK
| | - Veikko Salomaa
- Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Riia 23b, Tartu 51010, Estonia
| | - Markus Perola
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3] Estonian Genome Center, University of Tartu, Riia 23b, Tartu 51010, Estonia
| |
Collapse
|
29
|
Pataj Z, Ilisz I, Grecsó N, Palkó M, Fülöp F, Armstrong DW, Péter A. Enantiomeric Separation of Bicyclo[2.2.2]octane-Based 2-Amino-3-Carboxylic Acids on Macrocyclic Glycopeptide Chiral Stationary Phases. Chirality 2014; 26:200-8. [DOI: 10.1002/chir.22301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 12/09/2013] [Accepted: 12/26/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Zoltán Pataj
- Department of Inorganic and Analytical Chemistry; University of Szeged; Szeged Hungary
| | - István Ilisz
- Department of Inorganic and Analytical Chemistry; University of Szeged; Szeged Hungary
| | - Nóra Grecsó
- Department of Inorganic and Analytical Chemistry; University of Szeged; Szeged Hungary
- Institute of Pharmaceutical Chemistry; University of Szeged; Szeged Hungary
| | - Márta Palkó
- Institute of Pharmaceutical Chemistry; University of Szeged; Szeged Hungary
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry; University of Szeged; Szeged Hungary
| | - Daniel W. Armstrong
- Department of Chemistry and Biochemistry; University of Texas at Arlington; Arlington TX USA
| | - Antal Péter
- Department of Inorganic and Analytical Chemistry; University of Szeged; Szeged Hungary
| |
Collapse
|
30
|
Leiherer A, Geiger K, Muendlein A, Drexel H. Hypoxia induces a HIF-1α dependent signaling cascade to make a complex metabolic switch in SGBS-adipocytes. Mol Cell Endocrinol 2014; 383:21-31. [PMID: 24275182 PMCID: PMC3969228 DOI: 10.1016/j.mce.2013.11.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/18/2013] [Accepted: 11/15/2013] [Indexed: 12/11/2022]
Abstract
To elucidate the complex impact of hypoxia on adipose tissue, resulting in biased metabolism, insulin resistance and finally diabetes we used mature adipocytes derived from a Simpson-Golabi-Behmel syndrome patient for microarray analysis. We found a significantly increased transcription rate of genes involved in glycolysis and a striking association between the pattern of upregulated genes and disease biomarkers for diabetes mellitus and insulin resistance. Although their upregulation turned out to be HIF-1α-dependent, we identified further transcription factors mainly AP-1 components to play also an important role in hypoxia response. Analyzing the regulatory network of mentioned transcription factors and glycolysis targets we revealed a clear hint for directing glycolysis to glutathione and glycogen synthesis. This metabolic switch in adipocytes enables the cell to prevent oxidative damage in the short term but might induce lipogenesis and establish systemic metabolic disorders in the long run.
Collapse
MESH Headings
- Adipocytes/metabolism
- Adipocytes/pathology
- Adipogenesis
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Biomarkers/metabolism
- Cell Hypoxia/genetics
- Gene Expression Profiling
- Gene Expression Regulation
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/metabolism
- Genetic Diseases, X-Linked/pathology
- Gigantism/genetics
- Gigantism/metabolism
- Gigantism/pathology
- Glutathione/biosynthesis
- Glycogen/biosynthesis
- Glycolysis
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/metabolism
- Heart Defects, Congenital/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Insulin Resistance
- Intellectual Disability/genetics
- Intellectual Disability/metabolism
- Intellectual Disability/pathology
- Oligonucleotide Array Sequence Analysis
- Protein Interaction Mapping
- Signal Transduction/genetics
- Transcription Factor AP-1/genetics
- Transcription Factor AP-1/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein; Medical Central Laboratories, Feldkirch, Austria
| | - Kathrin Geiger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Department of Medicine and Cardiology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein; Drexel University College of Medicine, Philadelphia, USA.
| |
Collapse
|
31
|
Maechler P. Mitochondrial function and insulin secretion. Mol Cell Endocrinol 2013; 379:12-8. [PMID: 23792187 DOI: 10.1016/j.mce.2013.06.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 06/12/2013] [Accepted: 06/12/2013] [Indexed: 10/26/2022]
Abstract
In the endocrine fraction of the pancreas, the β-cell rapidly reacts to fluctuations in blood glucose concentrations by adjusting the rate of insulin secretion. Glucose-sensing coupled to insulin exocytosis depends on transduction of metabolic signals into intracellular messengers recognized by the secretory machinery. Mitochondria play a central role in this process by connecting glucose metabolism to insulin release. Mitochondrial activity is primarily regulated by metabolic fluxes, but also by dynamic morphology changes and free Ca(2+) concentrations. Recent advances of mitochondrial Ca(2+) homeostasis are discussed; in particular the roles of the newly-identified mitochondrial Ca(2+) uniporter MCU and its regulatory partner MICU1, as well as the mitochondrial Na(+)-Ca(2+) exchanger. This review describes how mitochondria function both as sensors and generators of metabolic signals; such as NADPH, long chain acyl-CoA, glutamate. The coupling factors are additive to the Ca(2+) signal and participate to the amplifying pathway of glucose-stimulated insulin secretion.
