51
|
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: 3.6] [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
|
52
|
Normal increases in insulin-stimulated glucose uptake after ex vivo contraction in neuronal nitric oxide synthase mu (nNOSμ) knockout mice. Pflugers Arch 2019; 471:961-969. [DOI: 10.1007/s00424-019-02268-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 11/25/2022]
|
53
|
Mo Z, Li L, Yu H, Wu Y, Li H. Coumarins ameliorate diabetogenic action of dexamethasone via Akt activation and AMPK signaling in skeletal muscle. J Pharmacol Sci 2019; 139:151-157. [PMID: 30733181 DOI: 10.1016/j.jphs.2019.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 12/29/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids are widely prescribed for lots of pathological conditions, however, can produce 'Cushingoid' side effects including central obesity, glucose intolerance, insulin resistance and so forth. Our study is intended to investigate the improving effects of coumarins on diabetogenic action of dexamethasone in vivo and in vitro and elucidate potential mechanisms. ICR mice treated with dexamethasone for 21 days exhibited decreased body weight, increased blood glucose and impaired glucose tolerance, which were prevented by fraxetin (40 mg/kg/day), esculin (40 mg/kg/day) and osthole (20 mg/kg/day), respectively. Esculin, fraxetin and osthole also could promote glucose uptake in normal C2C12 myotubes, and improve insulin resistance in myotubes induced by dexamethasone. Western blotting results indicated that esculin, fraxetin and osthole could boost Akt activation, stimulate GLUT4 translocation, thus alleviate insulin resistance. Esculin and osthole also could activate AMPK, thereby phosphorylate TBC1D1 at Ser237, and consequently ameliorate diabetogenic action of dexamethasone. Our study indicates coumarins as potential anti-diabetic candidates or leading compounds for drug development.
Collapse
Affiliation(s)
- Zejun Mo
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Linghuan Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Haiwen Yu
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Yingqi Wu
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Hanbing Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; Section of Endocrinology, School of Medicine, Yale University, New Haven 06520, USA.
| |
Collapse
|
54
|
Chemical denervation using botulinum toxin increases Akt expression and reduces submaximal insulin-stimulated glucose transport in mouse muscle. Cell Signal 2019; 53:224-233. [DOI: 10.1016/j.cellsig.2018.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 12/31/2022]
|
55
|
Diallo K, Oppong AK, Lim GE. Can 14-3-3 proteins serve as therapeutic targets for the treatment of metabolic diseases? Pharmacol Res 2019; 139:199-206. [DOI: 10.1016/j.phrs.2018.11.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022]
|
56
|
Mugabo Y, Lim GE. Scaffold Proteins: From Coordinating Signaling Pathways to Metabolic Regulation. Endocrinology 2018; 159:3615-3630. [PMID: 30204866 PMCID: PMC6180900 DOI: 10.1210/en.2018-00705] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/05/2018] [Indexed: 01/13/2023]
Abstract
Among their pleiotropic functions, scaffold proteins are required for the accurate coordination of signaling pathways. It has only been within the past 10 years that their roles in glucose homeostasis and metabolism have emerged. It is well appreciated that changes in the expression or function of signaling effectors, such as receptors or kinases, can influence the development of chronic diseases such as diabetes and obesity. However, little is known regarding whether scaffolds have similar roles in the pathogenesis of metabolic diseases. In general, scaffolds are often underappreciated in the context of metabolism or metabolic diseases. In the present review, we discuss various scaffold proteins and their involvement in signaling pathways related to metabolism and metabolic diseases. The aims of the present review were to highlight the importance of scaffold proteins and to raise awareness of their physiological contributions. A thorough understanding of how scaffolds influence metabolism could aid in the discovery of novel therapeutic approaches to treat chronic conditions, such as diabetes, obesity, and cardiovascular disease, for which the incidence of all continue to increase at alarming rates.
