1
|
Wei M, Wang Y, Zhang Y, Qiao Y. Plin5: A potential therapeutic target for type 2 diabetes mellitus. Diabetol Metab Syndr 2025; 17:114. [PMID: 40176076 PMCID: PMC11963521 DOI: 10.1186/s13098-025-01680-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/22/2025] [Indexed: 04/04/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a kind of metabolic disease characterized by aberrant insulin secretion as a result of -cell loss or injury, or by impaired insulin sensitivity of peripheral tissues, which finally results in insulin resistance and a disturbance of glucose and lipid metabolism. Among them, lipid metabolism disorders lead to lipotoxicity through oxidative stress and inflammatory response, destroying the structure and function of tissues and cells. Abnormal lipid metabolism can lead to abnormal insulin signaling and disrupt glucose metabolism through a variety of pathways. Therefore, emphasizing lipid metabolism may be a crucial step in the prevention and treatment of T2DM. Plin5 is a lipid droplet surface protein, which can bi-directionally regulate lipid metabolism and plays an important role in lipolysis and fat synthesis. Plin5 can simultaneously decrease the buildup of free fatty acids in the cytoplasm, improve mitochondrial uptake of free fatty acids, speed up fatty acid oxidation through lipid drops-mitochondria interaction, and lessen lipotoxicity. In oxidative tissues like the heart, liver, and skeletal muscle, Plin5 is extensively expressed. And Plin5 is widely involved in β-cell apoptosis, insulin resistance, oxidative stress, inflammatory response and other pathological processes, which has important implications for exploring the pathogenesis of T2DM. In addition, recent studies have found that Plin5 is also closely related to T2DM and cancer, which provides a new perspective for exploring the relationship between T2DM and cancer.
Collapse
Affiliation(s)
- Mengjuan Wei
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yan Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Yufei Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yun Qiao
- Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
3
|
Ezzati‐Mobaser S, Yarahmadi S, Dadkhah Nikroo N, Maleki MH, Yousefi Z, Golpour P, Nourbakhsh M, Nourbakhsh M. Adipose triglyceride lipase gene expression in peripheral blood mononuclear cells of subjects with obesity and its association with insulin resistance, inflammation and lipid accumulation in liver. Obes Sci Pract 2024; 10:e716. [PMID: 38263987 PMCID: PMC10804332 DOI: 10.1002/osp4.716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 01/25/2024] Open
Abstract
INTRODUCTION Adipose triglyceride lipase (ATGL) is a crucial enzyme responsible for the release of fatty acids from various tissues. The expression of ATGL is regulated by insulin and this enzyme is linked to Insulin resistance (IR). On the other hand, ATGL-mediated lipolysis is connected to macrophage function and thus, ATGL is involved in inflammation and the pathogenesis of lipid-related disorders. This study aimed to investigate the correlation between ATGL, obesity, Metabolic Syndrome (MetS), and inflammation. METHODS A total of 100 participants, including 50 individuals with obesity and 50 healthy participants, were recruited for this study and underwent comprehensive clinical evaluations. Blood samples were collected to measure plasma lipid profiles, glycemic indices, and liver function tests. Additionally, peripheral blood mononuclear cells (PBMCs) were isolated and used for the assessment of the gene expression of ATGL, using real-time PCR. Furthermore, PBMCs were cultured and exposed to lipopolysaccharides (LPS) with simultaneous ATGL inhibition, and the gene expression of inflammatory cytokines, along with the secretion of prostaglandin E2 (PGE2), were measured. RESULTS The gene expression of ATGL was significantly elevated in PBMCs obtained from participants with obesity and was particularly higher in those diagnosed with MetS. It exhibited a correlation with insulin levels and Homeostatic Model Assessment for IR (HOMA-IR), and it was associated with lipid accumulation in the liver. Stimulation with LPS increased ATGL expression in PBMCs, while inhibition of ATGL attenuated the inflammatory responses induced by LPS. CONCLUSIONS Obesity and MetS were associated with dysregulation of ATGL. ATGL might play a role in the upregulation of inflammatory cytokines and act as a significant contributor to the development of metabolic abnormalities related to obesity.
Collapse
Affiliation(s)
| | - Sahar Yarahmadi
- Department of BiochemistryFaculty of MedicineIran University of Medical SciencesTehranIran
| | - Nikta Dadkhah Nikroo
- Metabolic Disorders Research CenterEndocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Mohammad Hasan Maleki
- Department of BiochemistrySchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Zeynab Yousefi
- Department of Clinical BiochemistryFaculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Pegah Golpour
- Department of BiochemistryFaculty of MedicineShahid Sadoughi University of Medical SciencesTehranIran
| | - Mona Nourbakhsh
- Hazrat Aliasghar HospitalSchool of MedicineIran University of Medical SciencesTehranIran
| | - Mitra Nourbakhsh
- Finetech in Medicine Research CenterIran University of Medical SciencesTehranIran
- Department of BiochemistryFaculty of MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
4
|
Li J, Wang Y, Yang P, Han H, Zhang G, Xu H, Quan K. Overexpression of ATGL impairs lipid droplet accumulation by accelerating lipolysis in goat mammary epithelial cells. Anim Biotechnol 2023; 34:3126-3134. [PMID: 36306180 DOI: 10.1080/10495398.2022.2136678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Adipose triglyceride lipase (ATGL) is the key enzyme for the degradation of triacylglycerols (TAGs). It functions in concert with other enzymes to mobilize TAG and supply fatty acids (FAs) for energy production. Dysregulated lipolysis leads to excess concentrations of circulating FAs, which may lead to destructive and lipotoxic effects to the organism. To understand the role of ATGL in mammary lipid metabolism, ATGL was overexpressed in goat mammary epithelial cells (GMECs) by using a recombinant adenovirus system. ATGL overexpression decreased lipid droplet (LD) accumulation and cellular TG content (p < 0.05) along with a decrease in the expression of the key enzyme that catalyzes the final step of TG synthesis (DGAT). Significant increases were observed in the expression of genes related to lipolysis (hormone-sensitive lipase [HSL]) and FA desaturation (SCD) by ATGL overexpression. Genes responsible for FA oxidation (PPARα), LD formation and secretion (ADRP and BTN1A1), and long-chain FA uptake (CD36) were all decreased by ATGL overexpression (p < 0.05). The primary products of TAG lipolysis, free FAs (FFAs), were notably increased in the ATGL-overexpressing cells. Taken together, our results demonstrated that ATGL activation impairs lipid formation partially through accelerating lipolysis in GMECs.
Collapse
Affiliation(s)
- Jun Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Yaling Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, PR China
| | - Pengkun Yang
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Haoyuan Han
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Guizhi Zhang
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, PR China
| | - Kai Quan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| |
Collapse
|
5
|
Ghosh A, Peyot ML, Leung YH, Ravenelle F, Madiraju SRM, Prentki M. A peripherally restricted cannabinoid-1 receptor inverse agonist promotes insulin secretion and protects from cytokine toxicity in human pancreatic islets. Eur J Pharmacol 2023; 944:175589. [PMID: 36773683 DOI: 10.1016/j.ejphar.2023.175589] [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: 11/25/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
The cannabinoid receptor CB1R is expressed in pancreatic β-cells; CB1R increased activity is associated with diabetes, obesity, cardiovascular disorders as well as decreased insulin secretion and insulin resistance. CB1R was shown to signal through G-protein coupling as well as β-arrestins in β-cells. Peripherally restricted CB1R inverse agonists purportedly have beneficial effects on insulin secretion in β-cells, without the unwanted effects in the central nervous system. Here we show that a peripherally restricted CB1R inverse agonist, MRI-1891, augments glucose stimulated insulin secretion in isolated human pancreatic islets and mouse islets. The insulin secretion enhancing effect of MRI-1891 is comparable to exendin-4, an analogue of the glucagon like peptide-1 (GLP1). Moreover, MRI-1891 treatment protects isolated human islet cells against cytokine-induced apoptosis, similar to exendin-4. Thus, MRI-1891, a new class of CB1R inverse agonist, may be considered a potential therapeutic for both type 1 and type 2 diabetes because of its ability to protect pancreatic β-cells from cytokine toxicity and to promote insulin secretion.
Collapse
Affiliation(s)
- Anindya Ghosh
- Departments of Nutrition, Biochemistry and Molecular Medicine, Université de Montréal, Montreal Diabetes Research Center, CRCHUM, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - Marie-Line Peyot
- Departments of Nutrition, Biochemistry and Molecular Medicine, Université de Montréal, Montreal Diabetes Research Center, CRCHUM, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - Yat Hei Leung
- Departments of Nutrition, Biochemistry and Molecular Medicine, Université de Montréal, Montreal Diabetes Research Center, CRCHUM, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - François Ravenelle
- Inversago Pharma Inc., 1100 Rene-Levesque West, Suite 1110, Montreal, QC, H3B 4N4, Canada
| | - S R Murthy Madiraju
- Departments of Nutrition, Biochemistry and Molecular Medicine, Université de Montréal, Montreal Diabetes Research Center, CRCHUM, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular Medicine, Université de Montréal, Montreal Diabetes Research Center, CRCHUM, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada.
| |
Collapse
|
6
|
Deficiency of Adipose Triglyceride Lipase Induces Metabolic Syndrome and Cardiomyopathy in Zebrafish. Int J Mol Sci 2022; 24:ijms24010117. [PMID: 36613558 PMCID: PMC9820674 DOI: 10.3390/ijms24010117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Lipid metabolism dysfunction is related to clinical disorders including obesity, cancer, liver steatosis, and cardiomyopathy. Impaired lipolytic enzymes result in altered release of free fatty acids. The dramatic change in dyslipidemia is important in lipotoxic cardiomyopathy. Adipose triglyceride lipase (ATGL) catalyzes the lipolysis of triacylglycerol to reduce intramyocardial triglyceride levels in the heart and improve myocardial function. We examined the role of ATGL in metabolic cardiomyopathy by developing an Atgl knockout (ALKO) zebrafish model of metabolic cardiomyopathy disease by continuously expressing CRISPR/Cas9 protein and atgl gene guide RNAs (gRNAs). The expressed Cas9 protein bound to four gRNAs targeting the atgl gene locus, facilitating systemic gene KO. Ablation of Atgl interfered with lipid metabolism, which induced hyperlipidemia and hyperglycemia. ALKO adults and embryos displayed hypertrophic hearts. ALKO presented a typical dilated cardiomyopathy profile with a remarkable reduction in four sarcomere genes (myosin heavy chain 7-like, actin alpha cardiac muscle 1b, myosin binding protein C3, and troponin T type 2a) and two Ca2+ handling regulator genes (tropomyosin 4b and ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2b). Immune cell infiltration in cardiac tissue of ALKO provided direct evidence of advanced metabolic cardiomyopathy. The presently described model could become a powerful tool to clarify the underlying mechanism between metabolic disorders and cardiomyopathies.
Collapse
|
7
|
Chen S, Huang X. Cytosolic lipolysis in non-adipose tissues: energy provision and beyond. FEBS J 2022; 289:7385-7398. [PMID: 34407292 DOI: 10.1111/febs.16161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/18/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022]
Abstract
Cytosolic lipolysis is a well-defined biochemical process that plays important roles in the mobilization of stored neutral lipids. Lipid turnover, regulated by cytosolic lipolysis, has been extensively studied in adipose tissue, liver, and muscle. The storage and utilization of neutral lipids is a basic function of most, if not all, tissues and cells. In this review, we focus on the functions of cytosolic lipolysis mainly in non-adipose tissues and in several physiological processes, including cancer, longevity, and pathogen infection. The mechanisms underlying the impact of cytosolic lipolysis on these events will be discussed. Detailed understanding of cytosolic lipolysis in both adipose and non-adipose tissues will have implications for future clinical translation.