Collapse
Affiliation(s)
- Pierre Maechler
- Department of Cell Physiology and Metabolism, Geneva University Medical Centre, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland.
| |
Collapse
|
32
|
Melnik BC, Schmitz G, John S, Carrera-Bastos P, Lindeberg S, Cordain L. Metabolic effects of milk protein intake strongly depend on pre-existing metabolic and exercise status. Nutr Metab (Lond) 2013; 10:60. [PMID: 24225036 PMCID: PMC3856498 DOI: 10.1186/1743-7075-10-60] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 09/28/2013] [Indexed: 02/08/2023] Open
Abstract
Milk protein intake has recently been suggested to improve metabolic health. This Perspective provides evidence that metabolic effects of milk protein intake have to be regarded in the context of the individual’s pre-existing metabolic and exercise status. Milk proteins provide abundant branched-chain amino acids (BCAAs) and glutamine. Plasma BCAAs and glutamine are increased in obesity and insulin resistance, but decrease after gastric bypass surgery resulting in weight loss and improved insulin sensitivity. Milk protein consumption results in postprandial hyperinsulinemia in obese subjects, increases body weight of overweight adolescents and may thus deteriorate pre-existing metabolic disturbances of obese, insulin resistant individuals.
Collapse
|
33
|
Supale S, Thorel F, Merkwirth C, Gjinovci A, Herrera PL, Scorrano L, Meda P, Langer T, Maechler P. Loss of prohibitin induces mitochondrial damages altering β-cell function and survival and is responsible for gradual diabetes development. Diabetes 2013; 62:3488-99. [PMID: 23863811 PMCID: PMC3781460 DOI: 10.2337/db13-0152] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prohibitins are highly conserved proteins mainly implicated in the maintenance of mitochondrial function and architecture. Their dysfunctions are associated with aging, cancer, obesity, and inflammation. However, their possible role in pancreatic β-cells remains unknown. The current study documents the expression of prohibitins in human and rodent islets and their key role for β-cell function and survival. Ablation of Phb2 in mouse β-cells sequentially resulted in impairment of mitochondrial function and insulin secretion, loss of β-cells, progressive alteration of glucose homeostasis, and, ultimately, severe diabetes. Remarkably, these events progressed over a 3-week period of time after weaning. Defective insulin supply in β-Phb2(-/-) mice was contributed by both β-cell dysfunction and apoptosis, temporarily compensated by increased β-cell proliferation. At the molecular level, we observed that deletion of Phb2 caused mitochondrial abnormalities, including reduction of mitochondrial DNA copy number and respiratory chain complex IV levels, altered mitochondrial activity, cleavage of L-optic atrophy 1, and mitochondrial fragmentation. Overall, our data demonstrate that Phb2 is essential for metabolic activation of mitochondria and, as a consequence, for function and survival of β-cells.
Collapse
Affiliation(s)
- Sachin Supale
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
| | - Fabrizio Thorel
- Department of Genetic Medicine and Development, University of Geneva Medical Centre, Geneva, Switzerland
| | | | - Asllan Gjinovci
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
| | - Pedro L. Herrera
- Department of Genetic Medicine and Development, University of Geneva Medical Centre, Geneva, Switzerland
| | - Luca Scorrano
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
| | - Thomas Langer
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Corresponding author: Pierre Maechler,
| |
Collapse
|
34
|
Qi L, Qi Q, Prudente S, Mendonca C, Andreozzi F, di Pietro N, Sturma M, Novelli V, Mannino GC, Formoso G, Gervino EV, Hauser TH, Muehlschlegel JD, Niewczas MA, Krolewski AS, Biolo G, Pandolfi A, Rimm E, Sesti G, Trischitta V, Hu F, Doria A. Association between a genetic variant related to glutamic acid metabolism and coronary heart disease in individuals with type 2 diabetes. JAMA 2013; 310:821-8. [PMID: 23982368 PMCID: PMC3858847 DOI: 10.1001/jama.2013.276305] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Diabetes is associated with an elevated risk of coronary heart disease (CHD). Previous studies have suggested that the genetic factors predisposing to excess cardiovascular risk may be different in diabetic and nondiabetic individuals. OBJECTIVE To identify genetic determinants of CHD that are specific to patients with diabetes. DESIGN, SETTING, AND PARTICIPANTS We studied 5 independent sets of CHD cases and CHD-negative controls from the Nurses' Health Study (enrolled in 1976 and followed up through 2008), Health Professionals Follow-up Study (enrolled in 1986 and followed up through 2008), Joslin Heart Study (enrolled in 2001-2008), Gargano Heart Study (enrolled in 2001-2008), and Catanzaro Study (enrolled in 2004-2010). Included were a total of 1517 CHD cases and 2671 CHD-negative controls, all with type 2 diabetes. Results in