Collapse
Affiliation(s)
- Yves Mugabo
- Cardiometabolic Axis, Centre de Recherche de Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Montréal Diabetes Research Centre, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Gareth E Lim
- Cardiometabolic Axis, Centre de Recherche de Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Montréal Diabetes Research Centre, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
57
|
Wang H, Arias EB, Pataky MW, Goodyear LJ, Cartee GD. Postexercise improvement in glucose uptake occurs concomitant with greater γ3-AMPK activation and AS160 phosphorylation in rat skeletal muscle. Am J Physiol Endocrinol Metab 2018; 315:E859-E871. [PMID: 30130149 PMCID: PMC6293165 DOI: 10.1152/ajpendo.00020.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A single exercise session can increase insulin-stimulated glucose uptake (GU) by skeletal muscle, concomitant with greater Akt substrate of 160 kDa (AS160) phosphorylation on Akt-phosphosites (Thr642 and Ser588) that regulate insulin-stimulated GU. Recent research using mouse skeletal muscle suggested that ex vivo 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) or electrically stimulated contractile activity-inducing increased γ3-AMPK activity and AS160 phosphorylation on a consensus AMPK-motif (Ser704) resulted in greater AS160 Thr642 phosphorylation and GU by insulin-stimulated muscle. Our primary goal was to determine whether in vivo exercise that increases insulin-stimulated GU in rat skeletal muscle would also increase γ3-AMPK activity and AS160 site-selective phosphorylation (Ser588, Thr642, and Ser704) immediately postexercise (IPEX) and/or 3 h postexercise (3hPEX). Epitrochlearis muscles isolated from sedentary and exercised (2-h swim exercise; studied IPEX and 3hPEX) rats were incubated with 2-deoxyglucose to determine GU (without insulin at IPEX; without or with insulin at 3hPEX). Muscles were also assessed for γ1-AMPK activity, γ3-AMPK activity, phosphorylated AMPK (pAMPK), and phosphorylated AS160 (pAS160). IPEX versus sedentary had greater γ3-AMPK activity, pAS160 (Ser588, Thr642, Ser704), and GU with unaltered γ1-AMPK activity. 3hPEX versus sedentary had greater γ3-AMPK activity, pAS160 Ser704, and GU with or without insulin; greater pAS160 Thr642 only with insulin; and unaltered γ1-AMPK activity. These results using an in vivo exercise protocol that increased insulin-stimulated GU in rat skeletal muscle are consistent with the hypothesis that in vivo exercise-induced enhancement of γ3-AMPK activation and AS160 Ser704 IPEX and 3hPEX are important for greater pAS160 Thr642 and enhanced insulin-stimulated GU by skeletal muscle.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Mark W Pataky
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
| | - Laurie J Goodyear
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan , Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
- Institute of Gerontology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
58
|
Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev 2018; 98:2133-2223. [PMID: 30067154 PMCID: PMC6170977 DOI: 10.1152/physrev.00063.2017] [Citation(s) in RCA: 1460] [Impact Index Per Article: 243.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/15/2022] Open
Abstract
The 1921 discovery of insulin was a Big Bang from which a vast and expanding universe of research into insulin action and resistance has issued. In the intervening century, some discoveries have matured, coalescing into solid and fertile ground for clinical application; others remain incompletely investigated and scientifically controversial. Here, we attempt to synthesize this work to guide further mechanistic investigation and to inform the development of novel therapies for type 2 diabetes (T2D). The rational development of such therapies necessitates detailed knowledge of one of the key pathophysiological processes involved in T2D: insulin resistance. Understanding insulin resistance, in turn, requires knowledge of normal insulin action. In this review, both the physiology of insulin action and the pathophysiology of insulin resistance are described, focusing on three key insulin target tissues: skeletal muscle, liver, and white adipose tissue. We aim to develop an integrated physiological perspective, placing the intricate signaling effectors that carry out the cell-autonomous response to insulin in the context of the tissue-specific functions that generate the coordinated organismal response. First, in section II, the effectors and effects of direct, cell-autonomous insulin action in muscle, liver, and white adipose tissue are reviewed, beginning at the insulin receptor and working downstream. Section III considers the critical and underappreciated role of tissue crosstalk in whole body insulin action, especially the essential interaction between adipose lipolysis and hepatic gluconeogenesis. The pathophysiology of insulin resistance is then described in section IV. Special attention is given to which signaling pathways and functions become insulin resistant in the setting of chronic overnutrition, and an alternative explanation for the phenomenon of ‟selective hepatic insulin resistanceˮ is presented. Sections V, VI, and VII critically examine the evidence for and against several putative mediators of insulin resistance. Section V reviews work linking the bioactive lipids diacylglycerol, ceramide, and acylcarnitine to insulin resistance; section VI considers the impact of nutrient stresses in the endoplasmic reticulum and mitochondria on insulin resistance; and section VII discusses non-cell autonomous factors proposed to induce insulin resistance, including inflammatory mediators, branched-chain amino acids, adipokines, and hepatokines. Finally, in section VIII, we propose an integrated model of insulin resistance that links these mediators to final common pathways of metabolite-driven gluconeogenesis and ectopic lipid accumulation.
