1
|
Kristensen JM, Kjøbsted R, Larsen TJ, Carl CS, Hingst JR, Onslev J, Birk JB, Thorup A, Steenberg DE, Knudsen JR, Henriksen NS, Needham EJ, Halling JF, Gudiksen A, Rundsten CF, Hanghøj KE, Stinson SE, Hoier B, Hansen CC, Jensen TE, Hellsten Y, Pilegaard H, Grarup N, Olesen J, Humphrey SJ, James DE, Pedersen ML, Richter EA, Hansen T, Jørgensen ME, Wojtaszewski JFP. Skeletal muscle from TBC1D4 p.Arg684Ter variant carriers is severely insulin resistant but exhibits normal metabolic responses during exercise. Nat Metab 2024; 6:2254-2266. [PMID: 39482542 DOI: 10.1038/s42255-024-01153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 09/27/2024] [Indexed: 11/03/2024]
Abstract
In the Greenlandic Inuit population, 4% are homozygous carriers of a genetic nonsense TBC1D4 p.Arg684Ter variant leading to loss of the muscle-specific isoform of TBC1D4 and an approximately tenfold increased risk of type 2 diabetes1. Here we show the metabolic consequences of this variant in four female and four male homozygous carriers and matched controls. An extended glucose tolerance test reveals prolonged hyperglycaemia followed by reactive hypoglycaemia in the carriers. Whole-body glucose disposal is impaired during euglycaemic-hyperinsulinaemic clamp conditions and associates with severe insulin resistance in skeletal muscle only. Notably, a marked reduction in muscle glucose transporter GLUT4 and associated proteins is observed. While metabolic regulation during exercise remains normal, the insulin-sensitizing effect of a single exercise bout is compromised. Thus, loss of the muscle-specific isoform of TBC1D4 causes severe skeletal muscle insulin resistance without baseline hyperinsulinaemia. However, physical activity can ameliorate this condition. These observations offer avenues for personalized interventions and targeted preventive strategies.
Collapse
Affiliation(s)
- Jonas M Kristensen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kjøbsted
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Trine J Larsen
- Greenland Center of Health Research, Institute of Institute of Health and Nature, University of Greenland, Nuuk, Greenland
| | - Christian S Carl
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Janne R Hingst
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Johan Onslev
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jesper B Birk
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Anette Thorup
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Dorte E Steenberg
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jonas R Knudsen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai S Henriksen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Elise J Needham
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Jens F Halling
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Anders Gudiksen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Carsten F Rundsten
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristian E Hanghøj
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sara E Stinson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Hoier
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Camilla C Hansen
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Thomas E Jensen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Olesen
- Queen Ingrid Primary Health Care Center, Nuuk, Greenland
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Michael L Pedersen
- Greenland Center of Health Research, Institute of Institute of Health and Nature, University of Greenland, Nuuk, Greenland
- Steno Diabetes Center Greenland, Nuuk, Greenland
| | - Erik A Richter
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marit E Jørgensen
- Greenland Center of Health Research, Institute of Institute of Health and Nature, University of Greenland, Nuuk, Greenland
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- National Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jørgen F P Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Su S, Quan C, Chen Q, Wang R, Du Q, Zhu S, Li M, Yang X, Rong P, Chen J, Bai Y, Zheng W, Feng W, Liu M, Xie B, Ouyang K, Shi YS, Lan F, Zhang X, Xiao R, Chen X, Wang HY, Chen S. AS160 is a lipid-responsive regulator of cardiac Ca 2+ homeostasis by controlling lysophosphatidylinositol metabolism and signaling. Nat Commun 2024; 15:9602. [PMID: 39505896 PMCID: PMC11542008 DOI: 10.1038/s41467-024-54031-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
The obese heart undergoes metabolic remodeling and exhibits impaired calcium (Ca2+) homeostasis, which are two critical assaults leading to cardiac dysfunction. The molecular mechanisms underlying these alterations in obese heart are not well understood. Here, we show that the Rab-GTPase activating protein AS160 is a lipid-responsive regulator of Ca2+ homeostasis through governing lysophosphatidylinositol metabolism and signaling. Palmitic acid/high fat diet inhibits AS160 activity through phosphorylation by NEK6, which consequently activates its downstream target Rab8a. Inactivation of AS160 in cardiomyocytes elevates cytosolic Ca2+ that subsequently impairs cardiac contractility. Mechanistically, Rab8a downstream of AS160 interacts with DDHD1 to increase lysophosphatidylinositol metabolism and signaling that leads to Ca2+ release from sarcoplasmic reticulum. Inactivation of NEK6 prevents inhibition of AS160 by palmitic acid/high fat diet, and alleviates cardiac dysfunction in high fat diet-fed mice. Together, our findings reveal a regulatory mechanism governing metabolic remodeling and Ca2+ homeostasis in obese heart, and have therapeutic implications to combat obesity cardiomyopathy.
Collapse
Affiliation(s)
- Shu Su
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Chao Quan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ruizhen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Qian Du
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Sangsang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Min Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Xinyu Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Jiang Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yingyu Bai
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wen Zheng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Minjun Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Bingxian Xie
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Biomedical Engineering for Cardiovascular Disease Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Ruiping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Xiongwen Chen
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, China.
- Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China.
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Nanjing, China.
| |
Collapse
|
3
|
Tian R, Zhao P, Ding X, Wang X, Jiang X, Chen S, Cai Z, Li L, Chen S, Liu W, Sun Q. TBC1D4 antagonizes RAB2A-mediated autophagic and endocytic pathways. Autophagy 2024; 20:2426-2443. [PMID: 38964379 PMCID: PMC11572321 DOI: 10.1080/15548627.2024.2367907] [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] [Received: 09/25/2023] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024] Open
Abstract
Macroautophagic/autophagic and endocytic pathways play essential roles in maintaining homeostasis at different levels. It remains poorly understood how both pathways are coordinated and fine-tuned for proper lysosomal degradation of diverse cargoes. We and others recently identified a Golgi-resident RAB GTPase, RAB2A, as a positive regulator that controls both autophagic and endocytic pathways. In the current study, we report that TBC1D4 (TBC1 domain family member 4), a TBC domain-containing protein that plays essential roles in glucose homeostasis, suppresses RAB2A-mediated autophagic and endocytic pathways. TBC1D4 bound to RAB2A through its N-terminal PTB2 domain, which impaired RAB2A-mediated autophagy at the early stage by preventing ULK1 complex activation. During the late stage of autophagy, TBC1D4 impeded the association of RUBCNL/PACER and RAB2A with STX17 on autophagosomes by direct interaction with RUBCNL via its N-terminal PTB1 domain. Disruption of the autophagosomal trimeric complex containing RAB2A, RUBCNL and STX17 resulted in defective HOPS recruitment and eventually abortive autophagosome-lysosome fusion. Furthermore, TBC1D4 inhibited RAB2A-mediated endocytic degradation independent of RUBCNL. Therefore, TBC1D4 and RAB2A form a dual molecular switch to modulate autophagic and endocytic pathways. Importantly, hepatocyte- or adipocyte-specific tbc1d4 knockout in mice led to elevated autophagic flux and endocytic degradation and tissue damage. Together, this work establishes TBC1D4 as a critical molecular brake in autophagic and endocytic pathways, providing further mechanistic insights into how these pathways are intertwined both in vitro and in vivo.Abbreviations: ACTB: actin beta; ATG9: autophagy related 9; ATG14: autophagy related 14; ATG16L1: autophagy related 16 like 1; CLEM: correlative light electron microscopy; Ctrl: control; DMSO: dimethyl sulfoxide; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; FL: full length; GAP: GTPase-activating protein; GFP: green fluorescent protein; HOPS: homotypic fusion and protein sorting; IP: immunoprecipitation; KD: knockdown; KO: knockout; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; OE: overexpression; PG: phagophore; PtdIns3K: class III phosphatidylinositol 3-kinase; SLC2A4/GLUT4: solute carrier family 2 member 4; SQSTM1/p62: sequestosome 1; RUBCNL/PACER: rubicon like autophagy enhancer; STX17: syntaxin 17; TAP: tandem affinity purification; TBA: total bile acid; TBC1D4: TBC1 domain family member 4; TUBA1B: tubulin alpha 1b; ULK1: unc-51 like autophagy activating kinase 1; VPS39: VPS39 subunit of HOPS complex; WB: western blot; WT: wild type.