diabetic patients were compared with those in 737 nondiabetic CHD cases and 1637 nondiabetic CHD-negative controls from the Nurses' Health Study and Health Professionals Follow-up Study cohorts. Exposures included 2,543,016 common genetic variants occurring throughout the genome. MAIN OUTCOMES AND MEASURES Coronary heart disease--defined as fatal or nonfatal myocardial infarction, coronary artery bypass grafting, percutaneous transluminal coronary angioplasty, or angiographic evidence of significant stenosis of the coronary arteries. RESULTS A variant on chromosome 1q25 (rs10911021) was consistently associated with CHD risk among diabetic participants, with risk allele frequencies of 0.733 in cases vs 0.679 in controls (odds ratio, 1.36 [95% CI, 1.22-1.51]; P = 2 × 10(-8)). No association between this variant and CHD was detected among nondiabetic participants, with risk allele frequencies of 0.697 in cases vs 0.696 in controls (odds ratio, 0.99 [95% CI, 0.87-1.13]; P = .89), consistent with a significant gene × diabetes interaction on CHD risk (P = 2 × 10(-4)). Compared with protective allele homozygotes, rs10911021 risk allele homozygotes were characterized by a 32% decrease in the expression of the neighboring glutamate-ammonia ligase (GLUL) gene in human endothelial cells (P = .0048). A decreased ratio between plasma levels of γ-glutamyl cycle intermediates pyroglutamic and glutamic acid was also shown in risk allele homozygotes (P = .029). CONCLUSION AND RELEVANCE A single-nucleotide polymorphism (rs10911021) was identified that was significantly associated with CHD among persons with diabetes but not in those without diabetes and was functionally related to glutamic acid metabolism, suggesting a mechanistic link.
Collapse
Affiliation(s)
- Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Qibin Qi
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Sabrina Prudente
- IRCSS Casa Sollievo della Sofferenza-Mendel Laboratory, San Giovanni Rotondo, Italy
| | | | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Græcia, Catanzaro, Italy
| | - Natalia di Pietro
- Department of Experimental and Clinical Sciences, University ‘G. d'Annunzio’, Aging Research Center, Ce.S.I., ‘G. d'Annunzio’ University Foundation, Chieti-Pescara, Italy
| | - Mariella Sturma
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Valeria Novelli
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Gaia Chiara Mannino
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medical and Surgical Sciences, University Magna Græcia, Catanzaro, Italy
| | - Gloria Formoso
- Department of Medicine and Aging Sciences, University ‘G. d'Annunzio’, Aging Research Center, Ce.S.I., ‘G. d'Annunzio’ University Foundation, Chieti-Pescara, Italy
| | - Ernest V. Gervino
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Thomas H. Hauser
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Jochen D. Muehlschlegel
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Monika A. Niewczas
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Andrzej S. Krolewski
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Gianni Biolo
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Assunta Pandolfi
- Department of Experimental and Clinical Sciences, University ‘G. d'Annunzio’, Aging Research Center, Ce.S.I., ‘G. d'Annunzio’ University Foundation, Chieti-Pescara, Italy
| | - Eric Rimm
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University Magna Græcia, Catanzaro, Italy
| | - Vincenzo Trischitta
- IRCSS Casa Sollievo della Sofferenza-Mendel Laboratory, San Giovanni Rotondo, Italy
- Research Unit of Diabetes and Endocrine Diseases, IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Frank Hu
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Alessandro Doria
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Kim EA, Yang SJ, Choi SY, Lee WJ, Cho SW. Inhibition of glutamate dehydrogenase and insulin secretion by KHG26377 does not involve ADP-ribosylation by SIRT4 or deacetylation by SIRT3. BMB Rep 2012; 45:458-63. [PMID: 22917030 DOI: 10.5483/bmbrep.2012.45.8.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the mechanisms involved in KHG26377 regulation of glutamate dehydrogenase (GDH) activity, focusing on the roles of SIRT4 and SIRT3. Intraperitoneal injection of mice with KHG26377 reduced GDH activity with concomitant repression of glucose-induced insulin secretion. Consistent with their known functions, SIRT4 ribosylated GDH and reduced its activity, and SIRT3 deacetylated GDH, increasing its activity. However, KHG26377 did not affect SIRT4-mediated ADP-ribosylation/ inhibition or SIRT3-mediated deacetylation/activation of GDH. KHG26377 had no effect on SIRT4 protein levels, and did not alter total GDH, acetylated GDH, or SIRT3 protein levels in pancreatic mitochondrial lysates. These results suggest that the mechanism by which KHG26377 inhibits GDH activity and insulin secretion does not involve ADP-ribosylation of GDH by SIRT4 or deacetylation of GDH by SIRT3.