Collapse
Affiliation(s)
- Max C Petersen
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
59
|
Bazzano MV, Sarrible GB, Martinez N, Berón de Astrada M, Elia EM. Obesity alters the uterine environment before pregnancy. J Nutr Biochem 2018; 62:181-191. [PMID: 30300837 DOI: 10.1016/j.jnutbio.2018.09.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/03/2018] [Accepted: 09/12/2018] [Indexed: 11/30/2022]
Abstract
Obesity is a metabolic disorder that predisposes to numerous diseases and has become a major global public health concern. Cafeteria diet (CAF) is the animal model used for the study of obesity that more closely reflects Western diet habits. Previously, we described that CAF administration for 60 days induces obesity in female rats and their fetuses develop macrosomia. Given that, in our model, rats are not genetically modified and that obese mothers were fed standard chow during pregnancy, the aim of the current study was to test the hypothesis that obesity alters the intrauterine environment prior to pregnancy, and this may explain the exacerbated fetal weight gain. We found that uteri from obese rats during the estrous phase developed insulin resistance through mechanisms that involve the induction of uterine hypoxia and the down-regulation of the insulin receptor gene. Moreover, uterine cell proliferation was induced by obesity concomitantly with the reduction in the uterine contractile response to a β2 AR agonist, salbutamol, and this may be consequence of the down-regulation in the uterine β2 AR expression. We conclude that CAF-induced obesity alters the uterine environment in rats during the estrous phase and may cause the fetal macrosomia previously described by us in obese animals. The lower sensitivity of the uterus to a relaxation stimulus (salbutamol) is not a minor fact given that for implantation to occur the uterus must be relaxed for embryo nidation. Thus, the alteration in the uterine quiescence may impair implantation and, consequently, the foregoing pregnancy.
Collapse
Affiliation(s)
- María Victoria Bazzano
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA-Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires, Argentina
| | - Gisela Belén Sarrible
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA-Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires, Argentina; UBA, FCEN, Departamento de Biodiversidad y Bilogía Experimental, Buenos Aires, Argentina
| | - Nora Martinez
- UBA, Facultad de Medicina, Buenos Aires, Argentina; CONICET-UBA- Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO), Buenos Aires, Argentina
| | - Martín Berón de Astrada
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA-Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires, Argentina; UBA, FCEN, Departamento de Fisiología, Biología Molecular y Celular, Buenos Aires, Argentina
| | - Evelin Mariel Elia
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA-Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires, Argentina; UBA, FCEN, Departamento de Biodiversidad y Bilogía Experimental, Buenos Aires, Argentina.
| |
Collapse
|
60
|
Liu Y, Neumann D, Glatz JFC, Luiken JJFP. Molecular mechanism of lipid-induced cardiac insulin resistance and contractile dysfunction. Prostaglandins Leukot Essent Fatty Acids 2018; 136:131-141. [PMID: 27372802 DOI: 10.1016/j.plefa.2016.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/10/2016] [Indexed: 01/04/2023]
Abstract
Long-chain fatty acids are the main cardiac substrates from which ATP is generated continually to serve the high energy demand and sustain the normal function of the heart. Under healthy conditions, fatty acid β-oxidation produces 50-70% of the energy demands with the remainder largely accounted for by glucose. Chronically increased dietary lipid supply often leads to excess lipid accumulation in the heart, which is linked to a variety of maladaptive phenomena, such as insulin resistance, cardiac hypertrophy and contractile dysfunction. CD36, the predominant cardiac fatty acid transporter, has a key role in setting the heart on a road to contractile dysfunction upon the onset of chronic lipid oversupply by translocating to the cell surface and opening the cellular 'doors' for fatty acids. The sequence of events after the CD36-mediated myocellular lipid accumulation is less understood, but in general it has been accepted that the excessively imported lipids cause insulin resistance, which in turn leads to contractile dysfunction. There are several gaps of knowledge in this proposed order of events which this review aims to discuss. First, the molecular mechanisms underlying lipid-induced insulin resistance are not yet completely disclosed. Specifically, several mediators have been proposed, such as diacylglycerols, ceramides, peroxisome proliferator-activated receptors (PPAR), inflammatory kinases and reactive oxygen species (ROS), but their relative contributions to the onset of insulin resistance and their putatively synergistic actions are topics of controversy. Second, there are also pieces of evidence that lipids can induce contractile dysfunction independently of insulin resistance. Perhaps, a more integrative view is needed, in which several lipid-induced pathways operate synergistically or in parallel to induce contractile dysfunction. Unraveling of these processes is expected to be important in designing effective therapeutic strategies to protect the lipid-overloaded heart.