Collapse
Affiliation(s)
- Rui Tian
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengwei Zhao
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianming Ding
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyi Wang
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Jiang
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Zhijian Cai
- Institute of Immunology, and Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Li
- Proteomics Center, National Institute of Biological Sciences, Beijing, China
| | - She Chen
- Proteomics Center, National Institute of Biological Sciences, Beijing, China
| | - Wei Liu
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiming Sun
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
Springer C, Binsch C, Weide D, Toska L, Cremer AL, Backes H, Scheel AK, Espelage L, Kotzka J, Sill S, Kurowski A, Kim D, Karpinski S, Schnurr TM, Hansen T, Hartwig S, Lehr S, Cames S, Brüning JC, Lienhard M, Herwig R, Börno S, Timmermann B, Al-Hasani H, Chadt A. Depletion of TBC1D4 Improves the Metabolic Exercise Response by Overcoming Genetically Induced Peripheral Insulin Resistance. Diabetes 2024; 73:1058-1071. [PMID: 38608276 DOI: 10.2337/db23-0463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 04/02/2024] [Indexed: 04/14/2024]
Abstract
The Rab-GTPase-activating protein (RabGAP) TBC1D4 (AS160) represents a key component in the regulation of glucose transport into skeletal muscle and white adipose tissue (WAT) and is therefore crucial during the development of insulin resistance and type 2 diabetes. Increased daily activity has been shown to be associated with improved postprandial hyperglycemia in allele carriers of a loss-of-function variant in the human TBC1D4 gene. Using conventional Tbc1d4-deficient mice (D4KO) fed a high-fat diet, we show that moderate endurance exercise training leads to substantially improved glucose and insulin tolerance and enhanced expression levels of markers for mitochondrial activity and browning in WAT from D4KO animals. Importantly, in vivo and ex vivo analyses of glucose uptake revealed increased glucose clearance in interscapular brown adipose tissue and WAT from trained D4KO mice. Thus, chronic exercise is able to overcome the genetically induced insulin resistance caused by Tbc1d4 depletion. Gene variants in TBC1D4 may be relevant in future precision medicine as determinants of exercise response. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Christian Springer
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Christian Binsch
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Deborah Weide
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Laura Toska
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Anna L Cremer
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Cologne, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Cologne, Germany
| | - Anna K Scheel
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Lena Espelage
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Jörg Kotzka
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Sebastian Sill
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Anette Kurowski
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
| | - Daebin Kim
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
| | - Sandra Karpinski
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
| | - Theresia M Schnurr
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sonja Hartwig
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Stefan Lehr
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Sandra Cames
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Cologne, Germany
| | | | - Ralf Herwig
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Stefan Börno
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center (DDZ), Leibniz-Center for Diabetes Research at the Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| |
Collapse
|
5
|
Wang H, Kwak SE, Zheng A, Arias EB, Pan X, Duan D, Cartee GD. Phosphorylation of AS160-serine 704 is not essential for exercise-increase in insulin-stimulated glucose uptake by skeletal muscles from female or male rats. Am J Physiol Endocrinol Metab 2024; 326:E807-E818. [PMID: 38656130 PMCID: PMC11376492 DOI: 10.1152/ajpendo.00010.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024]
Abstract
One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle from rodents and humans of both sexes. We recently found that concurrent mutation of three key sites to prevent their phosphorylation (Ser588, Thr642, and Ser704) on Akt substrate of 160 kDa (AS160; also known as TBC1D4) reduced the magnitude of the enhancement of postexercise ISGU (PEX-ISGU) by muscle from male, but not female rats. However, we did not test the role of individual phosphorylation sites on PEX-ISGU. Accordingly, our current aim was to test whether AS160 Ser704 phosphorylation (pSer704) is required for elevated PEX-ISGU by muscle. AS160-knockout (AS160-KO) rats (female and male) were studied when either in sedentary or 3 h after acute exercise. Adeno-associated virus (AAV) vectors were used to enable muscle expression of wild-type AS160 (AAV-WT-AS160) or AS160 mutated Ser704 to alanine to prevent phosphorylation (AAV-1P-AS160). Paired epitrochlearis muscles from each rat were injected with AAV-WT-AS160 or AAV-1P-AS160. We discovered that regardless of sex 1) AS160 abundance in AS160-KO rats was similar in paired muscles expressing WT-AS160 versus 1P-AS160; 2) muscles from exercised versus sedentary rats had greater ISGU, and PEX-ISGU was slightly greater for muscles expressing 1P-AS160 versus contralateral muscles expressing WT-AS160; and 3) pAS160Thr642 was lower in muscles expressing 1P-AS160 versus paired muscles expressing WT-AS160. These results indicate that pAS160Ser704 was not essential for elevated PEX-ISGU by skeletal muscle from rats of either sex. Furthermore, elimination of the postexercise increase in pAS160Thr642 did not lessen the postexercise effect on ISGU.NEW & NOTEWORTHY The current study evaluated the role of Akt substrate of 160 kDa (AS160) phosphorylation on Ser704 in increased insulin-stimulated glucose uptake by skeletal muscle after exercise. Adeno-associated virus vectors were engineered to express either wild-type-AS160 or AS160 mutated so that it could not be phosphorylated on Ser704 in paired muscles from AS160-knockout rats. The results demonstrated that AS160 phosphorylation on Ser704 was not essential for exercise-induced elevation in insulin-stimulated glucose uptake by rats of either sex.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, United States
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Chemical and Biomedical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, United States
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
6
|
Kawakami F, Imai M, Isaka Y, Cookson MR, Maruyama H, Kubo M, Farrer MJ, Kanzaki M, Kawashima R, Maekawa T, Tamaki S, Kurosaki Y, Kojima F, Ohba K, Ichikawa T. LRRK2 negatively regulates glucose tolerance via regulation of membrane translocation of GLUT4 in adipocytes. FEBS Open Bio 2023; 13:2200-2214. [PMID: 37845194 PMCID: PMC10699104 DOI: 10.1002/2211-5463.13717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023] Open
Abstract
Epidemiological studies have shown that abnormalities of glucose metabolism are involved in leucine-rich repeat kinase 2 (LRRK2)-associated Parkinson's disease (PD). However, the physiological significance of this association is unclear. In the present study, we investigated the effect of LRRK2 on high-fat diet (HFD)-induced glucose intolerance using Lrrk2-knockout (KO) mice. We found for the first time that HFD-fed KO mice display improved glucose tolerance compared with their wild-type (WT) counterparts. In addition, high serum insulin and leptin, as well as low serum adiponectin resulting from HFD in WT mice were improved in KO mice. Using western blotting, we found that Lrrk2 is highly expressed in adipose tissues compared with other insulin-related tissues that are thought to be important in glucose tolerance, including skeletal muscle, liver, and pancreas. Lrrk2 expression and phosphorylation of its kinase substrates Rab8a and Rab10 were significantly elevated after HFD treatment in WT mice. In cell culture experiments, treatment with a LRRK2 kinase inhibitor stimulated insulin-dependent membrane translocation of glucose transporter 4 (Glut4) and glucose uptake in mouse 3T3-L1 adipocytes. We conclude that increased LRRK2 kinase activity in adipose tissue exacerbates glucose tolerance by suppressing Rab8- and Rab10-mediated GLUT4 membrane translocation.
Collapse
Affiliation(s)
- Fumitaka Kawakami
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
- Department of Health Administration, School of Allied Health SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
| | - Motoki Imai
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
- Department of Molecular Diagnostics, School of Allied Health SciencesKitasato UniversitySagamiharaJapan
| | - Yuki Isaka
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on AgingNational Institutes of HealthBethesdaMDUSA
| | - Hiroko Maruyama
- Department of Cytopathology, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
| | - Makoto Kubo
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
- Department of Microbiology, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
| | - Matthew J. Farrer
- Department of Neurology and Fixel InstituteUniversity of FloridaGainesvilleFLUSA
| | - Makoto Kanzaki
- Graduate School of Biomedical EngineeringTohoku UniversitySendaiJapan
| | - Rei Kawashima
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
| | - Tatsunori Maekawa
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
| | - Shun Tamaki
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
| | - Yoshifumi Kurosaki
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
- Department of Medical Laboratory Sciences, School of Allied Health SciencesKitasato UniversitySagamiharaJapan
| | - Fumiaki Kojima
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
- Department of Pharmacology, School of Allied Health SciencesKitasato UniversitySagamiharaJapan
| | - Kenichi Ohba
- Department of Health Administration, School of Allied Health SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
| | - Takafumi Ichikawa
- Department of Regulation Biochemistry, Graduate School of Medical SciencesKitasato UniversitySagamiharaJapan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health ScienceKitasato UniversitySagamiharaJapan
| |
Collapse
|
7
|
Wang H, Zheng A, Arias EB, Kwak SE, Pan X, Duan D, Cartee GD. AS160 expression, but not AS160 Serine-588, Threonine-642, and Serine-704 phosphorylation, is essential for elevated insulin-stimulated glucose uptake by skeletal muscle from female rats after acute exercise. FASEB J 2023; 37:e23021. [PMID: 37289137 DOI: 10.1096/fj.202300282rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle in both sexes. We recently found that muscle expression and phosphorylation of key sites of Akt substrate of 160 kDa (AS160; also called TBC1D4) are essential for the full-exercise effect on postexercise-ISGU (PEX-ISGU) in male rats. In striking contrast, AS160's role in increased PEX-ISGU has not been rigorously tested in females. Our rationale was to address this major knowledge gap. Wild-type (WT) and AS160-knockout (KO) rats were either sedentary or acutely exercised. Adeno-associated virus (AAV) vectors were engineered to express either WT-AS160 or AS160 mutated on key serine and threonine residues (Ser588, Thr642, and Ser704) to alanine to prevent their phosphorylation. AAV vectors were delivered to the muscle of AS160-KO rats to determine if WT-AS160 or phosphorylation-inactivated AS160 would influence PEX-ISGU. AS160-KO rats have lower skeletal muscle abundance of the GLUT4 glucose transporter protein. This GLUT4 deficit was rescued using AAV delivery of GLUT4 to determine if eliminating muscle GLUT4 deficiency would normalize PEX-ISGU. The novel results were as follows: (1) AS160 expression was required for greater PEX-ISGU; (2) rescuing muscle AS160 expression in AS160-KO rats restored elevated PEX-ISGU; (3) AS160's essential role for the postexercise increase in ISGU was not attributable to reduced muscle GLUT4 content; and (4) AS160 phosphorylation on Ser588, Thr642, and Ser704 was not essential for greater PEX-ISGU. In conclusion, these novel findings revealed that three phosphosites widely proposed to influence PEX-ISGU are not required for this important outcome in female rats.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, USA
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Zheng A, Wang H, Arias EB, Dong G, Zhao J, Cartee GD. Akt substrate of 160 kDa is essential for the calorie restriction-induced increase in insulin-stimulated glucose uptake by skeletal muscle of female rats. Appl Physiol Nutr Metab 2023; 48:283-292. [PMID: 36634338 PMCID: PMC11726319 DOI: 10.1139/apnm-2022-0414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We evaluated effects of calorie restriction (CR; consuming 65% of ad libitum (AL) intake) for 8 weeks on female wildtype (WT) and Akt substrate of 160 kDa knockout (AS160-KO) rats. Insulin-stimulated glucose uptake (ISGU) was determined in isolated epitrochlearis muscles incubated with 0, 50, 100, or 500 µU/mL insulin. Phosphorylation of key insulin signaling proteins that control ISGU (Akt and AS160) was assessed by immunoblotting (Akt phosphorylation on Threonine-308, pAktThr308 and Serine-473, pAktSer473; AS160 phosphorylation on Serine-588, pAS160Ser588, and Threonine-642, pAS160Thr642). Abundance of proteins that regulate ISGU (GLUT4 glucose transporter protein and hexokinase II) was also determined by immunoblotting. The major results were as follows: (i) WT-CR versus WT-AL rats had greater ISGU with 100 and 500 µU/mL insulin; (ii) CR versus WT-AL rats had greater GLUT4 protein abundance; (iii) WT-CR versus WT-AL rats had greater pAktThr308 with 500 µU/mL insulin; (iv) WT-CR versus WT-AL rats did not differ for pAktSer473, pAS160Ser588, or pAS160Thr642 at any insulin concentration; (v) AS160-KO versus WT rats with each diet had lower ISGU at each insulin concentration, but not lower pAkt on either phosphosite; (vi) AS160-KO versus WT rats had lower muscle GLUT4 abundance regardless of diet; and (vii) AS160-KO-CR versus AS160-KO-AL rats did not differ for ISGU, GLUT4 abundance, pAkt on either phosphosite, or pAS160 on either phosphosite. These novel results demonstrated that AS160 expression, but not greater pAS160 on key phosphosites, was essential for the CR-induced increases in muscle ISGU and GLUT4 abundance of female rats.