Collapse
Affiliation(s)
- Eun-A Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
36
|
Abstract
In the endocrine fraction of the pancreas, the task of the beta-cell is to continuously and perfectly adjust insulin secretion to fluctuating blood glucose levels, thereby maintaining glycemia and nutrient homeostasis. This glucose sensing coupled to insulin exocytosis depends on transduction of metabolic signals into intracellular messengers recognized by the exocytotic machinery. Central to this metabolism-secretion coupling, mitochondrial signal transduction refers to both integration and generation of metabolic signals, connecting glucose sensing to insulin exocytosis. In response to a glucose rise, nucleotides and metabolites are generated by mitochondria and participate, together with cytosolic calcium, in the stimulation of insulin release. This review describes the role of mitochondria in metabolic signal transduction regulating insulin secretion.
Collapse
Affiliation(s)
- Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, rue Michel-Servet 1, CH-1211 Geneva 4, Switzerland.
| |
Collapse
|
37
|
Li N, Li B, Brun T, Deffert-Delbouille C, Mahiout Z, Daali Y, Ma XJ, Krause KH, Maechler P. NADPH oxidase NOX2 defines a new antagonistic role for reactive oxygen species and cAMP/PKA in the regulation of insulin secretion. Diabetes 2012; 61:2842-50. [PMID: 22933115 PMCID: PMC3478534 DOI: 10.2337/db12-0009] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In insulin-secreting cells, expression of NADPH oxidase (NOX), a potent source of ROS, has been reported, along with controversial findings regarding its function. Here, the role of NOXs was investigated: first by expression and cellular localization in mouse and human pancreatic islets, and then by functional studies in islets isolated from Nox isoform-specific knockout mice. Both human and mouse β-cells express NOX, in particular NOX2. With use of Nox isoform-specific knockout mice, functional analysis revealed Nox2 as the predominant isoform. In human islets, NOX2 colocalized with both insulin granules and endosome/lysosome membranes. Nox2-deficient islets stimulated with 22.8 mmol/L glucose exhibited potentiation of insulin release compared with controls, an effect confirmed with in vitro knockdown of Nox2. The enhanced secretory function in Nox2-deficient islets was associated with both lower superoxide levels and elevated cAMP concentrations. In control islets, GLP-1 and other cAMP inducers suppressed glucose-induced ROS production similarly to Nox2 deficiency. Inhibiting cAMP-dependent protein kinase reduced the secretory response in Nox2-null islets, although not in control islets. This study ascribes a new role for NOX2 in pancreatic β-cells as negative modulator of the secretory response, reducing cAMP/PKA signaling secondary to ROS generation. Results also show reciprocal inhibition between the cAMP/PKA pathway and ROS.
Collapse
Affiliation(s)
- Ning Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
| | - Bin Li
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
| | - Thierry Brun
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
| | | | - Zahia Mahiout
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
| | - Youssef Daali
- Clinical Pharmacology and Toxicology, Geneva University Hospital, Geneva, Switzerland
| | - Xiao-Juan Ma
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva Medical Center, Geneva, Switzerland
- Corresponding authors: Pierre Maechler, , and Karl-Heinz Krause,
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
- Corresponding authors: Pierre Maechler, , and Karl-Heinz Krause,
| |
Collapse
|
38
|
Frigerio F, Karaca M, De Roo M, Mlynárik V, Skytt DM, Carobbio S, Pajęcka K, Waagepetersen HS, Gruetter R, Muller D, Maechler P. Deletion of glutamate dehydrogenase 1 (Glud1) in the central nervous system affects glutamate handling without altering synaptic transmission. J Neurochem 2012; 123:342-8. [PMID: 22924626 DOI: 10.1111/j.1471-4159.2012.07933.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 07/30/2012] [Accepted: 08/15/2012] [Indexed: 12/11/2022]
Abstract
Glutamate dehydrogenase (GDH), encoded by GLUD1, participates in the breakdown and synthesis of glutamate, the main excitatory neurotransmitter. In the CNS, besides its primary signaling function, glutamate is also at the crossroad of metabolic and neurotransmitter pathways. Importance of brain GDH was questioned here by generation of CNS-specific GDH-null mice (CnsGlud1(-/-)); which were viable, fertile and without apparent behavioral problems. GDH immunoreactivity as well as enzymatic activity were absent in Cns-Glud1(-/-) brains. Immunohistochemical analyses on brain sections revealed that the pyramidal cells of control animals were positive for GDH, whereas the labeling was absent in hippocampal sections of Cns-Glud1(-/-) mice. Electrophysiological recordings showed that deletion of GDH within the CNS did not alter synaptic transmission in standard conditions. Cns-Glud1(-/-) mice exhibited deficient oxidative catabolism of glutamate in astrocytes, showing that GDH is required for Krebs cycle pathway. As revealed by NMR studies, brain glutamate levels remained unchanged, whereas glutamine levels were increased. This pattern was favored by up-regulation of astrocyte-type glutamate and glutamine transporters and of glutamine synthetase. Present data show that the lack of GDH in the CNS modifies the metabolic handling of glutamate without altering synaptic transmission.