Collapse
Affiliation(s)
- Siyu Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
8
|
Ježek P, Holendová B, Jabůrek M, Dlasková A, Plecitá-Hlavatá L. Contribution of Mitochondria to Insulin Secretion by Various Secretagogues. Antioxid Redox Signal 2022; 36:920-952. [PMID: 34180254 PMCID: PMC9125579 DOI: 10.1089/ars.2021.0113] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Significance: Mitochondria determine glucose-stimulated insulin secretion (GSIS) in pancreatic β-cells by elevating ATP synthesis. As the metabolic and redox hub, mitochondria provide numerous links to the plasma membrane channels, insulin granule vesicles (IGVs), cell redox, NADH, NADPH, and Ca2+ homeostasis, all affecting insulin secretion. Recent Advances: Mitochondrial redox signaling was implicated in several modes of insulin secretion (branched-chain ketoacid [BCKA]-, fatty acid [FA]-stimulated). Mitochondrial Ca2+ influx was found to enhance GSIS, reflecting cytosolic Ca2+ oscillations induced by action potential spikes (intermittent opening of voltage-dependent Ca2+ and K+ channels) or the superimposed Ca2+ release from the endoplasmic reticulum (ER). The ATPase inhibitory factor 1 (IF1) was reported to tune the glucose sensitivity range for GSIS. Mitochondrial protein kinase A was implicated in preventing the IF1-mediated inhibition of the ATP synthase. Critical Issues: It is unknown how the redox signal spreads up to the plasma membrane and what its targets are, what the differences in metabolic, redox, NADH/NADPH, and Ca2+ signaling, and homeostasis are between the first and second GSIS phase, and whether mitochondria can replace ER in the amplification of IGV exocytosis. Future Directions: Metabolomics studies performed to distinguish between the mitochondrial matrix and cytosolic metabolites will elucidate further details. Identifying the targets of cell signaling into mitochondria and of mitochondrial retrograde metabolic and redox signals to the cell will uncover further molecular mechanisms for insulin secretion stimulated by glucose, BCKAs, and FAs, and the amplification of secretion by glucagon-like peptide (GLP-1) and metabotropic receptors. They will identify the distinction between the hub β-cells and their followers in intact and diabetic states. Antioxid. Redox Signal. 36, 920-952.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Blanka Holendová
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Jabůrek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Dlasková
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
9
|
Grabner GF, Xie H, Schweiger M, Zechner R. Lipolysis: cellular mechanisms for lipid mobilization from fat stores. Nat Metab 2021; 3:1445-1465. [PMID: 34799702 DOI: 10.1038/s42255-021-00493-6] [Citation(s) in RCA: 359] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022]
Abstract
The perception that intracellular lipolysis is a straightforward process that releases fatty acids from fat stores in adipose tissue to generate energy has experienced major revisions over the last two decades. The discovery of new lipolytic enzymes and coregulators, the demonstration that lipophagy and lysosomal lipolysis contribute to the degradation of cellular lipid stores and the characterization of numerous factors and signalling pathways that regulate lipid hydrolysis on transcriptional and post-transcriptional levels have revolutionized our understanding of lipolysis. In this review, we focus on the mechanisms that facilitate intracellular fatty-acid mobilization, drawing on canonical and noncanonical enzymatic pathways. We summarize how intracellular lipolysis affects lipid-mediated signalling, metabolic regulation and energy homeostasis in multiple organs. Finally, we examine how these processes affect pathogenesis and how lipolysis may be targeted to potentially prevent or treat various diseases.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Hao Xie
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
10
|
Han SL, Liu Y, Limbu SM, Chen LQ, Zhang ML, Du ZY. The reduction of lipid-sourced energy production caused by ATGL inhibition cannot be compensated by activation of HSL, autophagy, and utilization of other nutrients in fish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:173-188. [PMID: 33245450 DOI: 10.1007/s10695-020-00904-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/18/2020] [Indexed: 06/11/2023]
Abstract
The adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL)-mediated lipolysis play important roles in lipid catabolism. ATGL is considered the central rate-limiting enzyme in the mobilization of fatty acids in mammals. Currently, severe fat accumulation has been commonly detected in farmed fish globally. However, the ATGL-mediated lipolysis and the potential synergy among ATGL, HSL, and autophagy, which is another way for lipid breakdown, have not been intensively understood in fish. In the present study, we added Atglistatin as an ATGL-specific inhibitor into the zebrafish diet and fed to the fish for 5 weeks. The results showed that the Atglistatin-treated fish exhibited severe fat deposition, reduced oxygen consumption, and fatty acid β-oxidation, accompanied with increased oxidative stress and inflammation. Furthermore, the Atglistatin-treated fish elevated total and phosphorylation protein expressions of HSL. However, the free fatty acids and lipase activities in organs were still systemically reduced in the Atglistatin-treated fish, and the autophagy marker LC3 was also decreased in the liver. On the other hand, glycogenolysis was stimulated but blood glucose was higher in the Atglistatin-treated fish. The transcriptomic analysis also provided the hint that the protein turnover efficiency in Atglistatin-treated fish was likely to be accelerated, but the protein content in whole fish was not affected. Taken together, ATGL plays crucial roles in energy homeostasis such that its inhibition causes loss of lipid-sourced energy production, which cannot be compensated by activation of HSL, autophagy, and utilization of other nutrients.
Collapse
Affiliation(s)
- Si-Lan Han
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yan Liu
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Samwel M Limbu
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
- Department of Aquatic Sciences and Fisheries Technology, University of Dar es Salaam, P.O. Box 35064, Dar es Salaam, Tanzania
- ECNU-UDSM Joint Research Center for Aquaculture and Fish Biology (JRCAFB), Dar es Salaam, Tanzania
| | - Li-Qiao Chen
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Mei-Ling Zhang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen-Yu Du
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China.
- ECNU-UDSM Joint Research Center for Aquaculture and Fish Biology (JRCAFB), Shanghai, China.
| |
Collapse
|
11
|
Ježek P, Holendová B, Jabůrek M, Tauber J, Dlasková A, Plecitá-Hlavatá L. The Pancreatic β-Cell: The Perfect Redox System. Antioxidants (Basel) 2021; 10:antiox10020197. [PMID: 33572903 PMCID: PMC7912581 DOI: 10.3390/antiox10020197] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β-cell insulin secretion, which responds to various secretagogues and hormonal regulations, is reviewed here, emphasizing the fundamental redox signaling by NADPH oxidase 4- (NOX4-) mediated H2O2 production for glucose-stimulated insulin secretion (GSIS). There is a logical summation that integrates both metabolic plus redox homeostasis because the ATP-sensitive K+ channel (KATP) can only be closed when both ATP and H2O2 are elevated. Otherwise ATP would block KATP, while H2O2 would activate any of the redox-sensitive nonspecific calcium channels (NSCCs), such as TRPM2. Notably, a 100%-closed KATP ensemble is insufficient to reach the -50 mV threshold plasma membrane depolarization required for the activation of voltage-dependent Ca2+ channels. Open synergic NSCCs or Cl- channels have to act simultaneously to reach this threshold. The resulting intermittent cytosolic Ca2+-increases lead to the pulsatile exocytosis of insulin granule vesicles (IGVs). The incretin (e.g., GLP-1) amplification of GSIS stems from receptor signaling leading to activating the phosphorylation of TRPM channels and effects on other channels to intensify integral Ca2+-influx (fortified by endoplasmic reticulum Ca2+). ATP plus H2O2 are also required for branched-chain ketoacids (BCKAs); and partly for fatty acids (FAs) to secrete insulin, while BCKA or FA β-oxidation provide redox signaling from mitochondria, which proceeds by H2O2 diffusion or hypothetical SH relay via peroxiredoxin "redox kiss" to target proteins.
Collapse
|
12
|
Possik E, Al-Mass A, Peyot ML, Ahmad R, Al-Mulla F, Madiraju SRM, Prentki M. New Mammalian Glycerol-3-Phosphate Phosphatase: Role in β-Cell, Liver and Adipocyte Metabolism. Front Endocrinol (Lausanne) 2021; 12:706607. [PMID: 34326816 PMCID: PMC8313997 DOI: 10.3389/fendo.2021.706607] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/25/2021] [Indexed: 11/25/2022] Open
Abstract
Cardiometabolic diseases, including type 2 diabetes, obesity and non-alcoholic fatty liver disease, have enormous impact on modern societies worldwide. Excess nutritional burden and nutri-stress together with sedentary lifestyles lead to these diseases. Deranged glucose, fat, and energy metabolism is at the center of nutri-stress, and glycolysis-derived glycerol-3-phosphate (Gro3P) is at the crossroads of these metabolic pathways. Cellular levels of Gro3P can be controlled by its synthesis, utilization or hydrolysis. The belief that mammalian cells do not possess an enzyme that hydrolyzes Gro3P, as in lower organisms and plants, is challenged by our recent work showing the presence of a Gro3P phosphatase (G3PP) in mammalian cells. A previously described phosphoglycolate phosphatase (PGP) in mammalian cells, with no established physiological function, has been shown to actually function as G3PP, under physiological conditions, particularly at elevated glucose levels. In the present review, we summarize evidence that supports the view that G3PP plays an important role in the regulation of gluconeogenesis and fat storage in hepatocytes, glucose stimulated insulin secretion and nutri-stress in β-cells, and lipogenesis in adipocytes. We provide a balanced perspective on the pathophysiological significance of G3PP in mammals with specific reference to cardiometabolic diseases.
Collapse
Affiliation(s)
- Elite Possik
- Departments of Nutrition, Biochemistry and Molecular Medicine, and Montreal Diabetes Research Center, CRCHUM, Montréal, QC, Canada
| | - Anfal Al-Mass
- Departments of Nutrition, Biochemistry and Molecular Medicine, and Montreal Diabetes Research Center, CRCHUM, Montréal, QC, Canada
- Department of Medicine, McGill University, Montréal, QC, Canada
| | - Marie-Line Peyot
- Departments of Nutrition, Biochemistry and Molecular Medicine, and Montreal Diabetes Research Center, CRCHUM, Montréal, QC, Canada
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fahd Al-Mulla
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman, Kuwait
| | - S. R. Murthy Madiraju
- Departments of Nutrition, Biochemistry and Molecular Medicine, and Montreal Diabetes Research Center, CRCHUM, Montréal, QC, Canada
- *Correspondence: Marc Prentki, ; S. R. Murthy Madiraju,
| | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular Medicine, and Montreal Diabetes Research Center, CRCHUM, Montréal, QC, Canada
- *Correspondence: Marc Prentki, ; S. R. Murthy Madiraju,
| |
Collapse
|
13
|
Poursharifi P, Attané C, Mugabo Y, Al-Mass A, Ghosh A, Schmitt C, Zhao S, Guida J, Lussier R, Erb H, Chenier I, Peyot ML, Joly E, Noll C, Carpentier AC, Madiraju SRM, Prentki M. Adipose ABHD6 regulates tolerance to cold and thermogenic programs. JCI Insight 2020; 5:140294. [PMID: 33201859 PMCID: PMC7819748 DOI: 10.1172/jci.insight.140294] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022] Open
Abstract
Enhanced energy expenditure in brown (BAT) and white adipose tissues (WAT) can be therapeutic against metabolic diseases. We examined the thermogenic role of adipose α/β-hydrolase domain 6 (ABHD6), which hydrolyzes monoacylglycerol (MAG), by employing adipose-specific ABHD6-KO mice. Control and KO mice showed similar phenotypes at room temperature and thermoneutral conditions. However, KO mice were resistant to hypothermia, which can be accounted for by the simultaneously increased lipolysis and lipogenesis of the thermogenic glycerolipid/free fatty acid (GL/FFA) cycle in visceral fat, despite unaltered uncoupling protein 1 expression. Upon cold stress, nuclear 2-MAG levels increased in visceral WAT of the KO mice. Evidence is provided that 2-MAG causes activation of PPARα in white adipocytes, leading to elevated expression and activity of GL/FFA cycle enzymes. In the ABHD6-ablated BAT, glucose and oxidative metabolism were elevated upon cold induction, without changes in GL/FFA cycle and lipid turnover. Moreover, response to in vivo β3-adrenergic stimulation was comparable between KO and control mice. Our data reveal a MAG/PPARα/GL/FFA cycling metabolic signaling network in visceral adipose tissue, which contributes to cold tolerance, and that adipose ABHD6 is a negative modulator of adaptive thermogenesis. Visceral adipose adipose α/β-hydrolase domain 6 regulates cold adaptation and acts as a brake for heat production via the regulation of thermogenic glycerolipid/free fatty acid cycling.