Collapse
Affiliation(s)
- Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Edward B. Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Gengfu Dong
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Jiahui Zhao
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Gregory D. Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Ouyang Q, Chen Q, Ke S, Ding L, Yang X, Rong P, Feng W, Cao Y, Wang Q, Li M, Su S, Wei W, Liu M, Liu J, Zhang X, Li JZ, Wang HY, Chen S. Rab8a as a mitochondrial receptor for lipid droplets in skeletal muscle. Dev Cell 2023; 58:289-305.e6. [PMID: 36800997 DOI: 10.1016/j.devcel.2023.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 11/17/2022] [Accepted: 01/26/2023] [Indexed: 02/18/2023]
Abstract
Dynamic interaction between lipid droplets (LDs) and mitochondria controls the mobilization of long-chain fatty acids (LCFAs) from LDs for mitochondrial β-oxidation in skeletal muscle in response to energy stress. However, little is known about the composition and regulation of the tethering complex mediating LD-mitochondrion interaction. Here, we identify Rab8a as a mitochondrial receptor for LDs forming the tethering complex with the LD-associated PLIN5 in skeletal muscle. In rat L6 skeletal muscle cells, the energy sensor AMPK increases the GTP-bound active Rab8a that promotes LD-mitochondrion interaction through binding to PLIN5 upon starvation. The assembly of the Rab8a-PLIN5 tethering complex also recruits the adipose triglyceride lipase (ATGL), which couples LCFA mobilization from LDs with its transfer into mitochondria for β-oxidation. Rab8a deficiency impairs fatty acid utilization and decreases endurance during exercise in a mouse model. These findings may help to elucidate the regulatory mechanisms underlying the beneficial effects of exercise on lipid homeostasis control.
Collapse
Affiliation(s)
- Qian Ouyang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China; Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China; Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Shunyuan Ke
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Longfei Ding
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Xinyu Yang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Ye Cao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Qi Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Min Li
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Shu Su
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Wen Wei
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Minjun Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Jin Liu
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Xu Zhang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China; Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China; Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210061, China.
| |
Collapse
|
10
|
Lee SR, Jeong SH, Mukae M, Jeong KJ, Kwun HJ, Hong EJ. GLUT4 degradation by GLUTFOURINH® in mice resembles moderate-obese diabetes of human with hyperglycemia and low lipid accumulation. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166668. [PMID: 36822448 DOI: 10.1016/j.bbadis.2023.166668] [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] [Received: 11/19/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUNDS AND AIMS Type 2 diabetes mellitus (T2D) is a chronic disease characterized by insulin resistance and hyperglycemia. To investigate T2D, genetic and chemical induced hyper-obese rodent models have been experimentally developed. However, establishment of moderate-obese diabetes model will confer diverse opportunities for translational studies. In this study, we found the chemical, GLUTFOURINH® (GFI), induces post-translational degradation of glucose transporter 4 (GLUT4). We aimed to establish novel diabetic model by using GFI. METHODS AND RESULTS Low plasma membrane GLUT4 (pmGLUT4) levels by GFI resulted in reduction of intracellular glucose uptake and TG, and increase of intracellular FFA in A204 cells. Likewise, GFI treatment decreased intracellular TG and increased intracellular FFA levels in Hep3B and 3T3-L1 cells. Mice were administered with GFI (16 mg/kg) for short-term (3-day) and long-term (28- and 31-day) to compared with vehicle injection, HFD model, and T2D model, respectively. Short-term and long-term GFI treatments induced hyperglycemia and hyperinsulinemia with low pmGLUT4 levels. Compared to HFD model, long-term GFI with HFD reduced adipose weight and intracellular TG accumulation, but increased plasma FFA. GFI treatment resulted in insulin resistance by showing low QUICKI and high HOMA-IR values, and low insulin response during insulin tolerance test. Additionally, low pmGLUT4 by GFI heightened hyperglycemia, hyperinsulinemia, and insulin resistance compared to T2D model. CONCLUSIONS In summary, we report GLUT4 degradation by novel chemical (GFI) induces moderate-obese diabetes representing hyperglycemia, insulin resistance and low intracellular lipid accumulation. The GLUT4 degradation by GFI has translational value for studying diseases related to moderate-obese diabetes.
Collapse
Affiliation(s)
- Sang R Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Su Hee Jeong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moeka Mukae
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kang Joo Jeong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyo-Jung Kwun
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
11
|
Binsch C, Barbosa DM, Hansen-Dille G, Hubert M, Hodge SM, Kolasa M, Jeruschke K, Weiß J, Springer C, Gorressen S, Fischer JW, Lienhard M, Herwig R, Börno S, Timmermann B, Cremer AL, Backes H, Chadt A, Al-Hasani H. Deletion of Tbc1d4/As160 abrogates cardiac glucose uptake and increases myocardial damage after ischemia/reperfusion. Cardiovasc Diabetol 2023; 22:17. [PMID: 36707786 PMCID: PMC9881301 DOI: 10.1186/s12933-023-01746-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Type 2 Diabetes mellitus (T2DM) is a major risk factor for cardiovascular disease and associated with poor outcome after myocardial infarction (MI). In T2DM, cardiac metabolic flexibility, i.e. the switch between carbohydrates and lipids as energy source, is disturbed. The RabGTPase-activating protein TBC1D4 represents a crucial regulator of insulin-stimulated glucose uptake in skeletal muscle by controlling glucose transporter GLUT4 translocation. A human loss-of-function mutation in TBC1D4 is associated with impaired glycemic control and elevated T2DM risk. The study's aim was to investigate TBC1D4 function in cardiac substrate metabolism and adaptation to MI. METHODS Cardiac glucose metabolism of male Tbc1d4-deficient (D4KO) and wild type (WT) mice was characterized using in vivo [18F]-FDG PET imaging after glucose injection and ex vivo basal/insulin-stimulated [3H]-2-deoxyglucose uptake in left ventricular (LV) papillary muscle. Mice were subjected to cardiac ischemia/reperfusion (I/R). Heart structure and function were analyzed until 3 weeks post-MI using echocardiography, morphometric and ultrastructural analysis of heart sections, complemented by whole heart transcriptome and protein measurements. RESULTS Tbc1d4-knockout abolished insulin-stimulated glucose uptake in ex vivo LV papillary muscle and in vivo cardiac glucose uptake after glucose injection, accompanied by a marked reduction of GLUT4. Basal cardiac glucose uptake and GLUT1 abundance were not changed compared to WT controls. D4KO mice showed mild impairments in glycemia but normal cardiac function. However, after I/R D4KO mice showed progressively increased LV endsystolic volume and substantially increased infarction area compared to WT controls. Cardiac transcriptome analysis revealed upregulation of the unfolded protein response via ATF4/eIF2α in D4KO mice at baseline. Transmission electron microscopy revealed largely increased extracellular matrix (ECM) area, in line with decreased cardiac expression of matrix metalloproteinases of D4KO mice. CONCLUSIONS TBC1D4 is essential for insulin-stimulated cardiac glucose uptake and metabolic flexibility. Tbc1d4-deficiency results in elevated cardiac endoplasmic reticulum (ER)-stress response, increased deposition of ECM and aggravated cardiac damage following MI. Hence, impaired TBC1D4 signaling contributes to poor outcome after MI.