Collapse
Affiliation(s)
- Yilin Liu
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Dietbert Neumann
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Jan F C Glatz
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands.
| | - Joost J F P Luiken
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
61
|
Li S, Liu L, He G, Wu J. Molecular targets and mechanisms of bioactive peptides against metabolic syndromes. Food Funct 2018; 9:42-52. [PMID: 29188845 DOI: 10.1039/c7fo01323j] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bioactive peptides are present in all living organisms and have critical roles ranging from protection against infection as the key element of innate immunity, regulating blood pressure and glucose levels, to reducing signs of ageing by killing senescent cells. Bioactive peptides are also encrypted within food protein sequences that can be released during proteolysis or food processing. These specific food protein fragments are reported to have potential for improving human health and preventing metabolic diseases through their impact on inflammation, blood pressure, obesity, and type-2 diabetes. This review mainly focuses on the molecular targets and the underlying mechanisms of bioactive peptides against various metabolic syndromes including inflammation, high blood pressure, obesity, and type-2 diabetes, to provide new insights and perspectives on the potential of bioactive peptides for management of metabolic syndromes.
Collapse
Affiliation(s)
- Shanshan Li
- College of Biosystems Engineering and Food Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | | | | | | |
Collapse
|
62
|
Regulation of RabGAPs involved in insulin action. Biochem Soc Trans 2018; 46:683-690. [PMID: 29784647 DOI: 10.1042/bst20170479] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 12/31/2022]
Abstract
Rab (Ras-related proteins in brain) GTPases are key proteins responsible for a multiplicity of cellular trafficking processes. Belonging to the family of monomeric GTPases, they are regulated by cycling between their active GTP-bound and inactive GDP-bound conformations. Despite possessing a slow intrinsic GTP hydrolysis activity, Rab proteins rely on RabGAPs (Rab GTPase-activating proteins) that catalyze GTP hydrolysis and consequently inactivate the respective Rab GTPases. Two related RabGAPs, TBC1D1 and TBC1D4 (=AS160) have been described to be associated with obesity-related traits and type 2 diabetes in both mice and humans. Inactivating mutations of TBC1D1 and TBC1D4 lead to substantial changes in trafficking and subcellular distribution of the insulin-responsive glucose transporter GLUT4, and to subsequent alterations in energy substrate metabolism. The activity of the RabGAPs is controlled through complex phosphorylation events mediated by protein kinases including AKT and AMPK, and by putative regulatory interaction partners. However, the dynamics and downstream events following phosphorylation are not well understood. This review focuses on the specific role and regulation of TBC1D1 and TBC1D4 in insulin action.
Collapse
|
63
|
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: 4.5] [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
|
64
|
Serum Is Not Necessary for Prior Pharmacological Activation of AMPK to Increase Insulin Sensitivity of Mouse Skeletal Muscle. Int J Mol Sci 2018; 19:ijms19041201. [PMID: 29662023 PMCID: PMC5979416 DOI: 10.3390/ijms19041201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
Exercise, contraction, and pharmacological activation of AMP-activated protein kinase (AMPK) by 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) have all been shown to increase muscle insulin sensitivity for glucose uptake. Intriguingly, improvements in insulin sensitivity following contraction of isolated rat and mouse skeletal muscle and prior AICAR stimulation of isolated rat skeletal muscle seem to depend on an unknown factor present in serum. One study recently questioned this requirement of a serum factor by showing serum-independency with muscle from old rats. Whether a serum factor is necessary for prior AICAR stimulation to increase insulin sensitivity of mouse skeletal muscle is not known. Therefore, we investigated the necessity of serum for this effect of AICAR in mouse skeletal muscle. We found that the ability of prior AICAR stimulation to improve insulin sensitivity of mouse skeletal muscle did not depend on the presence of serum during AICAR stimulation. Although prior AICAR stimulation did not enhance proximal insulin signaling, insulin-stimulated phosphorylation of Tre-2/BUB2/CDC16- domain family member 4 (TBC1D4) Ser711 was greater in prior AICAR-stimulated muscle compared to all other groups. These results imply that the presence of a serum factor is not necessary for prior AMPK activation by AICAR to enhance insulin sensitivity of mouse skeletal muscle.