Collapse
Affiliation(s)
- Pegah Poursharifi
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Camille Attané
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yves Mugabo
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Anfal Al-Mass
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Anindya Ghosh
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Clémence Schmitt
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Shangang Zhao
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Julian Guida
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Roxane Lussier
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Heidi Erb
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Isabelle Chenier
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Marie-Line Peyot
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Erik Joly
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Christophe Noll
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - S R Murthy Madiraju
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Marc Prentki
- Departments of Nutrition, Biochemistry, and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| |
Collapse
|
14
|
Liu S, Promes JA, Harata M, Mishra A, Stephens SB, Taylor EB, Burand AJ, Sivitz WI, Fink BD, Ankrum JA, Imai Y. Adipose Triglyceride Lipase Is a Key Lipase for the Mobilization of Lipid Droplets in Human β-Cells and Critical for the Maintenance of Syntaxin 1a Levels in β-Cells. Diabetes 2020; 69:1178-1192. [PMID: 32312867 PMCID: PMC7243295 DOI: 10.2337/db19-0951] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 02/28/2020] [Indexed: 12/11/2022]
Abstract
Lipid droplets (LDs) are frequently increased when excessive lipid accumulation leads to cellular dysfunction. Distinct from mouse β-cells, LDs are prominent in human β-cells. However, the regulation of LD mobilization (lipolysis) in human β-cells remains unclear. We found that glucose increases lipolysis in nondiabetic human islets but not in islets in patients with type 2 diabetes (T2D), indicating dysregulation of lipolysis in T2D islets. Silencing adipose triglyceride lipase (ATGL) in human pseudoislets with shRNA targeting ATGL (shATGL) increased triglycerides (TGs) and the number and size of LDs, indicating that ATGL is the principal lipase in human β-cells. In shATGL pseudoislets, biphasic glucose-stimulated insulin secretion (GSIS), and insulin secretion to 3-isobutyl-1-methylxanthine and KCl were all reduced without altering oxygen consumption rate compared with scramble control. Like human islets, INS1 cells showed visible LDs, glucose-responsive lipolysis, and impairment of GSIS after ATGL silencing. ATGL-deficient INS1 cells and human pseudoislets showed reduced SNARE protein syntaxin 1a (STX1A), a key SNARE component. Proteasomal degradation of Stx1a was accelerated likely through reduced palmitoylation in ATGL-deficient INS1 cells. Therefore, ATGL is responsible for LD mobilization in human β-cells and supports insulin secretion by stabilizing STX1A. The dysregulated lipolysis may contribute to LD accumulation and β-cell dysfunction in T2D islets.
Collapse
Affiliation(s)
- Siming Liu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Joseph A Promes
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Mikako Harata
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Akansha Mishra
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Samuel B Stephens
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Eric B Taylor
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Anthony J Burand
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA
| | - William I Sivitz
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - Brian D Fink
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - James A Ankrum
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA
| | - Yumi Imai
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| |
Collapse
|
15
|
Trites MJ, Clugston RD. The role of adipose triglyceride lipase in lipid and glucose homeostasis: lessons from transgenic mice. Lipids Health Dis 2019; 18:204. [PMID: 31757217 PMCID: PMC6874817 DOI: 10.1186/s12944-019-1151-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022] Open
Abstract
The ability of mammals to store and draw on fat reserves has been a driving force throughout evolution in an environment with intermittent nutrient availability. The discovery of adipose triglyceride lipase (ATGL) as a triglyceride lipase provided a heightened understanding of the mechanisms governing mobilization of fat reserves from adipose tissue. ATGL catalyses the initial step in adipose triglyceride lipolysis, working in concert with other enzymes to mobilize triglyceride for energy production. In addition to the role of ATGL in adipose tissue triglyceride mobilization, ATGL plays crucial roles in regulating lipid homeostasis in other tissues. These roles have been characterized primarily using transgenic mice with tissue-specific ATGL ablation. For example, the global ATGL knockout induces a severe cardiac defect that results in premature mortality that is mimicked by inducible cardiomyocyte-specific ATGL knockout. Global- and adipose-specific ATGL ablation induces a whole-body shift from lipid metabolism to glucose metabolism to satisfy metabolic demand primarily facilitated by an increase in glucose uptake by skeletal muscle. Generation of liver-specific ATGL knockouts has implicated hepatic lipolysis as a critical component of normal liver function. Analysis of β-cell ATGL knockouts implicates the necessity of pancreatic ATGL in insulin secretion. The objective of this review is to discuss the contributions of ATGL to systemic lipid- and glucose-homeostasis discovered through the study of transgenic mice.
Collapse
Affiliation(s)
- Michael J Trites
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.,Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Robin D Clugston
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada. .,Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
16
|
Of mice and men: The physiological role of adipose triglyceride lipase (ATGL). Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:880-899. [PMID: 30367950 PMCID: PMC6439276 DOI: 10.1016/j.bbalip.2018.10.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
Abstract
Adipose triglyceride lipase (ATGL) has been discovered 14 years ago and revised our view on intracellular triglyceride (TG) mobilization – a process termed lipolysis. ATGL initiates the hydrolysis of TGs to release fatty acids (FAs) that are crucial energy substrates, precursors for the synthesis of membrane lipids, and ligands of nuclear receptors. Thus, ATGL is a key enzyme in whole-body energy homeostasis. In this review, we give an update on how ATGL is regulated on the transcriptional and post-transcriptional level and how this affects the enzymes' activity in the context of neutral lipid catabolism. In depth, we highlight and discuss the numerous physiological functions of ATGL in lipid and energy metabolism. Over more than a decade, different genetic mouse models lacking or overexpressing ATGL in a cell- or tissue-specific manner have been generated and characterized. Moreover, pharmacological studies became available due to the development of a specific murine ATGL inhibitor (Atglistatin®). The identification of patients with mutations in the human gene encoding ATGL and their disease spectrum has underpinned the importance of ATGL in humans. Together, mouse models and human data have advanced our understanding of the physiological role of ATGL in lipid and energy metabolism in adipose and non-adipose tissues, and of the pathophysiological consequences of ATGL dysfunction in mice and men. Summary of mouse models with genetic or pharmacological manipulation of ATGL. Summary of patients with mutations in the human gene encoding ATGL. In depth discussion of the role of ATGL in numerous physiological processes in mice and men.
Collapse
|
17
|
Zhang A, Li D, Liu Y, Li J, Zhang Y, Zhang CY. Islet β cell: An endocrine cell secreting miRNAs. Biochem Biophys Res Commun 2018; 495:1648-1654. [DOI: 10.1016/j.bbrc.2017.12.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/05/2017] [Indexed: 12/28/2022]
|
18
|
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: 6.5] [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
|
19
|
Mugabo Y, Zhao S, Lamontagne J, Al-Mass A, Peyot ML, Corkey BE, Joly E, Madiraju SRM, Prentki M. Metabolic fate of glucose and candidate signaling and excess-fuel detoxification pathways in pancreatic β-cells. J Biol Chem 2017; 292:7407-7422. [PMID: 28280244 DOI: 10.1074/jbc.m116.763060] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/06/2017] [Indexed: 12/28/2022] Open
Abstract
Glucose metabolism promotes insulin secretion in β-cells via metabolic coupling factors that are incompletely defined. Moreover, chronically elevated glucose causes β-cell dysfunction, but little is known about how cells handle excess fuels to avoid toxicity. Here we sought to determine which among the candidate pathways and coupling factors best correlates with glucose-stimulated insulin secretion (GSIS), define the fate of glucose in the β-cell, and identify pathways possibly involved in excess-fuel detoxification. We exposed isolated rat islets for 1 h to increasing glucose concentrations and measured various pathways and metabolites. Glucose oxidation, oxygen consumption, and ATP production correlated well with GSIS and saturated at 16 mm glucose. However, glucose utilization, glycerol release, triglyceride and glycogen contents, free fatty acid (FFA) content and release, and cholesterol and cholesterol esters increased linearly up to 25 mm glucose. Besides being oxidized, glucose was mainly metabolized via glycerol production and release and lipid synthesis (particularly FFA, triglycerides, and cholesterol), whereas glycogen production was comparatively low. Using targeted metabolomics in INS-1(832/13) cells, we found that several metabolites correlated well with GSIS, in particular some Krebs cycle intermediates, malonyl-CoA, and lower ADP levels. Glucose dose-dependently increased the dihydroxyacetone phosphate/glycerol 3-phosphate ratio in INS-1(832/13) cells, indicating a more oxidized state of NAD in the cytosol upon glucose stimulation. Overall, the data support a role for accelerated oxidative mitochondrial metabolism, anaplerosis, and malonyl-CoA/lipid signaling in β-cell metabolic signaling and suggest that a decrease in ADP levels is important in GSIS. The results also suggest that excess-fuel detoxification pathways in β-cells possibly comprise glycerol and FFA formation and release extracellularly and the diversion of glucose carbons to triglycerides and cholesterol esters.
Collapse
Affiliation(s)
- Yves Mugabo
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Departments of Nutrition, Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montréal, Montreal, Québec H3C 3J7, Canada, and
| | - Shangang Zhao
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, Québec H3A 1B1, Canada
| | - Julien Lamontagne
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Anfal Al-Mass
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Departments of Medicine and Human Genetics, McGill University, Montreal, Québec H3A 1B1, Canada
| | - Marie-Line Peyot
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Barbara E Corkey
- Department of Medicine, Obesity Research Center, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Erik Joly
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - S R Murthy Madiraju
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Marc Prentki
- From the Montreal Diabetes Research Center and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec H2X 0A9, Canada, .,Departments of Nutrition, Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montréal, Montreal, Québec H3C 3J7, Canada, and
| |
Collapse
|
20
|
Attané C, Peyot ML, Lussier R, Poursharifi P, Zhao S, Zhang D, Morin J, Pineda M, Wang S, Dumortier O, Ruderman NB, Mitchell GA, Simons B, Madiraju SRM, Joly E, Prentki M. A beta cell ATGL-lipolysis/adipose tissue axis controls energy homeostasis and body weight via insulin secretion in mice. Diabetologia 2016; 59:2654-2663. [PMID: 27677764 PMCID: PMC6518076 DOI: 10.1007/s00125-016-4105-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/15/2016] [Indexed: 12/02/2022]
Abstract
AIMS/HYPOTHESIS To directly assess the role of beta cell lipolysis in insulin secretion and whole-body energy homeostasis, inducible beta cell-specific adipose triglyceride lipase (ATGL)-deficient (B-Atgl-KO) mice were studied under normal diet (ND) and high-fat diet (HFD) conditions. METHODS Atgl flox/flox mice were cross-bred with Mip-Cre-ERT mice to generate Mip-Cre-ERT/+;Atgl flox/flox mice. At 8 weeks of age, these mice were injected with tamoxifen to induce deletion of beta cell-specific Atgl (also known as Pnpla2), and the mice were fed an ND or HFD. RESULTS ND-fed male B-Atgl-KO mice showed decreased insulinaemia and glucose-induced insulin secretion (GSIS) in vivo. Changes in GSIS correlated with the islet content of long-chain saturated monoacylglycerol (MAG) species that have been proposed to be metabolic coupling factors for insulin secretion. Exogenous MAGs restored GSIS in B-Atgl-KO islets. B-Atgl-KO male mice fed an HFD showed reduced insulinaemia, glycaemia in the fasted and fed states and after glucose challenge, as well as enhanced insulin sensitivity. Moreover, decreased insulinaemia in B-Atgl-KO mice was associated with increased energy expenditure, and lipid metabolism in brown (BAT) and white (WAT) adipose tissues, leading to reduced fat mass and body weight. CONCLUSIONS/INTERPRETATION ATGL in beta cells regulates insulin secretion via the production of signalling MAGs. Decreased insulinaemia due to lowered GSIS protects B-Atgl-KO mice from diet-induced obesity, improves insulin sensitivity, increases lipid mobilisation from WAT and causes BAT activation. The results support the concept that fuel excess can drive obesity and diabetes via hyperinsulinaemia, and that an islet beta cell ATGL-lipolysis/adipose tissue axis controls energy homeostasis and body weight via insulin secretion.