Collapse
Affiliation(s)
- Francesca Frigerio
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Vetterli L, Carobbio S, Pournourmohammadi S, Martin-Del-Rio R, Skytt DM, Waagepetersen HS, Tamarit-Rodriguez J, Maechler P. Delineation of glutamate pathways and secretory responses in pancreatic islets with β-cell-specific abrogation of the glutamate dehydrogenase. Mol Biol Cell 2012; 23:3851-62. [PMID: 22875990 PMCID: PMC3459861 DOI: 10.1091/mbc.e11-08-0676] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The amino acid profile and the secretory responses of glutamate dehydrogenase (GDH)-deficient β-cells are characterized. This study shows that GDH is essential for both insulin release and net glutamate synthesis evoked by glucose. Adding cellular glutamate restored the full development of glucose-stimulated insulin secretion, showing the requirement for permissive glutamate levels. In pancreatic β-cells, glutamate dehydrogenase (GDH) modulates insulin secretion, although its function regarding specific secretagogues is unclear. This study investigated the role of GDH using a β-cell–specific GDH knockout mouse model, called βGlud1−/−. The absence of GDH in islets isolated from βGlud1–/– mice resulted in abrogation of insulin release evoked by glutamine combined with 2-aminobicyclo[2.2.1]heptane-2-carboxylic acid or l-leucine. Reintroduction of GDH in βGlud1–/– islets fully restored the secretory response. Regarding glucose stimulation, insulin secretion in islets isolated from βGlud1–/– mice exhibited half of the response measured in control islets. The amplifying pathway, tested at stimulatory glucose concentrations in the presence of KCl and diazoxide, was markedly inhibited in βGlud1–/– islets. On glucose stimulation, net synthesis of glutamate from α-ketoglutarate was impaired in GDH-deficient islets. Accordingly, glucose-induced elevation of glutamate levels observed in control islets was absent in βGlud1–/– islets. Parallel biochemical pathways, namely alanine and aspartate aminotransferases, could not compensate for the lack of GDH. However, the secretory response to glucose was fully restored by the provision of cellular glutamate when βGlud1–/– islets were exposed to dimethyl glutamate. This shows that permissive levels of glutamate are required for the full development of glucose-stimulated insulin secretion and that GDH plays an indispensable role in this process.
Collapse
Affiliation(s)
- Laurène Vetterli
- Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Implication of mitochondrial cytoprotection in human islet isolation and transplantation. Biochem Res Int 2012; 2012:395974. [PMID: 22611495 PMCID: PMC3352213 DOI: 10.1155/2012/395974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Accepted: 01/30/2012] [Indexed: 12/23/2022] Open
Abstract
Islet transplantation is a promising therapy for type 1 diabetes mellitus; however, success rates in achieving both short- and long-term insulin independence are not consistent, due in part to inconsistent islet quality and quantity caused by the complex nature and multistep process of islet isolation and transplantation. Since the introduction of the Edmonton Protocol in 2000, more attention has been placed on preserving mitochondrial function as increasing evidences suggest that impaired mitochondrial integrity can adversely affect clinical outcomes. Some recent studies have demonstrated that it is possible to achieve islet cytoprotection by maintaining mitochondrial function and subsequently to improve islet transplantation outcomes. However, the benefits of mitoprotection in many cases are controversial and the underlying mechanisms are unclear. This article summarizes the recent progress associated with mitochondrial cytoprotection in each step of the islet isolation and transplantation process, as well as islet potency and viability assays based on the measurement of mitochondrial integrity. In addition, we briefly discuss immunosuppression side effects on islet graft function and how transplant site selection affects islet engraftment and clinical outcomes.
Collapse
|
41
|
da Silva PMR, Batista TM, Ribeiro RA, Zoppi CC, Boschero AC, Carneiro EM. Decreased insulin secretion in islets from protein malnourished rats is associated with impaired glutamate dehydrogenase function: effect of leucine supplementation. Metabolism 2012; 61:721-32. [PMID: 22078937 DOI: 10.1016/j.metabol.2011.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 08/27/2011] [Accepted: 09/27/2011] [Indexed: 02/06/2023]
Abstract
We herein studied the role of glutamate dehydrogenase (GDH), in response to leucine (LEU) supplementation, upon insulin secretion of malnourished rats. Weaned male Wistar rats were fed normal-protein (17%) or low-protein diet (6%, LP) for 8 weeks. Half of the rats of each group were supplemented with LEU (1.5%) in the drinking water for the following 4 weeks. Gene and protein expressions, static insulin secretion, and cytoplasmic Ca(2+) oscillations were measured. Glutamate dehydrogenase messenger RNA was 58% lower in LP islets, and LEU supplementation augmented it in 28%. The LP islets secreted less insulin when exposed to 20 mmol/L LEU, 20 mmol/L LEU + 2 mmol/L glutamine (with or without 5 mmol/L aminooxyacetic acid, a branched chain aminotransferase inhibitor, or 20 μmol/L epigallocatechin gallate, a GDH inhibitor), 20 mmol/L α-ketoisocaproate, glutamine + 20 mmol/L β-2-aminobicyclo[2.2.1]heptane-2-carboxylic acid (a GDH activator), and 22.2 mmol/L glucose. Leucine supplementation augmented insulin secretion to levels found in normal-protein islets in all the above conditions, an effect that was blunted when islets were incubated with epigallocatechin gallate. The glutamine + β-2-aminobicyclo[2.2.1]heptane-2-carboxylic acid-induced increased [Ca(2+)](i) and oscillations were higher than those for LP islets. Leucine supplementation normalized these parameters in LP islets. Impaired GDH function was associated with lower insulin release in LP islets, and LEU supplementation normalized insulin secretion via restoration of GDH function. In addition, GDH may contribute to insulin secretion through ameliorations of Ca(2+) handling in LP islets.