Collapse
|
65
|
Zhang X, Hiam D, Hong YH, Zulli A, Hayes A, Rattigan S, McConell GK. Nitric oxide is required for the insulin sensitizing effects of contraction in mouse skeletal muscle. J Physiol 2017; 595:7427-7439. [PMID: 29071734 DOI: 10.1113/jp275133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/18/2017] [Indexed: 01/15/2023] Open
Abstract
KEY POINTS People with insulin resistance or type 2 diabetes can substantially increase their skeletal muscle glucose uptake during exercise and insulin sensitivity after exercise. Skeletal muscle nitric oxide (NO) is important for glucose uptake during exercise, although how prior exercise increases insulin sensitivity is unclear. In the present study, we examined whether NO is necessary for normal increases in skeletal muscle insulin sensitivity after contraction ex vivo in mouse muscle. The present study uncovers, for the first time, a novel role for NO in the insulin sensitizing effects of ex vivo contraction, which is independent of blood flow. ABSTRACT The factors regulating the increase in skeletal muscle insulin sensitivity after exercise are unclear. We examined whether nitric oxide (NO) is required for the increase in insulin sensitivity after ex vivo contractions. Isolated C57BL/6J mouse EDL muscles were contracted for 10 min or remained at rest (basal) with or without the NO synthase (NOS) inhibition (NG -monomethyl-l-arginine; l-NMMA; 100 μm). Then, 3.5 h post contraction/basal, muscles were exposed to saline or insulin (120 μU ml-1 ) with or without l-NMMA during the last 30 min. l-NMMA had no effect on basal skeletal muscle glucose uptake. The increase in muscle glucose uptake with insulin (57%) was significantly (P < 0.05) greater after prior contraction (140% increase). NOS inhibition during the contractions had no effect on this insulin-sensitizing effect of contraction, whereas NOS inhibition during insulin prevented the increase in skeletal muscle insulin sensitivity post-contraction. Soluble guanylate cyclase inhibition, protein kinase G (PKG) inhibition or cyclic nucleotide phosphodiesterase inhibition each had no effect on the insulin-sensitizing effect of prior contraction. In conclusion, NO is required for increases in insulin sensitivity several hours after contraction of mouse skeletal muscle via a cGMP/PKG independent pathway.
Collapse
Affiliation(s)
- Xinmei Zhang
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Danielle Hiam
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia
| | - Yet-Hoi Hong
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia
| | - Anthony Zulli
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Alan Hayes
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia.,College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Stephen Rattigan
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Glenn K McConell
- Institute of Sport, Exercise and Active Living (ISEAL), College of Sport and Exercise Science, Victoria University, Melbourne, VIC, Australia.,College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| |
Collapse
|
66
|
Arias EB, Wang H, Cartee GD. Akt substrate of 160 kDa dephosphorylation rate is reduced in insulin-stimulated rat skeletal muscle after acute exercise. Physiol Res 2017; 67:143-147. [PMID: 29137480 DOI: 10.33549/physiolres.933591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Because greater Akt substrate of 160 kDa (AS160) phosphorylation has been reported in insulin-stimulated skeletal muscles without improved Akt activation several hours post-exercise, we hypothesized that prior exercise would result in attenuated AS160 dephosphorylation in insulin-stimulated rat skeletal muscle. Epitrochlearis muscles were isolated from rats that were sedentary (SED) or exercised 3 h earlier (3 h post-exercise; 3hPEX). Paired muscles were incubated with [(3)H]-2-deoxyglucose (2-DG) without insulin or with insulin. Lysates from other insulin-stimulated muscles from SED or 3hPEX rats were evaluated using AS160(Thr642) and AS160(Ser588) dephosphorylation assays. Prior exercise led to greater 2-DG uptake concomitant with greater AS160(Thr642) phosphorylation and a non-significant trend (P=0.087) for greater AS160(Ser588). Prior exercise also reduced AS160(Thr642) and AS160(Ser588) dephosphorylation rates. These results support the idea that attenuated AS160 dephosphorylation may favor greater AS160 phosphorylation post-exercise.