Collapse
Affiliation(s)
- Camille Attané
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Marie-Line Peyot
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Roxane Lussier
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Pegah Poursharifi
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Shangang Zhao
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
- UT Southwestern Medical Center, Dallas, TX, USA
| | - Dongwei Zhang
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
- Diabetes Research Center, Beijing University of Chinese Medicine, Beijing, China
| | - Johane Morin
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Marco Pineda
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Shupei Wang
- Division of Medical Genetics, Department of Pediatrics, Université de Montréal and CHU Sainte-Justine, Montreal, QC, Canada
| | - Olivier Dumortier
- University Nice Sophia Antipolis, Nice, France
- Inserm, U1081, Institute for Research on Cancer and Aging of Nice (IRCAN), Aging and Diabetes Team, Nice, France
- CNRS, UMR7284, IRCAN, Nice, France
| | - Neil B Ruderman
- Departments of Medicine and Physiology and Biophysics, Boston University School of Medicine, Boston, MA, USA
- Diabetes Unit, Boston Medical Center, Boston, MA, USA
| | - Grant A Mitchell
- Division of Medical Genetics, Department of Pediatrics, Université de Montréal and CHU Sainte-Justine, Montreal, QC, Canada
| | | | - S R Murthy Madiraju
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Erik Joly
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada
| | - Marc Prentki
- Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Room R08-412, Montreal, QC, H1W 4A4, Canada.
| |
Collapse
|
21
|
Mulder H. Metabolic coupling in pancreatic beta cells: lipolysis revisited. Diabetologia 2016; 59:2510-2513. [PMID: 27660005 DOI: 10.1007/s00125-016-4111-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 09/07/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Hindrik Mulder
- Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmö, Sweden.
| |
Collapse
|
22
|
Koppe L, Nyam E, Vivot K, Manning Fox JE, Dai XQ, Nguyen BN, Trudel D, Attané C, Moullé VS, MacDonald PE, Ghislain J, Poitout V. Urea impairs β cell glycolysis and insulin secretion in chronic kidney disease. J Clin Invest 2016; 126:3598-612. [PMID: 27525435 DOI: 10.1172/jci86181] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/24/2016] [Indexed: 12/25/2022] Open
Abstract
Disorders of glucose homeostasis are common in chronic kidney disease (CKD) and are associated with increased mortality, but the mechanisms of impaired insulin secretion in this disease remain unclear. Here, we tested the hypothesis that defective insulin secretion in CKD is caused by a direct effect of urea on pancreatic β cells. In a murine model in which CKD is induced by 5/6 nephrectomy (CKD mice), we observed defects in glucose-stimulated insulin secretion in vivo and in isolated islets. Similarly, insulin secretion was impaired in normal mouse and human islets that were cultured with disease-relevant concentrations of urea and in islets from normal mice treated orally with urea for 3 weeks. In CKD mouse islets as well as urea-exposed normal islets, we observed an increase in oxidative stress and protein O-GlcNAcylation. Protein O-GlcNAcylation was also observed in pancreatic sections from CKD patients. Impairment of insulin secretion in both CKD mouse and urea-exposed islets was associated with reduced glucose utilization and activity of phosphofructokinase 1 (PFK-1), which could be reversed by inhibiting O-GlcNAcylation. Inhibition of O-GlcNAcylation also restored insulin secretion in both mouse models. These results suggest that insulin secretory defects associated with CKD arise from elevated circulating levels of urea that increase islet protein O-GlcNAcylation and impair glycolysis.
Collapse
|
23
|
Attané C, Peyot ML, Lussier R, Zhang D, Joly E, Madiraju SRM, Prentki M. Differential Insulin Secretion of High-Fat Diet-Fed C57BL/6NN and C57BL/6NJ Mice: Implications of Mixed Genetic Background in Metabolic Studies. PLoS One 2016; 11:e0159165. [PMID: 27403868 PMCID: PMC4942110 DOI: 10.1371/journal.pone.0159165] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/28/2016] [Indexed: 01/02/2023] Open
Abstract
Many metabolic studies employ tissue-specific gene knockout mice, which requires breeding of floxed gene mice, available mostly on C57BL/6N (NN) genetic background, with cre or Flp recombinase-expressing mice, available on C57BL/6J (JJ) background, resulting in the generation of mixed C57BL/6NJ (NJ) genetic background mice. Recent awareness of many genetic differences between NN and JJ strains including the deletion of nicotinamide nucleotide transhydrogenase (nnt), necessitates examination of the consequence of mixed NJ background on glucose tolerance, beta cell function and other metabolic parameters. Male mice with NN and NJ genetic background were fed with normal or high fat diets (HFD) for 12 weeks and glucose and insulin homeostasis were studied. Genotype had no effect on body weight and food intake in mice fed normal or high fat diets. Insulinemia in the fed and fasted states and after a glucose challenge was lower in HFD-fed NJ mice, even though their glycemia and insulin sensitivity were similar to NN mice. NJ mice showed mild glucose intolerance. Moreover, glucose- but not KCl-stimulated insulin secretion in isolated islets was decreased in HFD-fed NJ vs NN mice without changes in insulin content and beta cell mass. Under normal diet, besides reduced fed insulinemia, NN and NJ mice presented similar metabolic parameters. However, HFD-fed NJ mice displayed lower fed and fasted insulinemia and glucose-induced insulin secretion in vivo and ex vivo, as compared to NN mice. These results strongly caution against using unmatched mixed genetic background C57BL/6 mice for comparisons, particularly under HFD conditions.
Collapse
Affiliation(s)
- Camille Attané
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Marie-Line Peyot
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Roxane Lussier
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Dongwei Zhang
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Erik Joly
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - S. R. Murthy Madiraju
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
| | - Marc Prentki
- Departments of Nutrition and Biochemistry, Montreal Diabetes Research Center, CRCHUM and Université de Montréal, Montréal, QC, Canada
- * E-mail:
| |
Collapse
|
24
|
Pearson GL, Mellett N, Chu KY, Boslem E, Meikle PJ, Biden TJ. A comprehensive lipidomic screen of pancreatic β-cells using mass spectroscopy defines novel features of glucose-stimulated turnover of neutral lipids, sphingolipids and plasmalogens. Mol Metab 2016; 5:404-414. [PMID: 27257600 PMCID: PMC4877660 DOI: 10.1016/j.molmet.2016.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 01/05/2023] Open
Abstract
Objective Glucose promotes lipid remodelling in pancreatic β-cells, and this is thought to contribute to the regulation of insulin secretion, but the metabolic pathways and potential signalling intermediates have not been fully elaborated. Methods Using mass spectrometry (MS) we quantified changes in approximately 300 lipid metabolites in MIN6 β-cells and isolated mouse islets following 1 h stimulation with glucose. Flux through sphingolipid pathways was also assessed in 3H-sphinganine-labelled cells using TLC. Results Glucose specifically activates the conversion of triacylglycerol (TAG) to diacylglycerol (DAG). This leads indirectly to the formation of 18:1 monoacylglycerol (MAG), via degradation of saturated/monounsaturated DAG species, such as 16:0_18:1 DAG, which are the most abundant, immediate products of glucose-stimulated TAG hydrolysis. However, 16:0-containing, di-saturated DAG species are a better direct marker of TAG hydrolysis since, unlike the 18:1-containing DAGs, they are predominately formed via this route. Using multiple reaction monitoring, we confirmed that in islets under basal conditions, 18:1 MAG is the most abundant species. We further demonstrated a novel site of glucose to enhance the conversion of ceramide to sphingomyelin (SM) and galactosylceramide (GalCer). Flux and product:precursor analyses suggest regulation of the enzyme SM synthase, which would constitute a separate mechanism for localized generation of DAG in response to glucose. Phosphatidylcholine (PC) plasmalogen (P) species, specifically those containing 20:4, 22:5 and 22:6 side chains, were also diminished in the presence of glucose, whereas the more abundant phosphatidylethanolamine plasmalogens were unchanged. Conclusion Our results highlight 18:1 MAG, GalCer, PC(P) and DAG/SM as potential contributors to metabolic stimulus-secretion coupling. Using mass spectroscopy lipidomics we have defined new aspects of glucose simulated lipid turnover in pancreatic beta cells. Glucose directly stimulates triacylglycerol hydrolysis, of which di-saturated diacylglycerol species are excellent markers. C18:1 is the most abundant monacylglycerol, and the one most obviously increased by glucose. Phosphatidylcholine plasmalogens with poly-unsaturated side chains are preferentially decreased by glucose. Glucose specifically enhances the conversion of ceramide to both sphingomyelin and galactosylceramide.
Collapse
Key Words
- (O), ether lipid
- (P), plasmalogen
- ATGL, adipose tissue glycerolipase
- CE, cholesterol ester
- COH, free cholesterol
- Ceramide
- DAG, diacylglycerol
- Diacylglycerol
- FA, fatty acid
- GSIS, glucose-stimulated insulin secretion
- GalCer, galactosylceramide
- GluCer, glucosylceramide
- HSL, hormone sensitive lipase
- Insulin secretion
- KRHB, Krebs Ringer Hepes Buffer
- MAG, monacylglycerol
- MHC, monohexosylceramide
- MS, mass spectrometry
- Monacylglycerol
- PC, phosphatidylcholine
- PE, phosphatidylethanolamine
- PI, phosphatidylinositol
- PKD, protein kinase D
- PLA2, phospholipase A2
- Pancreatic β-cell
- Plasmalogen
- SM, sphingomyelin
- TAG, triacylglycerol
Collapse
Affiliation(s)
- Gemma L Pearson
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Australia
| | - Natalie Mellett
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Kwan Yi Chu
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
| | - Ebru Boslem
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Australia
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia.
| | - Trevor J Biden
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Australia.
| |
Collapse
|
25
|
Pepin É, Al-Mass A, Attané C, Zhang K, Lamontagne J, Lussier R, Madiraju SRM, Joly E, Ruderman NB, Sladek R, Prentki M, Peyot ML. Pancreatic β-Cell Dysfunction in Diet-Induced Obese Mice: Roles of AMP-Kinase, Protein Kinase Cε, Mitochondrial and Cholesterol Metabolism, and Alterations in Gene Expression. PLoS One 2016; 11:e0153017. [PMID: 27043434 PMCID: PMC4820227 DOI: 10.1371/journal.pone.0153017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/22/2016] [Indexed: 12/27/2022] Open
Abstract
Diet induced obese (DIO) mice can be stratified according to their weight gain in response to high fat diet as low responders (LDR) and high responders (HDR). This allows the study of β-cell failure and the transitions to prediabetes (LDR) and early diabetes (HDR). C57BL/6N mice were fed for 8 weeks with a normal chow diet (ND) or a high fat diet and stratified as LDR and HDR. Freshly isolated islets from ND, LDR and HDR mice were studied ex-vivo for mitochondrial metabolism, AMPK activity and signalling, the expression and activity of key enzymes of energy metabolism, cholesterol synthesis, and mRNA profiling. Severely compromised glucose-induced insulin secretion in HDR islets, as compared to ND and LDR islets, was associated with suppressed AMP-kinase activity. HDR islets also showed reduced acetyl-CoA carboxylase activity and enhanced activity of 3-hydroxy-3-methylglutaryl-CoA reductase, which led respectively to elevated fatty acid oxidation and increased cholesterol biosynthesis. HDR islets also displayed mitochondrial membrane hyperpolarization and reduced ATP turnover in the presence of elevated glucose. Expression of protein kinase Cε, which reduces both lipolysis and production of signals for insulin secretion, was elevated in DIO islets. Genes whose expression increased or decreased by more than 1.2-fold were minor between LDR and ND islets (17 differentially expressed), but were prominent between HDR and ND islets (1508 differentially expressed). In HDR islets, particularly affected genes were related to cell cycle and proliferation, AMPK signaling, mitochondrial metabolism and cholesterol metabolism. In conclusion, chronically reduced AMPK activity, mitochondrial dysfunction, elevated cholesterol biosynthesis in islets, and substantial alterations in gene expression accompany β-cell failure in HDR islets. The β-cell compensation process in the prediabetic state (LDR) is largely independent of transcriptional adaptive changes, whereas the transition to early diabetes (HDR) is associated with major alterations in gene expression.