Collapse
Affiliation(s)
- C. Binsch
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - D. M. Barbosa
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - G. Hansen-Dille
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - M. Hubert
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - S. M. Hodge
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - M. Kolasa
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - K. Jeruschke
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - J. Weiß
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - C. Springer
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - S. Gorressen
- grid.411327.20000 0001 2176 9917Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - J. W. Fischer
- grid.411327.20000 0001 2176 9917Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - M. Lienhard
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - R. Herwig
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - S. Börno
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - B. Timmermann
- grid.419538.20000 0000 9071 0620Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - A. L. Cremer
- grid.418034.a0000 0004 4911 0702Max Planck Institute for Metabolism Research, Cologne, Germany
| | - H. Backes
- grid.418034.a0000 0004 4911 0702Max Planck Institute for Metabolism Research, Cologne, Germany
| | - A. Chadt
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany ,grid.452622.5German Center for Diabetes Research, Partner Düsseldorf, Munich-Neuherberg, Germany
| | - H. Al-Hasani
- grid.429051.b0000 0004 0492 602XMedical Faculty, Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz-Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany ,grid.452622.5German Center for Diabetes Research, Partner Düsseldorf, Munich-Neuherberg, Germany
| |
Collapse
|
12
|
Larsen JK, Larsen MR, Birk JB, Steenberg DE, Hingst JR, Højlund K, Chadt A, Al-Hasani H, Deshmukh AS, Wojtaszewski JF, Kjøbsted R. Illumination of the Endogenous Insulin-Regulated TBC1D4 Interactome in Human Skeletal Muscle. Diabetes 2022; 71:906-920. [PMID: 35192682 PMCID: PMC9074744 DOI: 10.2337/db21-0855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022]
Abstract
Insulin-stimulated muscle glucose uptake is a key process in glycemic control. This process depends on the redistribution of glucose transporters to the surface membrane, a process that involves regulatory proteins such as TBC1D1 and TBC1D4. Accordingly, a TBC1D4 loss-of-function mutation in human skeletal muscle is associated with an increased risk of type 2 diabetes, and observations from carriers of a TBC1D1 variant associate this protein to a severe obesity phenotype. Here, we identified interactors of the endogenous TBC1D4 protein in human skeletal muscle by an unbiased proteomics approach. We detected 76 proteins as candidate TBC1D4 interactors. The binding of 12 of these interactors was regulated by insulin, including proteins known to be involved in glucose metabolism (e.g., 14-3-3 proteins and α-actinin-4 [ACTN4]). TBC1D1 also coprecipitated with TBC1D4 and vice versa in both human and mouse skeletal muscle. This interaction was not regulated by insulin or exercise in young, healthy, lean individuals. Similarly, the exercise- and insulin-regulated phosphorylation of the TBC1D1-TBC1D4 complex was intact. In contrast, we observed an altered interaction as well as compromised insulin-stimulated phosphoregulation of the TBC1D1-TBC1D4 complex in muscle of obese individuals with type 2 diabetes. Altogether, we provide a repository of TBC1D4 interactors in human and mouse skeletal muscle that serve as potential regulators of TBC1D4 function and, thus, insulin-stimulated glucose uptake in human skeletal muscle.
Collapse
Affiliation(s)
- Jeppe K. Larsen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Magnus R. Larsen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jesper B. Birk
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Dorte E. Steenberg
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Janne R. Hingst
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Alexandra Chadt
- German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hadi Al-Hasani
- German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F.P. Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Corresponding authors: Rasmus Kjøbsted, , and Jørgen F.P. Wojtaszewski,
| | - Rasmus Kjøbsted
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Corresponding authors: Rasmus Kjøbsted, , and Jørgen F.P. Wojtaszewski,
| |
Collapse
|
13
|
Roberts BS, Yang CQ, Neher SB. Characterization of lipoprotein lipase storage vesicles in 3T3-L1 adipocytes. J Cell Sci 2022; 135:jcs258734. [PMID: 34382637 PMCID: PMC8403984 DOI: 10.1242/jcs.258734] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/10/2021] [Indexed: 12/12/2022] Open
Abstract
Lipoprotein lipase (LPL) is a secreted triglyceride lipase involved in the clearance of very-low-density lipoproteins and chylomicrons from circulation. LPL is expressed primarily in adipose and muscle tissues and transported to the capillary lumen. LPL secretion is regulated by insulin in adipose tissue; however, few studies have examined the regulatory and trafficking steps involved in secretion. Here, we describe the intracellular localization and insulin-dependent trafficking of LPL in 3T3-L1 adipocytes. We compared LPL trafficking to the better characterized trafficking pathways taken by leptin and GLUT4 (also known as SLC2A4). We show that the LPL trafficking pathway shares some characteristics of these other pathways, but that LPL subcellular localization and trafficking are distinct from those of GLUT4 and leptin. LPL secretion occurs slowly in response to insulin and rapidly in response to the Ca2+ ionophore ionomycin. This regulated trafficking is dependent on Golgi protein kinase D and the ADP-ribosylation factor GTPase ARF1. Together, these data give support to a new trafficking pathway for soluble cargo that is active in adipocytes.
Collapse
Affiliation(s)
| | | | - Saskia B. Neher
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
14
|
Raun SH, Knudsen JR, Han X, Jensen TE, Sylow L. Cancer causes dysfunctional insulin signaling and glucose transport in a muscle-type-specific manner. FASEB J 2022; 36:e22211. [PMID: 35195922 DOI: 10.1096/fj.202101759r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/21/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
Abstract
Metabolic dysfunction and insulin resistance are emerging as hallmarks of cancer and cachexia, and impair cancer prognosis. Yet, the molecular mechanisms underlying impaired metabolic regulation are not fully understood. To elucidate the mechanisms behind cancer-induced insulin resistance in muscle, we isolated extensor digitorum longus (EDL) and soleus muscles from Lewis Lung Carcinoma tumor-bearing mice. Three weeks after tumor inoculation, muscles were isolated and stimulated with or without a submaximal dose of insulin (1.5 nM). Glucose transport was measured using 2-[3 H]Deoxy-Glucose and intramyocellular signaling was investigated using immunoblotting. In soleus muscles from tumor-bearing mice, insulin-stimulated glucose transport was abrogated concomitantly with abolished insulin-induced TBC1D4 and GSK3 phosphorylation. In EDL, glucose transport and TBC1D4 phosphorylation were not impaired in muscles from tumor-bearing mice, while AMPK signaling was elevated. Anabolic insulin signaling via phosphorylation of the mTORC1 targets, p70S6K thr389, and ribosomal-S6 ser235, were decreased by cancer in soleus muscle while increased or unaffected in EDL. In contrast, the mTOR substrate, pULK1 ser757, was reduced in both soleus and EDL by cancer. Hence, cancer causes considerable changes in skeletal muscle insulin signaling that is dependent on muscle-type, which could contribute to metabolic dysregulation in cancer. Thus, the skeletal muscle could be a target for managing metabolic dysfunction in cancer.
Collapse
Affiliation(s)
- Steffen H Raun
- Section of Molecular Physiology, Department of nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Roland Knudsen
- Section of Molecular Physiology, Department of nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Xiuqing Han
- Section of Molecular Physiology, Department of nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Lykke Sylow
- Section of Molecular Physiology, Department of nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Zheng A, Arias EB, Wang H, Kwak SE, Pan X, Duan D, Cartee GD. Exercise-Induced Improvement in Insulin-Stimulated Glucose Uptake by Rat Skeletal Muscle Is Absent in Male AS160-Knockout Rats, Partially Restored by Muscle Expression of Phosphomutated AS160, and Fully Restored by Muscle Expression of Wild-Type AS160. Diabetes 2022; 71:219-232. [PMID: 34753801 PMCID: PMC8914290 DOI: 10.2337/db21-0601] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022]
Abstract
One exercise session can elevate insulin-stimulated glucose uptake (ISGU) in skeletal muscle, but the mechanisms remain elusive. Circumstantial evidence suggests a role for Akt substrate of 160 kDa (AS160 or TBC1D4). We used genetic approaches to rigorously test this idea. The initial experiment evaluated the role of AS160 in postexercise increase in ISGU using muscles from male wild-type (WT) and AS160-knockout (KO) rats. The next experiment used AS160-KO rats with an adeno-associated virus (AAV) approach to determine if rescuing muscle AS160 deficiency could restore the ability of exercise to improve ISGU. The third experiment tested if eliminating the muscle GLUT4 deficit in AS160-KO rats via AAV-delivered GLUT4 would enable postexercise enhancement of ISGU. The final experiment used AS160-KO rats and AAV delivery of AS160 mutated to prevent phosphorylation of Ser588, Thr642, and Ser704 to evaluate their role in postexercise ISGU. We discovered the following: 1) AS160 expression was essential for postexercise increase in ISGU; 2) rescuing muscle AS160 expression of AS160-KO rats restored postexercise enhancement of ISGU; 3) restoring GLUT4 expression in AS160-KO muscle did not rescue the postexercise increase in ISGU; and 4) although AS160 phosphorylation on three key sites was not required for postexercise elevation in ISGU, it was essential for the full exercise effect.