Collapse
Affiliation(s)
- E B Arias
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA.
| | | | | |
Collapse
|
67
|
Tunduguru R, Zhang J, Aslamy A, Salunkhe VA, Brozinick JT, Elmendorf JS, Thurmond DC. The actin-related p41ARC subunit contributes to p21-activated kinase-1 (PAK1)-mediated glucose uptake into skeletal muscle cells. J Biol Chem 2017; 292:19034-19043. [PMID: 28972183 DOI: 10.1074/jbc.m117.801340] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/21/2017] [Indexed: 02/04/2023] Open
Abstract
Defects in translocation of the glucose transporter GLUT4 are associated with peripheral insulin resistance, preclinical diabetes, and progression to type 2 diabetes. GLUT4 recruitment to the plasma membrane of skeletal muscle cells requires F-actin remodeling. Insulin signaling in muscle requires p21-activated kinase-1 (PAK1), whose downstream signaling triggers actin remodeling, which promotes GLUT4 vesicle translocation and glucose uptake into skeletal muscle cells. Actin remodeling is a cyclic process, and although PAK1 is known to initiate changes to the cortical actin-binding protein cofilin to stimulate the depolymerizing arm of the cycle, how PAK1 might trigger the polymerizing arm of the cycle remains unresolved. Toward this, we investigated whether PAK1 contributes to the mechanisms involving the actin-binding and -polymerizing proteins neural Wiskott-Aldrich syndrome protein (N-WASP), cortactin, and ARP2/3 subunits. We found that the actin-polymerizing ARP2/3 subunit p41ARC is a PAK1 substrate in skeletal muscle cells. Moreover, co-immunoprecipitation experiments revealed that insulin stimulates p41ARC phosphorylation and increases its association with N-WASP coordinately with the associations of N-WASP with cortactin and actin. Importantly, all of these associations were ablated by the PAK inhibitor IPA3, suggesting that PAK1 activation lies upstream of these actin-polymerizing complexes. Using the N-WASP inhibitor wiskostatin, we further demonstrated that N-WASP is required for localized F-actin polymerization, GLUT4 vesicle translocation, and glucose uptake. These results expand the model of insulin-stimulated glucose uptake in skeletal muscle cells by implicating p41ARC as a new component of the insulin-signaling cascade and connecting PAK1 signaling to N-WASP-cortactin-mediated actin polymerization and GLUT4 vesicle translocation.
Collapse
Affiliation(s)
- Ragadeepthi Tunduguru
- From the Departments of Biochemistry and Molecular Biology and.,the Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute of the City of Hope, Duarte, California 91010, and
| | - Jing Zhang
- the Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute of the City of Hope, Duarte, California 91010, and
| | - Arianne Aslamy
- the Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute of the City of Hope, Duarte, California 91010, and.,Cellular and Integrative Physiology, Center for Diabetes and Metabolic Diseases,Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Vishal A Salunkhe
- the Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute of the City of Hope, Duarte, California 91010, and
| | | | - Jeffrey S Elmendorf
- From the Departments of Biochemistry and Molecular Biology and.,Cellular and Integrative Physiology, Center for Diabetes and Metabolic Diseases,Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Debbie C Thurmond
- From the Departments of Biochemistry and Molecular Biology and .,the Department of Molecular and Cellular Endocrinology, Diabetes and Metabolism Research Institute and Beckman Research Institute of the City of Hope, Duarte, California 91010, and.,Cellular and Integrative Physiology, Center for Diabetes and Metabolic Diseases,Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
68
|
Chen Q, Rong P, Xu D, Zhu S, Chen L, Xie B, Du Q, Quan C, Sheng Y, Zhao TJ, Li P, Wang HY, Chen S. Rab8a Deficiency in Skeletal Muscle Causes Hyperlipidemia and Hepatosteatosis by Impairing Muscle Lipid Uptake and Storage. Diabetes 2017; 66:2387-2399. [PMID: 28696211 DOI: 10.2337/db17-0077] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 06/21/2017] [Indexed: 11/13/2022]
Abstract
Skeletal muscle absorbs long-chain fatty acids (LCFAs) that are either oxidized in mitochondria or temporarily stored as triglycerides in lipid droplets (LDs). So far, it is still not fully understood how lipid uptake and storage are regulated in muscle and whether these are important for whole-body lipid homeostasis. Here we show that the small GTPase Rab8a regulates lipid uptake and storage in skeletal muscle. Muscle-specific Rab8a deletion caused hyperlipidemia and exacerbated hepatosteatosis induced by a high-fat diet. Mechanistically, Rab8a deficiency decreased LCFA entry into skeletal muscle and inhibited LD fusion in muscle cells. Consequently, blood lipid levels were elevated and stimulated hepatic mammalian target of rapamycin, which enhanced hepatosteatosis by upregulating hepatic lipogenesis and cholesterol biosynthesis. Our results demonstrate the significance of lipid uptake and storage in muscle in regulating whole-body lipid homeostasis, and they shed light on the roles of skeletal muscle in the pathogenesis of hyperlipidemia and hepatosteatosis.