Collapse
Affiliation(s)
- Émilie Pepin
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Anfal Al-Mass
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Camille Attané
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Kezhuo Zhang
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Julien Lamontagne
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Roxane Lussier
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - S. R. Murthy Madiraju
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Erik Joly
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Neil B. Ruderman
- Departments of Medicine and Physiology and Biophysics, Boston University School of Medicine and Diabetes Unit, Boston Medical Center, Boston, MA, United States of America
| | - Robert Sladek
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Marc Prentki
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- Departments of Nutrition, Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montréal, Montreal, Québec, Canada
- * E-mail: (MP); (MLP)
| | - Marie-Line Peyot
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- * E-mail: (MP); (MLP)
| |
Collapse
|
26
|
Brun T, Maechler P. Beta-cell mitochondrial carriers and the diabetogenic stress response. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2540-9. [PMID: 26979549 DOI: 10.1016/j.bbamcr.2016.03.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 01/09/2023]
Abstract
Mitochondria play a central role in pancreatic beta-cells by coupling metabolism of the secretagogue glucose to distal events of regulated insulin exocytosis. This process requires transports of both metabolites and nucleotides in and out of the mitochondria. The molecular identification of mitochondrial carriers and their respective contribution to beta-cell function have been uncovered only recently. In type 2 diabetes, mitochondrial dysfunction is an early event and may precipitate beta-cell loss. Under diabetogenic conditions, characterized by glucotoxicity and lipotoxicity, the expression profile of mitochondrial carriers is selectively modified. This review describes the role of mitochondrial carriers in beta-cells and the selective changes in response to glucolipotoxicity. In particular, we discuss the importance of the transfer of metabolites (pyruvate, citrate, malate, and glutamate) and nucleotides (ATP, NADH, NADPH) for beta-cell function and dysfunction. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Thierry Brun
- Department of Cell Physiology and Metabolism, Faculty Diabetes Center, Geneva University Medical Centre, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, Faculty Diabetes Center, Geneva University Medical Centre, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland.
| |
Collapse
|
27
|
Identification of a mammalian glycerol-3-phosphate phosphatase: Role in metabolism and signaling in pancreatic β-cells and hepatocytes. Proc Natl Acad Sci U S A 2016; 113:E430-9. [PMID: 26755581 DOI: 10.1073/pnas.1514375113] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Obesity, and the associated disturbed glycerolipid/fatty acid (GL/FA) cycle, contribute to insulin resistance, islet β-cell failure, and type 2 diabetes. Flux through the GL/FA cycle is regulated by the availability of glycerol-3-phosphate (Gro3P) and fatty acyl-CoA. We describe here a mammalian Gro3P phosphatase (G3PP), which was not known to exist in mammalian cells, that can directly hydrolyze Gro3P to glycerol. We identified that mammalian phosphoglycolate phosphatase, with an uncertain function, acts in fact as a G3PP. We found that G3PP, by controlling Gro3P levels, regulates glycolysis and glucose oxidation, cellular redox and ATP production, gluconeogenesis, glycerolipid synthesis, and fatty acid oxidation in pancreatic islet β-cells and hepatocytes, and that glucose stimulated insulin secretion and the response to metabolic stress, e.g., glucolipotoxicity, in β-cells. In vivo overexpression of G3PP in rat liver lowers body weight gain and hepatic glucose production from glycerol and elevates plasma HDL levels. G3PP is expressed at various levels in different tissues, and its expression varies according to the nutritional state in some tissues. As Gro3P lies at the crossroads of glucose, lipid, and energy metabolism, control of its availability by G3PP adds a key level of metabolic regulation in mammalian cells, and G3PP offers a potential target for type 2 diabetes and cardiometabolic disorders.
Collapse
|
28
|
Morigny P, Houssier M, Mouisel E, Langin D. Adipocyte lipolysis and insulin resistance. Biochimie 2015; 125:259-66. [PMID: 26542285 DOI: 10.1016/j.biochi.2015.10.024] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/30/2015] [Indexed: 12/15/2022]
Abstract
Obesity-induced insulin resistance is a major risk factor for the development of type 2 diabetes. Basal fat cell lipolysis (i.e., fat cell triacylglycerol breakdown into fatty acids and glycerol in the absence of stimulatory factors) is elevated during obesity and is closely associated with insulin resistance. Inhibition of adipocyte lipolysis may therefore be a promising therapeutic strategy for treating insulin resistance and preventing obesity-associated type 2 diabetes. In this review, we explore the relationship between adipose lipolysis and insulin sensitivity. After providing an overview of the components of fat cell lipolytic machinery, we describe the hypotheses that may support the causality between lipolysis and insulin resistance. Excessive circulating fatty acids may ectopically accumulate in insulin-sensitive tissues and impair insulin action. Increased basal lipolysis may also modify the secretory profile of adipose tissue, influencing whole body insulin sensitivity. Finally, excessive fatty acid release may also worsen adipose tissue inflammation, a well-known parameter contributing to insulin resistance. Partial genetic or pharmacologic inhibition of fat cell lipases in mice as well as short term clinical trials using antilipolytic drugs in humans support the benefit of fat cell lipolysis inhibition on systemic insulin sensitivity and glucose metabolism, which occurs without an increase of fat mass. Modulation of fatty acid fluxes and, putatively, of fat cell secretory pattern may explain the amelioration of insulin sensitivity whereas changes in adipose tissue immune response do not seem involved.
Collapse
Affiliation(s)
- Pauline Morigny
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France
| | - Marianne Houssier
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France
| | - Etienne Mouisel
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France
| | - Dominique Langin
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, France; Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France.
| |
Collapse
|
29
|
Zhao S, Poursharifi P, Mugabo Y, Levens EJ, Vivot K, Attane C, Iglesias J, Peyot ML, Joly E, Madiraju SM, Prentki M. α/β-Hydrolase domain-6 and saturated long chain monoacylglycerol regulate insulin secretion promoted by both fuel and non-fuel stimuli. Mol Metab 2015; 4:940-50. [PMID: 26909310 PMCID: PMC4731734 DOI: 10.1016/j.molmet.2015.09.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 09/21/2015] [Accepted: 09/28/2015] [Indexed: 01/15/2023] Open
Abstract
Objective α/β-Hydrolase domain-6 (ABHD6) is a newly identified monoacylglycerol (MAG) lipase. We recently reported that it negatively regulates glucose stimulated insulin secretion (GSIS) in the β cells by hydrolyzing lipolysis-derived MAG that acts as a metabolic coupling factor and signaling molecule via exocytotic regulator Munc13-1. Whether ABHD6 and MAG play a role in response to all classes of insulin secretagogues, in particular various fuel and non-fuel stimuli, is unknown. Methods Insulin secretion in response to various classes of secretagogues, exogenous MAG and pharmacological agents was measured in islets of mice deficient in ABHD6 specifically in the β cell (BKO). Islet perifusion experiments and determinations of glucose and fatty acid metabolism, cytosolic Ca2+ and MAG species levels were carried out. Results Deletion of ABHD6 potentiated insulin secretion in response to the fuels glutamine plus leucine and α-ketoisocaproate and to the non-fuel stimuli glucagon-like peptide 1, carbamylcholine and elevated KCl. Fatty acids amplified GSIS in control and BKO mice to the same extent. Exogenous 1-MAG amplified insulin secretion in response to fuel and non-fuel stimuli. MAG hydrolysis activity was greatly reduced in BKO islets without changes in total diacylglycerol and triacylglycerol lipase activity. ABHD6 deletion induced insulin secretion independently from KATP channels and did not alter the glucose induced rise in intracellular Ca2+. Perifusion studies showed elevated insulin secretion during second phase of GSIS in BKO islets that was not due to altered cytosolic Ca2+ signaling or because of changes in glucose and fatty acid metabolism. Glucose increased islet saturated long chain 1-MAG species and ABHD6 deletion caused accumulation of these 1-MAG species at both low and elevated glucose. Conclusion ABHD6 regulates insulin secretion in response to fuel stimuli at large and some non-fuel stimuli by controlling long chain saturated 1-MAG levels that synergize with other signaling pathways for secretion. ABHD6 is the major monoacylglycerol (MAG) hydrolase in pancreatic β cells. 1-MAG level is elevated in islets from β cell specific ABHD6-KO mice (BKO). BKO islets show enhanced fuel and non-fuel induced insulin secretion. ABHD6 accessible 1-MAG synergizes with other signals for insulin secretion.
Collapse
Key Words
- 1-OG, 1-oleoylglycerol
- 1-PG, 1-palmitoylglycerol
- 1-SG, 1-stearoylglycerol
- ABHD6, α/β-hydrolase domain-6
- ATGL, adipose triglyceride lipase
- BKO, β cell specific ABHD6-knockout
- Carb, carbamylcholine
- Cytosolic Ca2+
- DAG, diacylglycerol
- FFA, free fatty acid
- Flox, flox/flox
- GL/FFA, glycerolipid/ free fatty acid
- GLP1, glucagon-like peptide 1
- GPCR, G-protein coupled receptor
- GSIS, glucose stimulated insulin secretion
- HSL, hormone sensitive lipase
- Insulin secretion
- KO, knockout
- Kic, α-ketoisocaproate
- MAG, monoacylglycerol
- Monoacylglycerol
- OGTT, oral glucose tolerance test
- Pancreatic islets
- ROS, reactive oxygen species
- TG, triacylglycerol
- WT, wild type
- α/β-Hydrolase domain-6
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - S.R. Murthy Madiraju
- Corresponding author. Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Rm R08-414, Montreal, QC H1W 4A4, Canada. Tel.: +1 514 890 8000x23610; fax: +1 514 412 7648.
| | - Marc Prentki
- Corresponding author. Montreal Diabetes Research Center, CRCHUM, 900 St-Denis (Viger Tower), Rm R08-412, Montreal, QC H1W 4A4, Canada. Tel.: +1 514 890 8000x23642; fax: +1 514 412 7648.
| |
Collapse
|
30
|
Iglesias J, Lamontagne J, Erb H, Gezzar S, Zhao S, Joly E, Truong VL, Skorey K, Crane S, Madiraju SRM, Prentki M. Simplified assays of lipolysis enzymes for drug discovery and specificity assessment of known inhibitors. J Lipid Res 2015; 57:131-41. [PMID: 26423520 DOI: 10.1194/jlr.d058438] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Indexed: 12/25/2022] Open
Abstract
Lipids are used as cellular building blocks and condensed energy stores and also act as signaling molecules. The glycerolipid/ fatty acid cycle, encompassing lipolysis and lipogenesis, generates many lipid signals. Reliable procedures are not available for measuring activities of several lipolytic enzymes for the purposes of drug screening, and this resulted in questionable selectivity of various known lipase inhibitors. We now describe simple assays for lipolytic enzymes, including adipose triglyceride lipase (ATGL), hormone sensitive lipase (HSL), sn-1-diacylglycerol lipase (DAGL), monoacylglycerol lipase, α/β-hydrolase domain 6, and carboxylesterase 1 (CES1) using recombinant human and mouse enzymes either in cell extracts or using purified enzymes. We observed that many of the reported inhibitors lack specificity. Thus, Cay10499 (HSL inhibitor) and RHC20867 (DAGL inhibitor) also inhibit other lipases. Marked differences in the inhibitor sensitivities of human ATGL and HSL compared with the corresponding mouse enzymes was noticed. Thus, ATGListatin inhibited mouse ATGL but not human ATGL, and the HSL inhibitors WWL11 and Compound 13f were effective against mouse enzyme but much less potent against human enzyme. Many of these lipase inhibitors also inhibited human CES1. Results describe reliable assays for measuring lipase activities that are amenable for drug screening and also caution about the specificity of the many earlier described lipase inhibitors.