Collapse
Affiliation(s)
- Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Edward B. Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO
| | - Gregory D. Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
- Institute of Gerontology, University of Michigan, Ann Arbor, MI
- Corresponding author: Gregory D. Cartee,
| |
Collapse
|
16
|
Stafeev I, Boldyreva M, Michurina S, Mamontova E, Ratner E, Menshikov M, Parfyonova Y. Grain-Based Dietary Background Impairs Restoration of Blood Flow and Skeletal Muscle During Hindlimb Ischemia in Comparison With Low-Fat and High-Fat Diets. Front Nutr 2022; 8:809732. [PMID: 35083264 PMCID: PMC8784406 DOI: 10.3389/fnut.2021.809732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/09/2021] [Indexed: 11/28/2022] Open
Abstract
Background: Among vascular pathologies associated with obesity, peripheral artery disease (PAD) occupies the important position. In clinical practice, nutritional interventions are recommended for patients with PAD. In this work, we investigated how the different dietary backgrounds affect the regeneration rate of ischemic hindlimb in mice. Methods: Male C57BL/6J mice were housed on three types of diet: low-fat (LFD), high-fat (HFD), and grain-based diet (GBD) for 13 weeks. Metabolic parameters including FBG level, ITT, and GTT were evaluated. The blood flow was assessed by laser Doppler scanning on 7, 14, and 21 days after hindlimb ischemia. Necrotic area of m.tibialis, macrophage infiltration, and angiogenesis/arteriogenesis were evaluated by histology. Glucose uptake in recovered skeletal muscle was analyzed using [3H]-2-deoxyglucose, and GLUT1 and GLUT4 expression were assessed by Western blotting. Results: In our work, we developed three experimental groups with different metabolic parameters: LFD with normal glucose metabolism, GBD with mild hyperglycemia, and HFD with impaired glucose tolerance. GBD-fed mice had a tendency to increase necrosis of m. tibialis and significantly higher macrophage infiltration than LFD and HFD groups. Moreover, GBD-fed mice had a trend to decreased blood flow recovery and significantly impaired arteriogenesis. Recovered skeletal muscle of GBD-fed mice had lower glucose uptake and decreased level of GLUT4 expression. Conclusion: Thus, we conclude that dietary background and metabolic status determine the rate of post-ischemic regeneration including angiogenesis, skeletal muscle recovery and metabolic activity. The most effective regeneration is supported by LFD, while the lowest rate of regeneration occurs on GBD.
Collapse
Affiliation(s)
- Iurii Stafeev
- National Medical Research Center for Cardiology, Moscow, Russia
| | - Maria Boldyreva
- National Medical Research Center for Cardiology, Moscow, Russia.,Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Svetlana Michurina
- National Medical Research Center for Cardiology, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | - Elizaveta Mamontova
- National Medical Research Center for Cardiology, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | | | | | - Yelena Parfyonova
- National Medical Research Center for Cardiology, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
17
|
Abstract
The Akt isoforms-AS160-GLUT4 axis is the primary axis that governs glucose homeostasis in the body. The first step on the path to insulin resistance is deregulated Akt isoforms. This could be Akt isoform expression, its phosphorylation, or improper isoform-specific redistribution to the plasma membrane in a specific tissue system. The second step is deregulated AS160 expression, its phosphorylation, improper dissociation from glucose transporter storage vesicles (GSVs), or its inability to bind to 14-3-3 proteins, thus not allowing it to execute its function. The final step is improper GLUT4 translocation and aberrant glucose uptake. These processes lead to insulin resistance in a tissue-specific way affecting the whole-body glucose homeostasis, eventually progressing to an overt diabetic phenotype. Thus, the relationship between these three key proteins and their proper regulation comes out as the defining axis of insulin signaling and -resistance. This review summarizes the role of this central axis in insulin resistance and disease in a new light.
Collapse
Affiliation(s)
- Medha Sharma
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
18
|
Rao XS, Cong XX, Gao XK, Shi YP, Shi LJ, Wang JF, Ni CY, He MJ, Xu Y, Yi C, Meng ZX, Liu J, Lin P, Zheng LL, Zhou YT. AMPK-mediated phosphorylation enhances the auto-inhibition of TBC1D17 to promote Rab5-dependent glucose uptake. Cell Death Differ 2021; 28:3214-3234. [PMID: 34045668 PMCID: PMC8630067 DOI: 10.1038/s41418-021-00809-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 02/04/2023] Open
Abstract
Dysregulation of glucose homeostasis contributes to insulin resistance and type 2 diabetes. Whilst exercise stimulated activation of AMP-activated protein kinase (AMPK), an important energy sensor, has been highlighted for its potential to promote insulin-stimulated glucose uptake, the underlying mechanisms for this remain largely unknown. Here we found that AMPK positively regulates the activation of Rab5, a small GTPase which is involved in regulating Glut4 translocation, in both myoblasts and skeletal muscles. We further verified that TBC1D17, identified as a potential interacting partner of Rab5 in our recent study, is a novel GTPase activating protein (GAP) of Rab5. TBC1D17-Rab5 axis regulates transport of Glut1, Glut4, and transferrin receptor. TBC1D17 interacts with Rab5 or AMPK via its TBC domain or N-terminal 1-306 region (N-Ter), respectively. Moreover, AMPK phosphorylates the Ser 168 residue of TBC1D17 which matches the predicted AMPK consensus motif. N-Ter of TBC1D17 acts as an inhibitory region by directly interacting with the TBC domain. Ser168 phosphorylation promotes intra-molecular interaction and therefore enhances the auto-inhibition of TBC1D17. Our findings reveal that TBC1D17 acts as a molecular bridge that links AMPK and Rab5 and delineate a previously unappreciated mechanism by which the activation of TBC/RabGAP is regulated.
Collapse
Affiliation(s)
- Xi Sheng Rao
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Xia Cong
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiu Kui Gao
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yin Pu Shi
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Jing Shi
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Feng Wang
- grid.13402.340000 0004 1759 700XDepartment of Respiratory Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen-Yao Ni
- grid.35403.310000 0004 1936 9991The School of Molecular and Cellular Biology, University of Illinois at Urbana Champaign, Urbana, IL USA
| | - Ming Jie He
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingke Xu
- grid.13402.340000 0004 1759 700XDepartment of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Endocrinology, the Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong Yi
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhuo-Xian Meng
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Zhejiang Provincial Key Laboratory of Pancreatic Disease of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinling Liu
- grid.13402.340000 0004 1759 700XDepartment of Pulmonology, the Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Peng Lin
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Ling Zheng
- grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Ting Zhou
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XZJU-UoE Institute, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Yang X, Chen Q, Ouyang Q, Rong P, Feng W, Quan C, Li M, Jiang Q, Liang H, Zhao TJ, Wang HY, Chen S. Tissue-Specific Splicing and Dietary Interaction of a Mutant As160 Allele Determine Muscle Metabolic Fitness in Rodents. Diabetes 2021; 70:1826-1842. [PMID: 33980689 DOI: 10.2337/db21-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022]
Abstract
Ethnic groups are physiologically and genetically adapted to their diets. Inuit bear a frequent AS160R684X mutation that causes type 2 diabetes. Whether this mutation evolutionarily confers adaptation in Inuit and how it causes metabolic disorders upon dietary changes are unknown due to limitations in human studies. Here, we develop a genetically modified rat model bearing an orthologous AS160R693X mutation, which mimics human patients exhibiting postprandial hyperglycemia and hyperinsulinemia. Importantly, a sugar-rich diet aggravates metabolic abnormalities in AS160R693X rats. The AS160R693X mutation diminishes a dominant long-variant AS160 without affecting a minor short-variant AS160 in skeletal muscle, which suppresses muscle glucose utilization but induces fatty acid oxidation. This fuel switch suggests a possible adaptation in Inuit who traditionally had lipid-rich hypoglycemic diets. Finally, induction of the short-variant AS160 restores glucose utilization in rat myocytes and a mouse model. Our findings have implications for development of precision treatments for patients bearing the AS160R684X mutation.
Collapse
Affiliation(s)
- Xinyu Yang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qiaoli Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qian Ouyang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Ping Rong
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Weikuan Feng
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Chao Quan
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Min Li
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, China
| | - Hui Liang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Tong-Jin Zhao
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hong Yu Wang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| |
Collapse
|
20
|
Bruhn L, Kjøbsted R, Quist JS, Gram AS, Rosenkilde M, Færch K, Wojtaszewski JF, Stallknecht B, Blond MB. Effect of exercise training on skeletal muscle protein expression in relation to insulin sensitivity: Per-protocol analysis of a randomized controlled trial (GO-ACTIWE). Physiol Rep 2021; 9:e14850. [PMID: 34042297 PMCID: PMC8157763 DOI: 10.14814/phy2.14850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 12/14/2022] Open
Abstract
Exercise training improves peripheral insulin sensitivity and leads to molecular adaptations in the skeletal muscle. We investigated changes in the expression of key muscle proteins in the glucose metabolic pathway following active commuting by bike or leisure-time exercise at two different intensities. In addition, potential associations between insulin sensitivity and muscle protein expression were examined. This per-protocol analysis included 72 out of 130 physically inactive, healthy women and men (20-45 years) with overweight/obesity (BMI: 25-35 kg/m2 ) who completed 6 months of no intervention (CON, n = 12), active commuting by bike (BIKE, n = 14), or leisure-time exercise of moderate (MOD, n = 28) or vigorous (VIG, n = 18) intensity. Exercise was prescribed 5 days/week with a weekly exercise energy expenditure of 1,600 kcal for women and 2,100 kcal for men. Insulin sensitivity was determined by a hyperinsulinemic euglycemic clamp and skeletal muscle biopsies were obtained from m. vastus lateralis and analyzed for protein expression at baseline and after 3 and 6 months of intervention. We found an increased expression of pyruvate dehydrogenase (PDH) in the exercise groups compared with the control group following 6 months of training. No differential effects were observed on the protein expression following moderate versus vigorous intensity exercise. In addition, we found a positive association between insulin sensitivity and the expression of glucose transporter type 4 as well as PDH. The positive association and the increase in expression of PDH after exercise training points toward a role for PDH in the training-induced enhancement of insulin sensitivity.