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 University, Pukou District, Nanjing, China
| | - Ping Rong
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Dijin Xu
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Sangsang Zhu
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Liang Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Bingxian Xie
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Qian Du
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Chao Quan
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Yang Sheng
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Tong-Jin Zhao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Peng Li
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 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 University, Pukou District, Nanjing, China
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study and State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| |
Collapse
|
69
|
Molecular mechanisms of ROS production and oxidative stress in diabetes. Biochem J 2017; 473:4527-4550. [PMID: 27941030 DOI: 10.1042/bcj20160503c] [Citation(s) in RCA: 536] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 12/18/2022]
Abstract
Oxidative stress and chronic inflammation are known to be associated with the development of metabolic diseases, including diabetes. Oxidative stress, an imbalance between oxidative and antioxidative systems of cells and tissues, is a result of over production of oxidative-free radicals and associated reactive oxygen species (ROS). One outcome of excessive levels of ROS is the modification of the structure and function of cellular proteins and lipids, leading to cellular dysfunction including impaired energy metabolism, altered cell signalling and cell cycle control, impaired cell transport mechanisms and overall dysfunctional biological activity, immune activation and inflammation. Nutritional stress, such as that caused by excess high-fat and/or carbohydrate diets, promotes oxidative stress as evident by increased lipid peroxidation products, protein carbonylation and decreased antioxidant status. In obesity, chronic oxidative stress and associated inflammation are the underlying factors that lead to the development of pathologies such as insulin resistance, dysregulated pathways of metabolism, diabetes and cardiovascular disease through impaired signalling and metabolism resulting in dysfunction to insulin secretion, insulin action and immune responses. However, exercise may counter excessive levels of oxidative stress and thus improve metabolic and inflammatory outcomes. In the present article, we review the cellular and molecular origins and significance of ROS production, the molecular targets and responses describing how oxidative stress affects cell function including mechanisms of insulin secretion and action, from the point of view of possible application of novel diabetic therapies based on redox regulation.
Collapse
|
70
|
Obesity alters the ovarian glucidic homeostasis disrupting the reproductive outcome of female rats. J Nutr Biochem 2017; 42:194-202. [DOI: 10.1016/j.jnutbio.2017.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 12/16/2016] [Accepted: 01/14/2017] [Indexed: 12/27/2022]
|
71
|
Kawamoto E, Koshinaka K, Yoshimura T, Masuda H, Kawanaka K. Immobilization rapidly induces muscle insulin resistance together with the activation of MAPKs (JNK and p38) and impairment of AS160 phosphorylation. Physiol Rep 2017; 4:4/15/e12876. [PMID: 27482072 PMCID: PMC4985544 DOI: 10.14814/phy2.12876] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/06/2016] [Indexed: 11/24/2022] Open
Abstract
Acute short‐duration physical inactivity induces the development of insulin resistance for glucose uptake in skeletal muscle. We examined the possibility that inactivity rapidly induces muscle insulin resistance via the excessive activation of proinflammatory/stress pathways including those of IKK/IκB/NF‐κB, JNK, and p38 MAPK. We also examined the other possibility that inactivity‐induced rapid development of insulin resistance is associated with reduced phosphorylation of AS160, the most distal insulin‐signaling protein that have been linked to the regulation of glucose uptake. Male Wistar rats were subjected to unilateral hindlimb immobilization for 6 h. At the end of the immobilization, the soleus muscles from both immobilized and contralateral non‐immobilized hindlimbs were dissected out. Immobilization decreased insulin‐stimulated 2‐deoxyglucose uptake in rat soleus muscle within 6 h. This rapid development of insulin resistance was accompanied by elevated phosphorylation of both JNK and p38 (commonly used indicator of JNK and p38 pathway activity, respectively). In addition, the abundance of SPT2, a rate‐limiting enzyme regulating ceramide biosynthesis, was increased in immobilized muscle. Immobilization did not alter the abundance of IκBα (commonly used indicator of IKK/IκB/NF‐κB pathway activity). The basal phosphorylation of AS160 at Thr642 and Ser588 was decreased together with the development of insulin resistance. These results suggest the possibility that inactivity‐induced rapid development of insulin resistance in immobilized muscle is related to enhanced activation of JNK and/or p38. Elevated ceramide biosynthesis pathway may contribute to this activation. Our results also indicate that decreased basal phosphorylation of AS160 may be involved in inactivity‐induced insulin resistance.