Collapse
Affiliation(s)
- Jose Iglesias
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Julien Lamontagne
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Heidi Erb
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Sari Gezzar
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Shangang Zhao
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Erik Joly
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | | | | | - Sheldon Crane
- NuChem Therapeutics, Montréal, Québec, Canada, H4P 2R2
| | - S R Murthy Madiraju
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| | - Marc Prentki
- Molecular Nutrition Unit and Montreal Diabetes Research Center, CRCHUM, Montréal, Québec, Canada H2X 0A9 Departments of Nutrition, Biochemistry, and Molecular Medicine, Université de Montréal, Montréal, Québec, Canada H2X 0A9
| |
Collapse
|
31
|
Oropeza D, Jouvet N, Bouyakdan K, Perron G, Ringuette LJ, Philipson LH, Kiss RS, Poitout V, Alquier T, Estall JL. PGC-1 coactivators in β-cells regulate lipid metabolism and are essential for insulin secretion coupled to fatty acids. Mol Metab 2015; 4:811-22. [PMID: 26629405 PMCID: PMC4632114 DOI: 10.1016/j.molmet.2015.08.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 07/30/2015] [Accepted: 08/05/2015] [Indexed: 11/30/2022] Open
Abstract
Objectives Peroxisome proliferator-activated receptor γ coactivator 1 (PPARGCA1, PGC-1) transcriptional coactivators control gene programs important for nutrient metabolism. Islets of type 2 diabetic subjects have reduced PGC-1α expression and this is associated with decreased insulin secretion, yet little is known about why this occurs or what role it plays in the development of diabetes. Our goal was to delineate the role and importance of PGC-1 proteins to β-cell function and energy homeostasis. Methods We investigated how nutrient signals regulate coactivator expression in islets and the metabolic consequences of reduced PGC-1α and PGC-1β in primary and cultured β-cells. Mice with inducible β-cell specific double knockout of Pgc-1α/Pgc-1β (βPgc-1 KO) were created to determine the physiological impact of reduced Pgc1 expression on glucose homeostasis. Results Pgc-1α and Pgc-1β expression was increased in primary mouse and human islets by acute glucose and palmitate exposure. Surprisingly, PGC-1 proteins were dispensable for the maintenance of mitochondrial mass, gene expression, and oxygen consumption in response to glucose in adult β-cells. However, islets and mice with an inducible, β-cell-specific PGC-1 knockout had decreased insulin secretion due in large part to loss of the potentiating effect of fatty acids. Consistent with an essential role for PGC-1 in lipid metabolism, β-cells with reduced PGC-1s accumulated acyl-glycerols and PGC-1s controlled expression of key enzymes in lipolysis and the glycerolipid/free fatty acid cycle. Conclusions These data highlight the importance of PGC-1s in coupling β-cell lipid metabolism to promote efficient insulin secretion. Loss of Pgc-1s in adult β-cells decreases insulin secretion in response to glucose/palmitate. Pgc-1α/β is not required to maintain basal mitochondrial mass or oxidative capacity in mature β-cells. Pgc-1α/β regulates expression of the lipolytic enzymes HSL and ATGL in β-cells. Reduced β-cell Pgc-1 causes accumulation of intracellular acyl-glycerols and cholesterol esters.
Collapse
Affiliation(s)
- Daniel Oropeza
- Laboratory of Molecular Mechanisms of Diabetes, Institut de Recherches Cliniques de Montreal (IRCM), 110 Ave des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada ; Department of Anatomy and Cell Biology, McGill University, 845 Rue Sherbrooke Ouest, Montreal, Quebec, H3A 0G4, Canada
| | - Nathalie Jouvet
- Laboratory of Molecular Mechanisms of Diabetes, Institut de Recherches Cliniques de Montreal (IRCM), 110 Ave des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada ; Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Khalil Bouyakdan
- Montreal Diabetes Research Center, CRCHUM, Department of Medicine, University of Montreal, 2900 Boulevard Edouard-Montpetit, Montreal, Quebec, H3T 1J4, Canada
| | - Gabrielle Perron
- Department of Anatomy and Cell Biology, McGill University, 845 Rue Sherbrooke Ouest, Montreal, Quebec, H3A 0G4, Canada
| | - Lea-Jeanne Ringuette
- Department of Anatomy and Cell Biology, McGill University, 845 Rue Sherbrooke Ouest, Montreal, Quebec, H3A 0G4, Canada
| | - Louis H Philipson
- Department of Medicine, University of Chicago, 5801 South Ellis Avenue, Chicago, IL, USA
| | - Robert S Kiss
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center, CRCHUM, Department of Medicine, University of Montreal, 2900 Boulevard Edouard-Montpetit, Montreal, Quebec, H3T 1J4, Canada
| | - Thierry Alquier
- Montreal Diabetes Research Center, CRCHUM, Department of Medicine, University of Montreal, 2900 Boulevard Edouard-Montpetit, Montreal, Quebec, H3T 1J4, Canada
| | - Jennifer L Estall
- Laboratory of Molecular Mechanisms of Diabetes, Institut de Recherches Cliniques de Montreal (IRCM), 110 Ave des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada ; Department of Anatomy and Cell Biology, McGill University, 845 Rue Sherbrooke Ouest, Montreal, Quebec, H3A 0G4, Canada ; Montreal Diabetes Research Center, CRCHUM, Department of Medicine, University of Montreal, 2900 Boulevard Edouard-Montpetit, Montreal, Quebec, H3T 1J4, Canada
| |
Collapse
|
32
|
Ramanadham S, Ali T, Ashley JW, Bone RN, Hancock WD, Lei X. Calcium-independent phospholipases A2 and their roles in biological processes and diseases. J Lipid Res 2015; 56:1643-68. [PMID: 26023050 DOI: 10.1194/jlr.r058701] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Indexed: 12/24/2022] Open
Abstract
Among the family of phospholipases A2 (PLA2s) are the Ca(2+)-independent PLA2s (iPLA2s) and they are designated group VI iPLA2s. In relation to secretory and cytosolic PLA2s, the iPLA2s are more recently described and details of their expression and roles in biological functions are rapidly emerging. The iPLA2s or patatin-like phospholipases (PNPLAs) are intracellular enzymes that do not require Ca(2+) for activity, and contain lipase (GXSXG) and nucleotide-binding (GXGXXG) consensus sequences. Though nine PNPLAs have been recognized, PNPLA8 (membrane-associated iPLA2γ) and PNPLA9 (cytosol-associated iPLA2β) are the most widely studied and understood. The iPLA2s manifest a variety of activities in addition to phospholipase, are ubiquitously expressed, and participate in a multitude of biological processes, including fat catabolism, cell differentiation, maintenance of mitochondrial integrity, phospholipid remodeling, cell proliferation, signal transduction, and cell death. As might be expected, increased or decreased expression of iPLA2s can have profound effects on the metabolic state, CNS function, cardiovascular performance, and cell survival; therefore, dysregulation of iPLA2s can be a critical factor in the development of many diseases. This review is aimed at providing a general framework of the current understanding of the iPLA2s and discussion of the potential mechanisms of action of the iPLA2s and related involved lipid mediators.
Collapse
Affiliation(s)
- Sasanka Ramanadham
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Tomader Ali
- Undergraduate Research Office, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jason W Ashley
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Robert N Bone
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - William D Hancock
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Xiaoyong Lei
- Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
33
|
Trevino MB, Machida Y, Hallinger DR, Garcia E, Christensen A, Dutta S, Peake DA, Ikeda Y, Imai Y. Perilipin 5 regulates islet lipid metabolism and insulin secretion in a cAMP-dependent manner: implication of its role in the postprandial insulin secretion. Diabetes 2015; 64:1299-310. [PMID: 25392244 PMCID: PMC4375085 DOI: 10.2337/db14-0559] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Elevation of circulating fatty acids (FA) during fasting supports postprandial (PP) insulin secretion that is critical for glucose homeostasis and is impaired in diabetes. We tested our hypothesis that lipid droplet (LD) protein perilipin 5 (PLIN5) in β-cells aids PP insulin secretion by regulating intracellular lipid metabolism. We demonstrated that PLIN5 serves as an LD protein in human islets. In vivo, Plin5 and triglycerides were increased by fasting in mouse islets. MIN6 cells expressing PLIN5 (adenovirus [Ad]-PLIN5) and those expressing perilipin 2 (PLIN2) (Ad-PLIN2) had higher [(3)H]FA incorporation into triglycerides than Ad-GFP control, which support their roles as LD proteins. However, Ad-PLIN5 cells had higher lipolysis than Ad-PLIN2 cells, which increased further by 8-Br-cAMP, indicating that PLIN5 facilitates FA mobilization upon cAMP stimulation as seen postprandially. Ad-PLIN5 in islets enhanced the augmentation of glucose-stimulated insulin secretion by FA and 8-Br-cAMP in G-protein-coupled receptor 40 (GPR40)- and cAMP-activated protein kinase-dependent manners, respectively. When PLIN5 was increased in mouse β-cells in vivo, glucose tolerance after an acute exenatide challenge was improved. Therefore, the elevation of islet PLIN5 during fasting allows partitioning of FA into LD that is released upon refeeding to support PP insulin secretion in cAMP- and GPR40-dependent manners.