Collapse
Affiliation(s)
- Priscilla Muniz Ribeiro da Silva
- Department of Anatomy, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas,PO Box 6109, CEP 13083-970 Campinas, SP, Brazil
| | | | | | | | | | | |
Collapse
|
42
|
Göhring I, Mulder H. Glutamate dehydrogenase, insulin secretion, and type 2 diabetes: a new means to protect the pancreatic β-cell? J Endocrinol 2012; 212:239-42. [PMID: 22232141 DOI: 10.1530/joe-11-0481] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this issue of Journal of Endocrinology, Dr Han and colleagues report a protective effect of the glutamate dehydrogenase activator 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) under diabetes-like conditions that impair β-cell function in both a pancreatic β-cell line and db/db mice. Based on these observations, the authors suggest that BCH could serve as a novel treatment modality in type 2 diabetes. The present commentary discusses the importance of the findings. Some additional questions are raised, which may be addressed in future investigations, as there is some concern regarding the BCH treatment of β-cell failure.
Collapse
Affiliation(s)
- Isabel Göhring
- Unit of Molecular Metabolism, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Clinical Research Center 91:11, SE-205 02, Malmö, Sweden
| | | |
Collapse
|
43
|
Han SJ, Choi SE, Yi SA, Lee SJ, Kim HJ, Kim DJ, Lee HC, Lee KW, Kang Y. β-Cell-protective effect of 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid as a glutamate dehydrogenase activator in db/db mice. J Endocrinol 2012; 212:307-15. [PMID: 22131441 DOI: 10.1530/joe-11-0340] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
2-Aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) is an activator of glutamate dehydrogenase (GDH), which is a mitochondrial enzyme with an important role in insulin secretion. We investigated the effect of BCH on the high-glucose (HG)-induced reduction in glucose-stimulated insulin secretion (GSIS), the HG/palmitate (PA)-induced reduction in insulin gene expression, and HG/PA-induced β-cell death. We also studied whether long-term treatment with BCH lowers blood glucose and improves β-cell integrity in db/db mice. We evaluated GSIS, insulin gene expression, and DNA fragmentation in INS-1 cells exposed to HG or HG/PA in the presence or absence of BCH. An in vivo study was performed in which 7-week-old diabetic db/db mice were treated with BCH (0.7 g/kg, n = 10) and placebo (n = 10) every other day for 6 weeks. After treatment, an intraperitoneal glucose tolerance test and immunohistological examinations were performed. Treatment with BCH blocked HG-induced GSIS inhibition and the HG/PA-induced reduction in insulin gene expression in INS-1 cells. In addition, BCH significantly reduced HG/PA-induced INS-1 cell death and phospho-JNK level. BCH treatment improved glucose tolerance and insulin secretion in db/db mice. BCH treatment also increased the ratio of insulin-positive β-cells to total islet area (P < 0.05) and reduced the percentage of β-cells expressing cleaved caspase 3 (P < 0.05). In conclusion, the GDH activator BCH improved glycemic control in db/db mice. This anti-diabetic effect may be associated with improved insulin secretion, preserved islet architecture, and reduced β-cell apoptosis.
Collapse
Affiliation(s)
- Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, San 5, Wonchon-dong, Yeongtong-gu, Suwon 443-721, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Huypens PR, Huang M, Joseph JW. Overcoming the spatial barriers of the stimulus secretion cascade in pancreatic β-cells. Islets 2012; 4:1-116. [PMID: 22143007 DOI: 10.4161/isl.18338] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The ability of the pancreatic β-cells to adapt the rate of insulin release in accordance to changes in circulating glucose levels is essential for glucose homeostasis. Two spatial barriers imposed by the plasma membrane and inner mitochondrial membrane need to be overcome in order to achieve stringent coupling between the different steps in the stimulus-secretion cascade. The first spatial barrier is overcome by the presence of a glucose transporter (GLUT) in the plasma membrane, whereas a low affinity hexokinase IV (glucokinase, GK) in the cytosol conveys glucose availability into a metabolic flux that triggers and accelerates insulin release. The mitochondrial inner membrane comprises a second spatial barrier that compartmentalizes glucose metabolism into glycolysis (cytosol) and tricarboxylate (TCA) cycle (mitochondrial matrix). The exchange of metabolites between cytosol and mitochondrial matrix is mediated via a set of mitochondrial carriers, including the aspartate-glutamate carrier (aralar1), α- ketoglutarate carrier (OGC), ATP/ADP carrier (AAC), glutamate carrier (GC1), dicarboxylate carrier (DIC) and citrate/isocitrate carrier (CIC). The scope of this review is to provide an overview of the role these carriers play in stimulus-secretion coupling and discuss the importance of these findings in the context of the exquisite glucose responsive state of the pancreatic β-cell.