Collapse
Affiliation(s)
- Lea Bruhn
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Steno Diabetes Center CopenhagenGentofteDenmark
| | - Rasmus Kjøbsted
- Section of Molecular PhysiologyAugust Krogh ClubDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Jonas Salling Quist
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Steno Diabetes Center CopenhagenGentofteDenmark
| | - Anne Sofie Gram
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Mads Rosenkilde
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Kristine Færch
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Steno Diabetes Center CopenhagenGentofteDenmark
| | - Jørgen F.P. Wojtaszewski
- Section of Molecular PhysiologyAugust Krogh ClubDepartment of Nutrition, Exercise and SportsUniversity of CopenhagenCopenhagenDenmark
| | - Bente Stallknecht
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Martin Bæk Blond
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Steno Diabetes Center CopenhagenGentofteDenmark
| |
Collapse
|
21
|
Bledzka KM, Manaserh IH, Grondolsky J, Pfleger J, Roy R, Gao E, Chuprun JK, Koch WJ, Schumacher SM. A peptide of the amino-terminus of GRK2 induces hypertrophy and yet elicits cardioprotection after pressure overload. J Mol Cell Cardiol 2021; 154:137-153. [PMID: 33548241 DOI: 10.1016/j.yjmcc.2021.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/14/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptor (GPCR) kinase 2 (GRK2) expression and activity are elevated early on in response to several forms of cardiovascular stress and are a hallmark of heart failure. Interestingly, though, in addition to its well-characterized role in regulating GPCRs, mounting evidence suggests a GRK2 "interactome" that underlies a great diversity in its functional roles. Several such GRK2 interacting partners are important for adaptive and maladaptive myocyte growth; therefore, an understanding of domain-specific interactions with signaling and regulatory molecules could lead to novel targets for heart failure therapy. Herein, we subjected transgenic mice with cardiac restricted expression of a short, amino terminal fragment of GRK2 (βARKnt) to pressure overload and found that unlike their littermate controls or previous GRK2 fragments, they exhibited an increased left ventricular wall thickness and mass prior to cardiac stress that underwent proportional hypertrophic growth to controls after acute pressure overload. Importantly, despite this enlarged heart, βARKnt mice did not undergo the expected transition to heart failure observed in controls. Further, βARKnt expression limited adverse left ventricular remodeling and increased cell survival signaling. Proteomic analysis to identify βARKnt binding partners that may underlie the improved cardiovascular phenotype uncovered a selective functional interaction of both endogenous GRK2 and βARKnt with AKT substrate of 160 kDa (AS160). AS160 has emerged as a key downstream regulator of insulin signaling, integrating physiological and metabolic cues to couple energy demand to membrane recruitment of Glut4. Our preliminary data indicate that in βARKnt mice, cardiomyocyte insulin signaling is improved during stress, with a coordinate increase in spare respiratory activity and ATP production without metabolite switching. Surprisingly, these studies also revealed a significant decrease in gonadal fat weight, equivalent to human abdominal fat, in male βARKnt mice at baseline and following cardiac stress. These data suggest that the enhanced AS160-mediated signaling in the βARKnt mice may ameliorate pathological cardiac remodeling through direct modulation of insulin signaling within cardiomyocytes, and translate these to beneficial effects on systemic metabolism.
Collapse
Affiliation(s)
- Kamila M Bledzka
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Iyad H Manaserh
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica Grondolsky
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica Pfleger
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Rajika Roy
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - J Kurt Chuprun
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Sarah M Schumacher
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
22
|
Benninghoff T, Espelage L, Eickelschulte S, Zeinert I, Sinowenka I, Müller F, Schöndeling C, Batchelor H, Cames S, Zhou Z, Kotzka J, Chadt A, Al-Hasani H. The RabGAPs TBC1D1 and TBC1D4 Control Uptake of Long-Chain Fatty Acids Into Skeletal Muscle via Fatty Acid Transporter SLC27A4/FATP4. Diabetes 2020; 69:2281-2293. [PMID: 32868338 DOI: 10.2337/db20-0180] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/24/2020] [Indexed: 11/13/2022]
Abstract
The two closely related RabGTPase-activating proteins (RabGAPs) TBC1D1 and TBC1D4 play a crucial role in the regulation of GLUT4 translocation in response to insulin and contraction in skeletal muscle. In mice, deficiency in one or both RabGAPs leads to reduced insulin- and contraction-stimulated glucose uptake and to elevated fatty acid (FA) uptake and oxidation in both glycolytic and oxidative muscle fibers without altering mitochondrial copy number and the abundance of proteins for oxidative phosphorylation. Here we present evidence for a novel mechanism of skeletal muscle lipid utilization involving the two RabGAPs and the FA transporter SLC27A4/FATP4. Both RabGAPs control the uptake of saturated and unsaturated long-chain FAs (LCFAs) into skeletal muscle and knockdown (Kd) of a subset of RabGAP substrates, Rab8, Rab10, or Rab14, decreased LCFA uptake into these cells. In skeletal muscle from Tbc1d1 and Tbc1d4 knockout animals, SLC27A4/FATP4 abundance was increased and depletion of SLC27A4/FATP4 but not FAT/CD36 completely abrogated the enhanced FA oxidation in RabGAP-deficient skeletal muscle and cultivated C2C12 myotubes. Collectively, our data demonstrate that RabGAP-mediated control of skeletal muscle lipid metabolism converges with glucose metabolism at the level of downstream RabGTPases and involves regulated transport of LCFAs via SLC27A4/FATP4.
Collapse
Affiliation(s)
- Tim Benninghoff
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Lena Espelage
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Samaneh Eickelschulte
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Isabel Zeinert
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Isabelle Sinowenka
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Frank Müller
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Christina Schöndeling
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Hannah Batchelor
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Sandra Cames
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Zhou Zhou
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Jörg Kotzka
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Duesseldorf, Medical Faculty, Duesseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| |
Collapse
|
23
|
Zheng X, Arias EB, Qi NR, Saunders TL, Cartee GD. In vivo glucoregulation and tissue-specific glucose uptake in female Akt substrate 160 kDa knockout rats. PLoS One 2020; 15:e0223340. [PMID: 32053588 PMCID: PMC7018090 DOI: 10.1371/journal.pone.0223340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/26/2020] [Indexed: 01/31/2023] Open
Abstract
The Rab GTPase activating protein known as Akt substrate of 160 kDa (AS160 or TBC1D4) regulates insulin-stimulated glucose uptake in skeletal muscle, the heart, and white adipose tissue (WAT). A novel rat AS160-knockout (AS160-KO) was created with CRISPR/Cas9 technology. Because female AS160-KO versus wild type (WT) rats had not been previously evaluated, the primary objective of this study was to compare female AS160-KO rats with WT controls for multiple, important metabolism-related endpoints. Body mass and composition, physical activity, and energy expenditure were not different between genotypes. AS160-KO versus WT rats were glucose intolerant based on an oral glucose tolerance test (P<0.001) and insulin resistant based on a hyperinsulinemic-euglycemic clamp (HEC; P<0.001). Tissue glucose uptake during the HEC of female AS160-KO versus WT rats was: 1) significantly lower in epitrochlearis (P<0.05) and extensor digitorum longus (EDL; P<0.01) muscles of AS160-KO compared to WT rats; 2) not different in soleus, gastrocnemius or WAT; and 3) ~3-fold greater in the heart (P<0.05). GLUT4 protein content was reduced in AS160-KO versus WT rats in the epitrochlearis (P<0.05), EDL (P<0.05), gastrocnemius (P<0.05), soleus (P<0.05), WAT (P<0.05), and the heart (P<0.005). Insulin-stimulated glucose uptake by isolated epitrochlearis and soleus muscles was lower (P<0.001) in AS160-KO versus WT rats. Akt phosphorylation of insulin-stimulated tissues was not different between the genotypes. A secondary objective was to probe processes that might account for the genotype-related increase in myocardial glucose uptake, including glucose transporter protein abundance (GLUT1, GLUT4, GLUT8, SGLT1), hexokinase II protein abundance, and stimulation of the AMP-activated protein kinase (AMPK) pathway. None of these parameters differed between genotypes. Metabolic phenotyping in the current study revealed AS160 deficiency produced a profound glucoregulatory phenotype in female AS160-KO rats that was strikingly similar to the results previously reported in male AS160-KO rats.