Collapse
Affiliation(s)
- Michelle B Trevino
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA
| | - Yui Machida
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA
| | - Daniel R Hallinger
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA
| | - Eden Garcia
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA
| | - Aaron Christensen
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA
| | - Sucharita Dutta
- Leroy T. Canoles Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA
| | | | - Yasuhiro Ikeda
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN
| | - Yumi Imai
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA
| |
Collapse
|
34
|
Takei M, Komatsu M, Aizawa T. Intracellular free fatty acid upholds β-cell glucose competence: The role of peroxisome proliferator-activated receptor δ and mitochondrial metabolism. J Diabetes Investig 2015; 6:133-6. [PMID: 25802719 PMCID: PMC4364846 DOI: 10.1111/jdi.12257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 11/30/2022] Open
Affiliation(s)
- Masahiro Takei
- Division of Diabetes Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine Matsumoto, Japan
| | - Mitsuhisa Komatsu
- Division of Diabetes Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine Matsumoto, Japan
| | - Toru Aizawa
- Diabetes Center, Aizawa Hospital Matsumoto, Japan
| |
Collapse
|
35
|
Cai Y, Lydic TA, Turkette T, Reid GE, Olson LK. Impact of alogliptin and pioglitazone on lipid metabolism in islets of prediabetic and diabetic Zucker Diabetic Fatty rats. Biochem Pharmacol 2015; 95:46-57. [PMID: 25801003 DOI: 10.1016/j.bcp.2015.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/13/2015] [Indexed: 12/30/2022]
Abstract
Prolonged exposure of pancreatic beta (β) cells to elevated glucose and free fatty acids (FFA) as occurs in type 2 diabetes results in loss of β cell function and survival. In Zucker Diabetic Fatty (ZDF) rats, β cell failure is associated with increased triacylglyceride (TAG) synthesis and disruption of the glycerolipid/FFA (GL/FFA) cycle, a critical arm of glucose-stimulated insulin secretion (GSIS). The aim of this study was to determine the impact of activation of PPARγ and increased incretin action via dipeptidyl-peptidase inhibition using pioglitazone and/or alogliptin, respectively, on islet lipid metabolism in prediabetic and diabetic ZDF rats. Transition of control prediabetic ZDF rats to diabetes was associated with reduced plasma insulin levels, reduced islet insulin content and GSIS, reduced stearoyl-CoA desaturase 2 (SCD 2) expression, and increased islet TAG, diacylglyceride (DAG) and ceramides species containing saturated FA. Treatment of prediabetic ZDF rats with a combination of pioglitazone and alogliptin, but not individually, prevented the transition to diabetes and was associated with marked lowering of islet TAG and DAG levels. Pioglitazone and alogliptin, however, did not restore SCD2 expression, the degree of FA saturation in TAG, DAG or ceramides, islet insulin content, or lower ceramide levels. These findings are consistent with activation of PPARγ and increased incretin action working in concert to restore GL/FFA cycle in β cells of ZDF rats. Restoration of the GL/FFA cycle without correcting islet FA desaturation, production of islet ceramides, and/or insulin sensitivity, however, may place these islets at risk for β cell failure.
Collapse
Affiliation(s)
- Ying Cai
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| | - Todd A Lydic
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA.
| | - Thomas Turkette
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| | - Gavin E Reid
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Department of Chemistry, Michigan State University, East Lansing, MI 48824 USA.
| | - L Karl Olson
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
36
|
Lord CC, Brown JM. Distinct roles for alpha-beta hydrolase domain 5 (ABHD5/CGI-58) and adipose triglyceride lipase (ATGL/PNPLA2) in lipid metabolism and signaling. Adipocyte 2014; 1:123-131. [PMID: 23145367 PMCID: PMC3492958 DOI: 10.4161/adip.20035] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Catabolism of stored triacylglycerol (TAG) from cytoplasmic lipid droplets is critical for providing energy substrates, membrane building blocks and signaling lipids in most cells of the body. However, the lipolytic machinery dictating TAG hydrolysis varies greatly among different cell types. Within the adipocyte, TAG hydrolysis is dynamically regulated by hormones to ensure appropriate metabolic adaptation to nutritional and physiologic cues. In other cell types such as hepatocytes, myocytes and macrophages, mobilization of stored TAG is regulated quite differently. Within the last decade, mutations in two key genes involved in TAG hydrolysis, α-β hydrolase domain 5 (ABHD5/CGI-58) and adipose triglyceride lipase (ATGL/PNPLA2), were found to cause two distinct neutral lipid storage diseases (NLSD) in humans. These genetic links, along with supporting evidence in mouse models, have prompted a number of studies surrounding the biochemical function(s) of these proteins. Although both CGI-58 and ATGL have been clearly implicated in TAG hydrolysis in multiple tissues and have even been shown to physically interact with each other, recent evidence suggests that they may also have distinct roles. The purpose of this review is to summarize the most recent insights into how CGI-58 and ATGL regulate lipid metabolism and signaling.
Collapse
|
37
|
MacPherson REK, Castellani L, Beaudoin MS, Wright DC. Evidence for fatty acids mediating CL 316,243-induced reductions in blood glucose in mice. Am J Physiol Endocrinol Metab 2014; 307:E563-70. [PMID: 25096179 PMCID: PMC4187028 DOI: 10.1152/ajpendo.00287.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CL 316,243, a β3-adrenergic agonist, was developed as an antiobesity and diabetes drug and causes rapid decreases in blood glucose levels in mice. The mechanisms mediating this effect have not been fully elucidated; thus, the purpose of the current study was to examine the role of fatty acids and interleukin-6, reputed mediators of insulin secretion, in this process. To address this question, we used physiological and pharmacological approaches in combination with knockout mouse models. CL 316,243 treatment in male C57BL6 mice increased plasma fatty acids, glycerol, interleukin-6, and insulin and reduced blood glucose concentrations 2 h following injections. The ability of CL 316,243 to increase insulin and fatty acids and reduce glucose was preserved in interleukin-6-deficient mice. CL 316,243-induced drops in blood glucose occurred in parallel with increases in circulating fatty acids but prior to increases in plasma interleukin-6. CL 316,243-mediated increases in plasma insulin levels and reductions in blood glucose were attenuated when mice were pretreated with the lipase inhibitor nicotinic acid or in whole body adipose tissue triglyceride lipase knockout mice. Collectively, our findings demonstrate an important role for fatty acids in mediating the effects of CL 316,243 in mice. Not only do our results provide new insight into the mechanisms of action of CL 316,243, but they also hint at an unappreciated aspect of adipose tissue -pancreas cross-talk.
Collapse
Affiliation(s)
- Rebecca E K MacPherson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Laura Castellani
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Marie-Soleil Beaudoin
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
38
|
α/β-Hydrolase domain-6-accessible monoacylglycerol controls glucose-stimulated insulin secretion. Cell Metab 2014; 19:993-1007. [PMID: 24814481 DOI: 10.1016/j.cmet.2014.04.003] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 01/24/2014] [Accepted: 03/26/2014] [Indexed: 02/07/2023]
Abstract
Glucose metabolism in pancreatic β cells stimulates insulin granule exocytosis, and this process requires generation of a lipid signal. However, the signals involved in lipid amplification of glucose-stimulated insulin secretion (GSIS) are unknown. Here we show that in β cells, glucose stimulates production of lipolysis-derived long-chain saturated monoacylglycerols, which further increase upon inhibition of the membrane-bound monoacylglycerol lipase α/β-Hydrolase Domain-6 (ABHD6). ABHD6 expression in β cells is inversely proportional to GSIS. Exogenous monoacylglycerols stimulate β cell insulin secretion and restore GSIS suppressed by the pan-lipase inhibitor orlistat. Whole-body and β-cell-specific ABHD6-KO mice exhibit enhanced GSIS, and their islets show elevated monoacylglycerol production and insulin secretion in response to glucose. Inhibition of ABHD6 in diabetic mice restores GSIS and improves glucose tolerance. Monoacylglycerol binds and activates the vesicle priming protein Munc13-1, thereby inducing insulin exocytosis. We propose saturated monoacylglycerol as a signal for GSIS and ABHD6 as a negative modulator of insulin secretion.
Collapse
|
39
|
Nielsen TS, Jessen N, Jørgensen JOL, Møller N, Lund S. Dissecting adipose tissue lipolysis: molecular regulation and implications for metabolic disease. J Mol Endocrinol 2014; 52:R199-222. [PMID: 24577718 DOI: 10.1530/jme-13-0277] [Citation(s) in RCA: 278] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lipolysis is the process by which triglycerides (TGs) are hydrolyzed to free fatty acids (FFAs) and glycerol. In adipocytes, this is achieved by sequential action of adipose TG lipase (ATGL), hormone-sensitive lipase (HSL), and monoglyceride lipase. The activity in the lipolytic pathway is tightly regulated by hormonal and nutritional factors. Under conditions of negative energy balance such as fasting and exercise, stimulation of lipolysis results in a profound increase in FFA release from adipose tissue (AT). This response is crucial in order to provide the organism with a sufficient supply of substrate for oxidative metabolism. However, failure to efficiently suppress lipolysis when FFA demands are low can have serious metabolic consequences and is believed to be a key mechanism in the development of type 2 diabetes in obesity. As the discovery of ATGL in 2004, substantial progress has been made in the delineation of the remarkable complexity of the regulatory network controlling adipocyte lipolysis. Notably, regulatory mechanisms have been identified on multiple levels of the lipolytic pathway, including gene transcription and translation, post-translational modifications, intracellular localization, protein-protein interactions, and protein stability/degradation. Here, we provide an overview of the recent advances in the field of AT lipolysis with particular focus on the molecular regulation of the two main lipases, ATGL and HSL, and the intracellular and extracellular signals affecting their activity.
Collapse
Affiliation(s)
- Thomas Svava Nielsen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, DenmarkThe Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Niels Jessen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, DenmarkThe Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Jens Otto L Jørgensen
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Niels Møller
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| | - Sten Lund
- The Novo Nordisk Foundation Center for Basic Metabolic ResearchSection on Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 6.6.30, DK-2200 N Copenhagen, DenmarkDepartment of Endocrinology and Internal MedicineAarhus University Hospital, Nørrebrogade 44, Bldg. 3.0, 8000 Aarhus C, DenmarkDepartment of Molecular MedicineAarhus University Hospital, Brendstrupgårdsvej 100, 8200 Aarhus N, Denmark
| |
Collapse
|
40
|
Kratky D, Obrowsky S, Kolb D, Radovic B. Pleiotropic regulation of mitochondrial function by adipose triglyceride lipase-mediated lipolysis. Biochimie 2014; 96:106-12. [PMID: 23827855 PMCID: PMC3859496 DOI: 10.1016/j.biochi.2013.06.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 06/20/2013] [Indexed: 12/12/2022]
Abstract
Lipolysis is defined as the catabolism of triacylglycerols (TGs) stored in cellular lipid droplets. Recent discoveries of essential lipolytic enzymes and characterization of numerous regulatory proteins and mechanisms have fundamentally changed our perception of lipolysis and its impact on cellular metabolism. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme for TG catabolism in most cells and tissues. This review focuses on recent advances in understanding the (patho)physiological impact due to defective lipolysis by ATGL deficiency on mitochondrial (dys)function. Depending on the type of cells and tissues investigated, absence of ATGL has pleiotropic roles in mitochondrial function.
Collapse
Affiliation(s)
- Dagmar Kratky
- Institute of Molecular Biology and Biochemistry, Center for Molecular Medicine, Medical University of Graz, Harrachgasse 21, 8010 Graz, Austria.
| | | | | | | |
Collapse
|
41
|
Pearson GL, Mellett N, Chu KY, Cantley J, Davenport A, Bourbon P, Cosner CC, Helquist P, Meikle PJ, Biden TJ. Lysosomal acid lipase and lipophagy are constitutive negative regulators of glucose-stimulated insulin secretion from pancreatic beta cells. Diabetologia 2014; 57:129-39. [PMID: 24149836 DOI: 10.1007/s00125-013-3083-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 09/26/2013] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS Lipolytic breakdown of endogenous lipid pools in pancreatic beta cells contributes to glucose-stimulated insulin secretion (GSIS) and is thought to be mediated by acute activation of neutral lipases in the amplification pathway. Recently it has been shown in other cell types that endogenous lipid can be metabolised by autophagy, and this lipophagy is catalysed by lysosomal acid lipase (LAL). This study aimed to elucidate a role for LAL and lipophagy in pancreatic beta cells. METHODS We employed pharmacological and/or genetic inhibition of autophagy and LAL in MIN6 cells and primary islets. Insulin secretion following inhibition was measured using RIA. Lipid accumulation was assessed by MS and confocal microscopy (to visualise lipid droplets) and autophagic flux was analysed by western blot. RESULTS Insulin secretion was increased following chronic (≥ 8 h) inhibition of LAL. This was more pronounced with glucose than with non-nutrient stimuli and was accompanied by augmentation of neutral lipid species. Similarly, following inhibition of autophagy in MIN6 cells, the number of lipid droplets was increased and GSIS was potentiated. Inhibition of LAL or autophagy in primary islets also increased insulin secretion. This augmentation of GSIS following LAL or autophagy inhibition was dependent on the acute activation of neutral lipases. CONCLUSIONS/INTERPRETATION Our data suggest that lysosomal lipid degradation, using LAL and potentially lipophagy, contributes to neutral lipid turnover in beta cells. It also serves as a constitutive negative regulator of GSIS by depletion of substrate for the non-lysosomal neutral lipases that are activated acutely by glucose.