Collapse
Affiliation(s)
- Peter R Huypens
- School of Pharmacy; Health Science Campus; University of Waterloo; Kitchener, CN Canada
| | - Mei Huang
- School of Pharmacy; Health Science Campus; University of Waterloo; Kitchener, CN Canada
| | - Jamie W Joseph
- School of Pharmacy; Health Science Campus; University of Waterloo; Kitchener, CN Canada
| |
Collapse
|
45
|
Palkó M, Sohár P, Fülöp F. Synthesis and transformations of di-endo-3-aminobicyclo-[2.2.2]oct-5-ene-2-carboxylic acid derivatives. Molecules 2011; 16:7691-705. [PMID: 21900870 PMCID: PMC6264698 DOI: 10.3390/molecules16097691] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/24/2011] [Accepted: 09/06/2011] [Indexed: 12/02/2022] Open
Abstract
all-endo-3-amino-5-hydroxybicyclo[2.2.2]octane-2-carboxylic acid (13) and all-endo-5-amino-6-(hydroxymethyl)bicyclo[2.2.2]octan-2-ol (10) were prepared via dihydro-1,3-oxazine or γ-lactone intermediates by the stereoselective functionalization of an N-protected derivative of endo-3-aminobicyclo[2.2.2]oct-5-ene-2-carboxylic acid (2). Ring closure of β-amino ester 4 resulted in tricyclic pyrimidinones 15 and 16. The structures, stereochemistry and relative configurations of the synthesized compounds were determined by IR and NMR.
Collapse
Affiliation(s)
- Márta Palkó
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Eötvös utca 6, Hungary
| | - Pál Sohár
- Institute of Chemistry, Eötvös Lóránd University, H-1518 Budapest, POB 32, Hungary
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Eötvös utca 6, Hungary
- Author to whom correspondence should be addressed; ; Tel.: +36-62-545-562; Fax: +36-62-545-705
| |
Collapse
|
46
|
Karaca M, Frigerio F, Maechler P. From pancreatic islets to central nervous system, the importance of glutamate dehydrogenase for the control of energy homeostasis. Neurochem Int 2011; 59:510-7. [DOI: 10.1016/j.neuint.2011.03.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 03/21/2011] [Accepted: 03/23/2011] [Indexed: 11/25/2022]
|
47
|
Li Y, Zhao S, Zhang W, Zhao P, He B, Wu N, Han P. Epigallocatechin-3-O-gallate (EGCG) attenuates FFAs-induced peripheral insulin resistance through AMPK pathway and insulin signaling pathway in vivo. Diabetes Res Clin Pract 2011; 93:205-214. [PMID: 21514684 DOI: 10.1016/j.diabres.2011.03.036] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 03/20/2011] [Accepted: 03/28/2011] [Indexed: 12/16/2022]
Abstract
We aimed to investigate the effects and possible mechanisms of Epigallocatechin-3-O-gallate (EGCG) on free fatty acids (FFAs)-induced peripheral insulin resistance in vivo. Overnight-fasted Wistar rats were subjected to 48-h intravenous infusion of either saline or Intralipid plus heparin (IH) with or without different doses of EGCG co-injection. Hyperinsulinemic-euglycemic clamp was performed in awake rats to assess peripheral insulin sensitivity. Co-injection with EGCG significantly prevented FFAs-induced peripheral insulin resistance, decreased plasma markers of oxidative stress: malondialdehyde (MDA) and 8-isoprostaglandin, and increased antioxidant enzymes: superoxide dismutases (SOD) and Glutathione peroxidase (GPx). Furthermore, EGCG treatment reversed IH-induced: (1) decrease in Thr172 phosphorylation of AMP activated protein kinase (AMPK); (2) increase in protein kinase Cθ(PKCθ) membrane translocation and Ser307 phosphorylation of insulin receptor substrate-1 (IRS-1); (3) decrease in Ser473 phosphorylation of Akt and Glucose transporter 4 (GLUT4) translocation in skeletal muscle and adipose tissue. Our data suggest that EGCG treatment ameliorated FFAs-induced peripheral insulin resistance in vivo, and this might be through decreasing oxidative stress and PKCθ membrane translocation, activating the AMPK pathway and improving insulin signaling pathway in vivo. This study suggests the therapeutic value of EGCG in protecting from insulin resistance caused by elevated FFAs.