Collapse
Affiliation(s)
- Xiaohua Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Edward B. Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Nathan R. Qi
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Thomas L. Saunders
- Transgenic Animal Model Core, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Gregory D. Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States of America
- Division of Genetic Medicine Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
24
|
Arias EB, Zheng X, Agrawal S, Cartee GD. Whole body glucoregulation and tissue-specific glucose uptake in a novel Akt substrate of 160 kDa knockout rat model. PLoS One 2019; 14:e0216236. [PMID: 31034517 PMCID: PMC6488193 DOI: 10.1371/journal.pone.0216236] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/16/2019] [Indexed: 11/26/2022] Open
Abstract
Akt substrate of 160 kDa (also called AS160 or TBC1D4) is a Rab GTPase activating protein and key regulator of insulin-stimulated glucose uptake which is expressed by multiple tissues, including skeletal muscle, white adipose tissue (WAT) and the heart. This study introduces a novel rat AS160-knockout (AS160-KO) model that was created using CRISPR/Cas9 technology. We compared male AS160-KO versus wildtype (WT) rats for numerous metabolism-related endpoints. Body mass, body composition, energy expenditure and physical activity did not differ between genotypes. Oral glucose intolerance was detected in AS160-KO versus WT rats (P<0.005). A hyperinsulinemic-euglycemic clamp (HEC) revealed insulin resistance for glucose infusion rate (P<0.05) with unaltered hepatic glucose production in AS160-KO versus WT rats. Genotype-effects on glucose uptake during the HEC: 1) was significantly lower in epitrochlearis (P<0.01) and extensor digitorum longus (P<0.05) of AS160-KO versus WT rats, and tended to be lower for AS160-KO versus WT rats in the soleus (P<0.06) and gastrocnemius (P<0.08); 2) tended to be greater for AS160-KO versus WT rats in white adipose tissue (P = 0.09); and 3) was significantly greater in the heart (P<0.005) of AS160-KO versus WT rats. GLUT4 protein abundance was significantly lower for AS160-KO versus WT rats in each tissue analyzed (P<0.01–0.001) except the gastrocnemius. Ex vivo insulin-stimulated glucose uptake was significantly lower (P<0.001) for AS160-KO versus WT rats in isolated epitrochlearis or soleus. Insulin-stimulated Akt phosphorylation (in vivo or ex vivo) did not differ between genotypes for any tissue tested. Ex vivo AICAR-stimulated glucose uptake by isolated epitrochlearis was significantly lower for AS160-KO versus WT rats (P<0.01) without genotype-induced alteration in AMP-activated protein phosphorylation. This unique AS160-KO rat model, which elucidated striking genotype-related modifications in glucoregulation, will enable future research aimed at understanding AS160’s roles in numerous physiological processes in response to various interventions (e.g., diet and/or exercise).
Collapse
Affiliation(s)
- Edward B. Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Xiaohua Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Swati Agrawal
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Gregory D. Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
25
|
Chen Q, Rong P, Zhu S, Yang X, Ouyang Q, Wang HY, Chen S. Targeting RalGAPα1 in skeletal muscle to simultaneously improve postprandial glucose and lipid control. SCIENCE ADVANCES 2019; 5:eaav4116. [PMID: 30989113 PMCID: PMC6459767 DOI: 10.1126/sciadv.aav4116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/12/2019] [Indexed: 05/14/2023]
Abstract
How insulin stimulates postprandial uptake of glucose and long-chain fatty acids (LCFAs) into skeletal muscle and the mechanisms by which these events are dampened in diet-induced obesity are incompletely understood. Here, we show that RalGAPα1 is a critical regulator of muscle insulin action and governs both glucose and lipid homeostasis. A high-fat diet increased RalGAPα1 protein but decreased its insulin-responsive Thr735-phosphorylation in skeletal muscle. A RalGAPα1Thr735Ala mutation impaired insulin-stimulated muscle assimilation of glucose and LCFAs and caused metabolic syndrome in mice. In contrast, skeletal muscle-specific deletion of RalGAPα1 improved postprandial glucose and lipid control. Mechanistically, these mutations of RalGAPα1 affected translocation of insulin-responsive glucose transporter GLUT4 and fatty acid translocase CD36 via RalA to affect glucose and lipid homeostasis. These data indicated RalGAPα1 as a dual-purpose target, for which we developed a peptide-blockade for improving muscle insulin sensitivity. Our findings have implications for drug discovery to combat metabolic disorders.
Collapse
|
26
|
Wu J, Cheng D, Liu L, Lv Z, Liu K. TBC1D15 affects glucose uptake by regulating GLUT4 translocation. Gene 2018; 683:210-215. [PMID: 30316925 DOI: 10.1016/j.gene.2018.10.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/06/2018] [Accepted: 10/11/2018] [Indexed: 10/28/2022]
Abstract
Glucose transport into skeletal muscle is mediated by the principal glucose transporter protein, GLUT4, which can be transported from intracellular vesicles to the cytoplasmic membrane, and the translocation of GLUT4 vesicles requires a variety of Rab proteins. Previously we reported a new type of TBC1D15 from C. plagiosum with Rab-GAP activity for Rab7. Here we reported that TBC1D15 regulated glucose uptake by affecting the translocation of GLUT4 through late endosomal pathway. When TBC1D15 was knocked out by CRISPR/Cas9, a significant reduction in 2-NBDG uptake was observed, and the total amount of GLUT4 was significantly reduced in TBC1D15-/- cells compared to that in WT cells. Furthermore, concentrated distribution of Rab7 in Lamp1-decorated late endosome/lysosome and an increase in co-localization between GLUT4 and Rab7 was observed in TBC1D15-/- cells. These results suggested that TBC1D15 served as a master regulator in GLUT4 translocation and further affected GLUT4-mediated glucose uptake.
Collapse
Affiliation(s)
- Jia Wu
- College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, China
| | - Dandan Cheng
- College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, China
| | - Li Liu
- College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, China
| | - Zhengbing Lv
- College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, China
| | - Kuancheng Liu
- College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Hangzhou 310018, China.
| |
Collapse
|
27
|
Li Z, Yue Y, Hu F, Zhang C, Ma X, Li N, Qiu L, Fu M, Chen L, Yao Z, Bilan PJ, Klip A, Niu W. Electrical pulse stimulation induces GLUT4 translocation in C 2C 12 myotubes that depends on Rab8A, Rab13, and Rab14. Am J Physiol Endocrinol Metab 2018; 314:E478-E493. [PMID: 29089333 DOI: 10.1152/ajpendo.00103.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The signals mobilizing GLUT4 to the plasma membrane in response to muscle contraction are less known than those elicited by insulin. This disparity is undoubtedly due to lack of suitable in vitro models to study skeletal muscle contraction. We generated C2C12 myotubes stably expressing HA-tagged GLUT4 (C2C12-GLUT4 HA) that contract in response to electrical pulse stimulation (EPS) and investigated molecular mechanisms regulating GLUT4 HA. EPS (60 min, 20 V, 1 Hz, 24-ms pulses at 976-ms intervals) elicited a gain in surface GLUT4 HA (GLUT4 translocation) comparably to insulin or 5-amino imidazole-4-carboxamide ribonucleotide (AICAR). A myosin II inhibitor prevented EPS-stimulated myotube contraction and reduced surface GLUT4 by 56%. EPS stimulated AMPK and CaMKII phosphorylation, and EPS-stimulated GLUT4 translocation was reduced in part by small interfering (si)RNA-mediated AMPKα1/α2 knockdown, compound C, siRNA-mediated Ca2+/calmodulin-dependent protein kinase (CaMKII)δ knockdown, or CaMKII inhibitor KN93. Key regulatory residues on the Rab-GAPs AS160 and TBC1D1 were phosphorylated in response to EPS. Stable expression of an activated form of the Rab-GAP AS160 (AS160-4A) diminished EPS- and insulin-stimulated GLUT4 translocation, suggesting regulation of GLUT4 vesicle traffic by Rab GTPases. Knockdown of each Rab8a, Rab13, or Rab14 reduced, in part, GLUT4 translocation induced by EPS, whereas only Rab8a, or Rab14 knockdown reduced the AICAR response. In conclusion, EPS involves Rab8a, Rab13, and Rab14 to elicit GLUT4 translocation but not Rab10; moreover, Rab10 and Rab13 are not engaged by AMPK activation alone. C2C12-GLUT4 HA cultures constitute a valuable in vitro model to investigate molecular mechanisms of contraction-stimulated GLUT4 translocation.
Collapse
Affiliation(s)
- Zhu Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Yingying Yue
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Fang Hu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Chang Zhang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Xiaofang Ma
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
- Central Laboratory, The Fifth Central Hospital of Tianjin , Tianjin , China
| | - Nana Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Lihong Qiu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Maolong Fu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
- Tianjin Third Central Hospital , Tianjin , China
| | - Liming Chen
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Zhi Yao
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children , Toronto, Ontario , Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children , Toronto, Ontario , Canada
| | - Wenyan Niu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Metabolic Diseases Hospital, Tianjin Medical University , Tianjin , China
| |
Collapse
|
28
|
Su Z, Deshpande V, James DE, Stöckli J. Tankyrase modulates insulin sensitivity in skeletal muscle cells by regulating the stability of GLUT4 vesicle proteins. J Biol Chem 2018; 293:8578-8587. [PMID: 29669812 DOI: 10.1074/jbc.ra117.001058] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/05/2018] [Indexed: 11/06/2022] Open
Abstract
Tankyrase 1 and 2, members of the poly(ADP-ribose) polymerase family, have previously been shown to play a role in insulin-mediated glucose uptake in adipocytes. However, their precise mechanism of action, and their role in insulin action in other cell types, such as myocytes, remains elusive. Treatment of differentiated L6 myotubes with the small molecule tankyrase inhibitor XAV939 resulted in insulin resistance as determined by impaired insulin-stimulated glucose uptake. Proteomic analysis of XAV939-treated myotubes identified down-regulation of several glucose transporter GLUT4 storage vesicle (GSV) proteins including RAB10, VAMP8, SORT1, and GLUT4. A similar effect was observed following knockdown of tankyrase 1 in L6 myotubes. Inhibition of the proteasome using MG132 rescued GSV protein levels as well as insulin-stimulated glucose uptake in XAV939-treated L6 myotubes. These studies reveal an important role for tankyrase in maintaining the stability of key GLUT4 regulatory proteins that in turn plays a role in regulating cellular insulin sensitivity.