Collapse
Affiliation(s)
- Gemma L Pearson
- Diabetes and Obesity Department, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tang T, Abbott MJ, Ahmadian M, Lopes AB, Wang Y, Sul HS. Desnutrin/ATGL activates PPARδ to promote mitochondrial function for insulin secretion in islet β cells. Cell Metab 2013; 18:883-895. [PMID: 24268737 PMCID: PMC3871209 DOI: 10.1016/j.cmet.2013.10.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 07/31/2013] [Accepted: 10/08/2013] [Indexed: 02/02/2023]
Abstract
Excessive caloric intake leading to obesity is associated with insulin resistance and dysfunction of islet β cells. High-fat feeding decreases desnutrin (also called ATGL/PNPLA2) levels in islets. Here we show that desnutrin ablation via RIP-Cre (βKO) or RIP-CreER results in hyperglycemia with impaired glucose-stimulated insulin secretion (GSIS). Due to decreased lipolysis, islets have higher TAG content but lower free FA levels. βKO islets exhibit impaired mitochondrial respiration and lower production of ATP required for GSIS, along with decreased expression of PPARδ target genes involved in mitochondrial oxidation. Furthermore, synthetic PPARδ, but not PPARα, agonist restores GSIS and expression of mitochondrial oxidative genes in βKO mice, revealing that desnutrin-catalyzed lipolysis generates PPARδ ligands. Finally, adenoviral expression of desnutrin in βKO islets restores all defects of βKO islet phenotype and function, including GSIS and mitochondrial defects, demonstrating the critical role of the desnutrin-PPARδ-mitochondrial oxidation axis in regulating islet β cell GSIS.
Collapse
Affiliation(s)
- Tianyi Tang
- Endocrinology Program, University of California, Berkeley, CA, USA 94720
| | - Marcia J. Abbott
- Department of Nutritional Science and Toxicology, University of California, Berkeley, CA, USA 94720
| | - Maryam Ahmadian
- Department of Nutritional Science and Toxicology, University of California, Berkeley, CA, USA 94720
| | | | - Yuhui Wang
- Department of Nutritional Science and Toxicology, University of California, Berkeley, CA, USA 94720
| | - Hei Sook Sul
- Department of Nutritional Science and Toxicology, University of California, Berkeley, CA, USA 94720
- Endocrinology Program, University of California, Berkeley, CA, USA 94720
| |
Collapse
|
43
|
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: 100] [Impact Index Per Article: 8.3] [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
|
44
|
Serr J, Li X, Lee K. The Regulation of Lipolysis in Adipose Tissue. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2013. [DOI: 10.5187/jast.2013.55.4.303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Abstract
The pancreatic islet β cell senses circulating levels of calorigenic nutrients to secrete insulin according to the needs of the organism. Altered insulin secretion is linked to various disorders such as diabetes, hypoglycemic states, and cardiometabolic diseases. Fuel stimuli, including glucose, free fatty acids, and amino acids, promote insulin granule exocytosis primarily via their metabolism in β cells and the production of key signaling metabolites. This paper reviews our current knowledge of the pathways involved in both positive and negative metabolic signaling for insulin secretion and assesses the role of established and candidate metabolic coupling factors, keeping recent developments in focus.
Collapse
Affiliation(s)
- Marc Prentki
- Molecular Nutrition Unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, QC, Canada.
| | | | | |
Collapse
|
46
|
Rancoule C, Attané C, Grès S, Fournel A, Dusaulcy R, Bertrand C, Vinel C, Tréguer K, Prentki M, Valet P, Saulnier-Blache JS. Lysophosphatidic acid impairs glucose homeostasis and inhibits insulin secretion in high-fat diet obese mice. Diabetologia 2013; 56:1394-402. [PMID: 23508306 DOI: 10.1007/s00125-013-2891-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 03/04/2013] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS Lysophosphatidic acid (LPA) is a lipid mediator produced by adipocytes that acts via specific G-protein-coupled receptors; its synthesis is modulated in obesity. We previously reported that reducing adipocyte LPA production in high-fat diet (HFD)-fed obese mice is associated with improved glucose tolerance, suggesting a negative impact of LPA on glucose homeostasis. Here, our aim was to test this hypothesis. METHODS First, glucose tolerance and plasma insulin were assessed after acute (30 min) injection of LPA (50 mg/kg) or of the LPA1/LPA3 receptor antagonist Ki16425 (5 mg kg(-1) day(-1), i.p.) in non-obese mice fed a normal diet (ND) and in obese/prediabetic (defined as glucose-intolerant) HFD mice. Glucose and insulin tolerance, pancreas morphology, glycogen storage, glucose oxidation and glucose transport were then studied after chronic treatment (3 weeks) of HFD mice with Ki16425. RESULTS In ND and HFD mice, LPA acutely impaired glucose tolerance by inhibiting glucose-induced insulin secretion. These effects were blocked by pre-injection of Ki16425 (5 mg/kg, i.p.). Inhibition of glucose-induced insulin secretion by LPA also occurred in isolated mouse islets. Plasma LPA was higher in HFD mice than in ND mice and Ki16425 transiently improved glucose tolerance. The beneficial effect of Ki16425 became permanent after chronic treatment and was associated with increased pancreatic islet mass and higher fasting insulinaemia. Chronic treatment with Ki16425 also improved insulin tolerance and increased liver glycogen storage and basal glucose use in skeletal muscle. CONCLUSIONS/INTERPRETATION Exogenous and endogenous LPA exerts a deleterious effect on glucose disposal through a reduction of plasma insulin; pharmacological blockade of LPA receptors improves glucose homeostasis in obese/prediabetic mice.
Collapse
Affiliation(s)
- C Rancoule
- Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabaties, Inserm U1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
All organisms use fatty acids (FAs) for energy substrates and as precursors for membrane and signaling lipids. The most efficient way to transport and store FAs is in the form of triglycerides (TGs); however, TGs are not capable of traversing biological membranes and therefore need to be cleaved by TG hydrolases ("lipases") before moving in or out of cells. This biochemical process is generally called "lipolysis." Intravascular lipolysis degrades lipoprotein-associated TGs to FAs for their subsequent uptake by parenchymal cells, whereas intracellular lipolysis generates FAs and glycerol for their release (in the case of white adipose tissue) or use by cells (in the case of other tissues). Although the importance of lipolysis has been recognized for decades, many of the key proteins involved in lipolysis have been uncovered only recently. Important new developments include the discovery of glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), the molecule that moves lipoprotein lipase from the interstitial spaces to the capillary lumen, and the discovery of adipose triglyceride lipase (ATGL) and comparative gene identification-58 (CGI-58) as crucial molecules in the hydrolysis of TGs within cells. This review summarizes current views of lipolysis and highlights the relevance of this process to human disease.
Collapse
Affiliation(s)
- Stephen G. Young
- Department of Medicine
- Department of Human Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| |
Collapse
|
48
|
Zierler KA, Jaeger D, Pollak NM, Eder S, Rechberger GN, Radner FPW, Woelkart G, Kolb D, Schmidt A, Kumari M, Preiss-Landl K, Pieske B, Mayer B, Zimmermann R, Lass A, Zechner R, Haemmerle G. Functional cardiac lipolysis in mice critically depends on comparative gene identification-58. J Biol Chem 2013; 288:9892-9904. [PMID: 23413028 DOI: 10.1074/jbc.m112.420620] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Efficient catabolism of cellular triacylglycerol (TG) stores requires the TG hydrolytic activity of adipose triglyceride lipase (ATGL). The presence of comparative gene identification-58 (CGI-58) strongly increased ATGL-mediated TG catabolism in cell culture experiments. Mutations in the genes coding for ATGL or CGI-58 in humans cause neutral lipid storage disease characterized by TG accumulation in multiple tissues. ATGL gene mutations cause a severe phenotype especially in cardiac muscle leading to cardiomyopathy that can be lethal. In contrast, CGI-58 gene mutations provoke severe ichthyosis and hepatosteatosis in humans and mice, whereas the role of CGI-58 in muscle energy metabolism is less understood. Here we show that mice lacking CGI-58 exclusively in muscle (CGI-58KOM) developed severe cardiac steatosis and cardiomyopathy linked to impaired TG catabolism and mitochondrial fatty acid oxidation. The marked increase in ATGL protein levels in cardiac muscle of CGI-58KOM mice was unable to compensate the lack of CGI-58. The addition of recombinant CGI-58 to cardiac lysates of CGI-58KOM mice completely reconstituted TG hydrolytic activities. In skeletal muscle, the lack of CGI-58 similarly provoked TG accumulation. The addition of recombinant CGI-58 increased TG hydrolytic activities in control and CGI-58KOM tissue lysates, elucidating the limiting role of CGI-58 in skeletal muscle TG catabolism. Finally, muscle CGI-58 deficiency affected whole body energy homeostasis, which is caused by impaired muscle TG catabolism and increased cardiac glucose uptake. In summary, this study demonstrates that functional muscle lipolysis depends on both CGI-58 and ATGL.
Collapse
Affiliation(s)
- Kathrin A Zierler
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Doris Jaeger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Nina M Pollak
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Sandra Eder
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Franz P W Radner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Gerald Woelkart
- Department of Pharmacology and Toxicology, University of Graz, 8010 Graz, Austria
| | - Dagmar Kolb
- Center for Medical Research, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Albrecht Schmidt
- Department of Cardiology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Manju Kumari
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Burkert Pieske
- Department of Cardiology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Bernd Mayer
- Department of Pharmacology and Toxicology, University of Graz, 8010 Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria.
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| |
Collapse
|
49
|
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
|
50
|
Ynsa MD, Minquin R, Rajendran R, Pinheiro T, Watt F. Consequences of a fat diet in the distribution of minerals within pancreatic tissues of rats and rabbits. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2012; 18:1060-1066. [PMID: 23046744 DOI: 10.1017/s1431927612001547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The effects of plasma lipid overload on pancreatic islet function and on mineral imbalance are issues under debate. However, the outcomes may be biased by the different metabolisms of different species. This prospective study evaluated whether a high fat diet intake changed the distribution of physiologically relevant elements within pancreatic endocrine and exocrine tissues of Sprague Dawley rats and New Zealand White rabbits. Nuclear microscopy techniques provided images of the specimen density and structure as well as the elemental distributions and quantification of P, S, Cl, K, Ca, Fe, and Zn using unstained cryosections of pancreas. Our results indicate that pancreatic islets in normal rats and rabbits had lower tissue density and higher Ca, Fe, and Zn content compared to exocrine tissue, and that rabbit islets exhibit the highest Zn content (3,300 μg/g in rabbits versus 510 μg/g in rats). Fat diet intake resulted in large deposits of fat in the pancreas, which modified the density contrast of tissues and also resulted in a twofold decrease of Ca and Zn concentrations in islets of both rats and rabbits. This result indicates that a fat diet leads to a reduction in essential trace element concentrations in pancreas, which in turn may hamper endocrine function.
Collapse
Affiliation(s)
- M Dolores Ynsa
- Centro de Micro-Análisis de Materiales, Universidad Autonoma de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain.
| | | | | | | | | |
Collapse
|