Collapse
Affiliation(s)
- Yan Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Sheng Zhao
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Wei Zhang
- Department of Endocrinology, Affiliated Fourth Hospital, China Medical University, Shenyang 110032, China.
| | - Peng Zhao
- Department of Medical Record, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Bing He
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Na Wu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ping Han
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
48
|
Zoppi CC, Calegari VC, Silveira LR, Carneiro EM, Boschero AC. Exercise training enhances rat pancreatic islets anaplerotic enzymes content despite reduced insulin secretion. Eur J Appl Physiol 2011; 111:2369-74. [PMID: 21287194 DOI: 10.1007/s00421-011-1842-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 01/14/2011] [Indexed: 01/09/2023]
Abstract
Endurance exercise has been shown to reduce pancreatic islets glucose-stimulated insulin secretion (GSIS). Anaplerotic/cataplerotic pathways are directly related to GSIS signaling. However, the effect of endurance training upon pancreatic islets anaplerotic enzymes is still unknown. In this sense, we tested the hypothesis that endurance exercise decreases GSIS by reducing anaplerotic/cataplerotic enzymes content. Male Wistar rats were randomly assigned to one of the four experimental groups as follows: control sedentary group (CTL), trained 1 day per week (TRE1×), trained 3 days per week (TRE3×) and trained 5 days per week (TRE5x) and submitted to an 8 weeks endurance-training protocol. After the training protocol, pancreatic islets were isolated and incubated with basal (2.8 mM) and stimulating (16.7 mM) glucose concentrations for GSIS measurement by radioimmunoassay. In addition, pyruvate carboxylase (PYC), pyruvate dehydrogenase (PDH), pyruvate dehydrogenase kinase 4 (PDK4), ATP-citrate lyase (ACL) and glutamate dehydrogenase (GDH) content were quantified by western blotting. Our data showed that 8 weeks of chronic endurance exercise reduced GSIS by 50% in a dose-response manner according to weekly exercise frequency. PYC showed significant twofold increase in TRE3×. PYC enhancement was even higher in TRE5× (p < 0.0001). PDH and PDK4 reached significant 25 and 50% enhancement, respectively compared with CTL. ACL and GDH also reported significant 50 and 75% increase, respectively. The absence of exercise-induced correlations among GSIS and anaplerotic/cataplerotic enzymes suggests that exercise may control insulin release by activating other signaling pathways. The observed anaplerotic and cataplerotic enzymes enhancement might be related to β-cell surviving rather than insulin secretion.
Collapse
Affiliation(s)
- Claudio C Zoppi
- Department of Anatomy, Cellular Biology and Physiology and Biophysics, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP: 13083-865, Brazil.
| | | | | | | | | |
Collapse
|
49
|
Yang J, Chi Y, Burkhardt BR, Guan Y, Wolf BA. Leucine metabolism in regulation of insulin secretion from pancreatic beta cells. Nutr Rev 2010; 68:270-9. [PMID: 20500788 DOI: 10.1111/j.1753-4887.2010.00282.x] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Leucine, a branched-chain amino acid that must be supplied in the daily diet, plays an important role in controlling protein synthesis and regulating cell metabolism in various cell types. In pancreatic beta cells, leucine acutely stimulates insulin secretion by serving as both metabolic fuel and allosteric activator of glutamate dehydrogenase to enhance glutaminolysis. Leucine has also been shown to regulate gene transcription and protein synthesis in pancreatic islet beta cells via both mTOR-dependent and -independent pathways at physiological concentrations. Long-term treatment with leucine has been shown to improve insulin secretory dysfunction of human diabetic islets via upregulation of certain key metabolic genes. In vivo, leucine administration improves glycemic control in humans and rodents with type 2 diabetes. This review summarizes and discusses the recent findings regarding the effects of leucine metabolism on pancreatic beta-cell function.
Collapse
Affiliation(s)
- Jichun Yang
- Department of Physiology and Pathophysiology, Peking University Diabetes Center, Peking University Health Science Center, Beijing, China.
| | | | | | | | | |
Collapse
|
50
|
Yang SJ, Hahn HG, Choi SY, Cho SW. Inhibitory effects of KHG26377 on glutamate dehydrogenase activity in cultured islets. BMB Rep 2010; 43:245-9. [PMID: 20423608 DOI: 10.5483/bmbrep.2010.43.4.245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
GDH has been known to be related with hyperinsulinismhyperammonemia syndrome. We have screened new drugs with a view to developing effective drugs modulating GDH activity. In the present work, we investigated the effects of a new drug, KHG26377 on glutamate formation and GDH activity in cultured rat islets. When KHG26377 was added to the culture medium for 24 h prior to kinetic analysis, the V(max) of GDH was decreased by 59% whereas K(m) is not significantly changed. The concentration of glutamate decreased by 50% and perfusion of islets with KHG26377 reduced insulin release by up to 55%. Our results show that KHG26377 regulates insulin release by inhibiting GDH activity in primary cultured islets and support the previous studies for the connection between GDH activity and insulin release. Further studies are required to determine in vivo effects and pharmacokinetics of the drug.
Collapse
Affiliation(s)
- Seung-Ju Yang
- 1Department of Biomedical Laboratory Science, Konyang University, Daejeon 302-718, USA
| | | | | | | |
Collapse
|