Collapse
Affiliation(s)
- Zhiduan Su
- From the Charles Perkins Centre, School of Life and Environmental Sciences and
| | - Vinita Deshpande
- From the Charles Perkins Centre, School of Life and Environmental Sciences and
| | - David E James
- From the Charles Perkins Centre, School of Life and Environmental Sciences and .,the Sydney Medical School, University of Sydney, Sydney 2006, Australia
| | - Jacqueline Stöckli
- From the Charles Perkins Centre, School of Life and Environmental Sciences and
| |
Collapse
|
29
|
Wu J, Tao WW, Chong DY, Lai SS, Wang C, Liu Q, Zhang TY, Xue B, Li CJ. Early growth response-1 negative feedback regulates skeletal muscle postprandial insulin sensitivity via activating Ptp1b transcription. FASEB J 2018. [PMID: 29543533 DOI: 10.1096/fj.201701340r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Postprandial insulin desensitization plays a critical role in maintaining whole-body glucose homeostasis by avoiding the excessive absorption of blood glucose; however, the detailed mechanisms that underlie how the major player, skeletal muscle, desensitizes insulin action remain to be elucidated. Herein, we report that early growth response gene-1 ( Egr-1) is activated by insulin in skeletal muscle and provides feedback inhibition that regulates insulin sensitivity after a meal. The inhibition of the transcriptional activity of Egr-1 enhanced the phosphorylation of the insulin receptor (InsR) and Akt, thus increasing glucose uptake in L6 myotubes after insulin stimulation, whereas overexpression of Egr-1 decreased insulin sensitivity. Furthermore, deletion of Egr-1 in the skeletal muscle improved systemic insulin sensitivity and glucose tolerance, which resulted in lower blood glucose levels after refeeding. Mechanistic analysis demonstrated that EGR-1 inhibited InsR phosphorylation and glucose uptake in skeletal muscle by binding to the proximal promoter region of protein tyrosine phosphatase-1B (PTP1B) and directly activating transcription. PTP1B knockdown largely restored insulin sensitivity and enhanced glucose uptake, even under conditions of EGR-1 overexpression. Our results indicate that EGR-1/PTP1B signaling negatively regulates postprandial insulin sensitivity and suggest a potential therapeutic target for the prevention and treatment of excessive glucose absorption.-Wu, J., Tao, W.-W., Chong, D.-Y., Lai, S.-S., Wang, C., Liu, Q., Zhang, T.-Y., Xue, B., Li, C.-J. Early growth response-1 negative feedback regulates skeletal muscle postprandial insulin sensitivity via activating Ptp1b transcription.
Collapse
Affiliation(s)
- Jing Wu
- Medicine School of Nanjing University, Nanjing, China
| | - Wei-Wei Tao
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Dan-Yang Chong
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Shan-Shan Lai
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Chuang Wang
- Medicine School of Nanjing University, Nanjing, China
| | - Qi Liu
- Medicine School of Nanjing University, Nanjing, China
| | - Tong-Yu Zhang
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Bin Xue
- Medicine School of Nanjing University, Nanjing, China
| | - Chao-Jun Li
- Medicine School of Nanjing University, Nanjing, China.,Model Animal Research Center, Nanjing University, Nanjing, China
| |
Collapse
|
30
|
Tang W, Tang S, Wang H, Ge Z, Zhu D, Bi Y. Insulin restores UCP3 activity and decreases energy surfeit to alleviate lipotoxicity in skeletal muscle. Int J Mol Med 2017; 40:2000-2010. [PMID: 29039450 DOI: 10.3892/ijmm.2017.3169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 09/19/2017] [Indexed: 11/06/2022] Open
Abstract
An early insulin regimen ameliorates glucotoxicity but also lipotoxicity in type 2 diabetes; however, the underlying mechanism remains elusive. In the present study, we investigated the role of mitochondria in lipid regulation following early insulin administration in insulin-resistant skeletal muscle cells. Male C57BL/6 mice, fed a high-fat diet (HFD) for 8 weeks, were treated with insulin for 3 weeks, and L6 myotubes cultured with palmitate (PA) for 24 h were incubated with insulin for another 12 h. The results showed that insulin facilitated systemic glucose disposal and attenuated muscular triglyceride accumulation in vivo. Recovery of AMP-activated protein kinase (AMPK) phosphorylation, inhibition of sterol-regulated element binding protein-1c (SREBP-1c) and increased carnitine palmitoyltransferase‑1B (CPT1B) expression were observed after insulin administration. Moreover, increased ATP concentration and cellular energy charge elicited by over-nutrition were suppressed by insulin. Despite maintaining respiratory complex activities, insulin restored muscular uncoupling protein 3 (UCP3) protein expression in vitro and in vivo. By contrast, knockdown of UCP3 abrogated insulin-induced restoration of AMPK phosphorylation in vitro. Importantly, the PA-induced decrease in UCP3 was blocked by the proteasome inhibitor MG132, and insulin reduced UCP3 ubiquitination, thereby prohibiting its degradation. Our findings, focusing on energy balance, provide a mechanistic understanding of the promising effect of early insulin initiation on lipotoxicity. Insulin, by recovering UCP3 activity, alleviated energy surfeit and potentiated AMPK-mediated lipid homeostasis in skeletal muscle cells following exposure to PA and in gastrocnemius of mice fed HFD.
Collapse
Affiliation(s)
- Wenjuan Tang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Sunyinyan Tang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Zhijuan Ge
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
31
|
Chen Q, Xie B, Zhu S, Rong P, Sheng Y, Ducommun S, Chen L, Quan C, Li M, Sakamoto K, MacKintosh C, Chen S, Wang HY. A Tbc1d1 Ser231Ala-knockin mutation partially impairs AICAR- but not exercise-induced muscle glucose uptake in mice. Diabetologia 2017; 60:336-345. [PMID: 27826658 DOI: 10.1007/s00125-016-4151-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/14/2016] [Indexed: 11/30/2022]
Abstract
AIMS/HYPOTHESIS TBC1D1 (tre-2/USP6, BUB2, cdc16 domain family member 1) is a Rab GTPase-activating protein (RabGAP) that has been implicated in regulating GLUT4 trafficking. TBC1D1 can be phosphorylated by the AMP-activated protein kinase (AMPK) on Ser231, which consequently interacts with 14-3-3 proteins. Given the key role for AMPK in regulating insulin-independent muscle glucose uptake, we hypothesised that TBC1D1-Ser231 phosphorylation and/or 14-3-3 binding may mediate AMPK-governed glucose homeostasis. METHODS Whole-body glucose homeostasis and muscle glucose uptake were assayed in mice bearing a Tbc1d1 Ser231Ala-knockin mutation or harbouring skeletal muscle-specific Ampkα1/α2 (also known as Prkaa1/2) double-knockout mutations in response to an AMPK-activating agent, 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR). Exercise-induced muscle glucose uptake and exercise capacity were also determined in the Tbc1d1 Ser231Ala-knockin mice. RESULTS Skeletal muscle-specific deletion of Ampkα1/a2 in mice prevented AICAR-induced hypoglycaemia and muscle glucose uptake. The Tbc1d1 Ser231Ala-knockin mutation also attenuated the glucose-lowering effect of AICAR in mice. Glucose uptake and cell surface GLUT4 content were significantly lower in muscle isolated from the Tbc1d1 Ser231Ala-knockin mice upon stimulation with a submaximal dose of AICAR. However, this Tbc1d1 Ser231Ala-knockin mutation neither impaired exercise-induced muscle glucose uptake nor affected exercise capacity in mice. CONCLUSIONS/INTERPRETATION TBC1D1-Ser231 phosphorylation and/or 14-3-3 binding partially mediates AMPK-governed glucose homeostasis and muscle glucose uptake in a context-dependent manner.
Collapse
Affiliation(s)
- Qiaoli Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Bingxian Xie
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Sangsang Zhu
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Ping Rong
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Yang Sheng
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Serge Ducommun
- Nestlé Institute of Health Sciences SA, Campus EPFL, Quartier de l'Innovation, Bâtiment G, Lausanne, Switzerland
| | - Liang Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Chao Quan
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Min Li
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China
| | - Kei Sakamoto
- Nestlé Institute of Health Sciences SA, Campus EPFL, Quartier de l'Innovation, Bâtiment G, Lausanne, Switzerland
| | - Carol MacKintosh
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China.
- Collaborative Innovation Center of Genetics and Development, Shanghai, China.
| | - Hong Yu Wang
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Pukou District, Nanjing, 210061, China.
- Collaborative Innovation Center of Genetics and Development, Shanghai, China.
| |
Collapse
|