Collapse
Affiliation(s)
- Emi Kawamoto
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan Department of Materials Engineering, Nagaoka National College of Technology, Nagaoka, Japan
| | - Keiichi Koshinaka
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan
| | - Tatsuhiko Yoshimura
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan
| | - Hiroyuki Masuda
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan
| | - Kentaro Kawanaka
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
72
|
Kjøbsted R, Munk-Hansen N, Birk JB, Foretz M, Viollet B, Björnholm M, Zierath JR, Treebak JT, Wojtaszewski JFP. Enhanced Muscle Insulin Sensitivity After Contraction/Exercise Is Mediated by AMPK. Diabetes 2017; 66:598-612. [PMID: 27797909 DOI: 10.2337/db16-0530] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/24/2016] [Indexed: 12/11/2022]
Abstract
Earlier studies have demonstrated that muscle insulin sensitivity to stimulate glucose uptake is enhanced several hours after an acute bout of exercise. Using AICAR, we recently demonstrated that prior activation of AMPK is sufficient to increase insulin sensitivity in mouse skeletal muscle. Here we aimed to determine whether activation of AMPK is also a prerequisite for the ability of muscle contraction to increase insulin sensitivity. We found that prior in situ contraction of m. extensor digitorum longus (EDL) and treadmill exercise increased muscle and whole-body insulin sensitivity in wild-type (WT) mice, respectively. These effects were not found in AMPKα1α2 muscle-specific knockout mice. Prior in situ contraction did not increase insulin sensitivity in m. soleus from either genotype. Improvement in muscle insulin sensitivity was not associated with enhanced glycogen synthase activity or proximal insulin signaling. However, in WT EDL muscle, prior in situ contraction enhanced insulin-stimulated phosphorylation of TBC1D4 Thr649 and Ser711 Such findings are also evident in prior exercised and insulin-sensitized human skeletal muscle. Collectively, our data suggest that the AMPK-TBC1D4 signaling axis is likely mediating the improved muscle insulin sensitivity after contraction/exercise and illuminates an important and physiologically relevant role of AMPK in skeletal muscle.
Collapse
Affiliation(s)
- Rasmus Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nanna Munk-Hansen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jesper B Birk
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Marc Foretz
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie Björnholm
- Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jonas T Treebak
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
73
|
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: 4.1] [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
|
74
|
Zheng X, Cartee GD. Insulin-induced Effects on the Subcellular Localization of AKT1, AKT2 and AS160 in Rat Skeletal Muscle. Sci Rep 2016; 6:39230. [PMID: 27966646 PMCID: PMC5155274 DOI: 10.1038/srep39230] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/18/2016] [Indexed: 12/02/2022] Open
Abstract
AKT1 and AKT2, the AKT isoforms that are highly expressed in skeletal muscle, have distinct and overlapping functions, with AKT2 more important for insulin-stimulated glucose metabolism. In adipocytes, AKT2 versus AKT1 has greater susceptibility for insulin-mediated redistribution from cytosolic to membrane localization, and insulin also causes subcellular redistribution of AKT Substrate of 160 kDa (AS160), an AKT2 substrate and crucial mediator of insulin-stimulated glucose transport. Although skeletal muscle is the major tissue for insulin-mediated glucose disposal, little is known about AKT1, AKT2 or AS160 subcellular localization in skeletal muscle. The major aim of this study was to determine insulin’s effects on the subcellular localization and phosphorylation of AKT1, AKT2 and AS160 in skeletal muscle. Rat skeletal muscles were incubated ex vivo ± insulin, and differential centrifugation was used to isolate cytosolic and membrane fractions. The results revealed that: 1) insulin increased muscle membrane localization of AKT2, but not AKT1; 2) insulin increased AKT2 phosphorylation in the cytosol and membrane fractions; 3) insulin increased AS160 localization to the cytosol and membranes; and 4) insulin increased AS160 phosphorylation in the cytosol, but not membranes. These results demonstrate distinctive insulin effects on the subcellular redistribution of AKT2 and its substrate AS160 in skeletal muscle.
Collapse
Affiliation(s)
- Xiaohua Zheng
- 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
|
75
|
Xie B, Chen Q, Chen L, Sheng Y, Wang HY, Chen S. The Inactivation of RabGAP Function of AS160 Promotes Lysosomal Degradation of GLUT4 and Causes Postprandial Hyperglycemia and Hyperinsulinemia. Diabetes 2016; 65:3327-3340. [PMID: 27554475 DOI: 10.2337/db16-0416] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/16/2016] [Indexed: 11/13/2022]
Abstract
The AS160 (Akt substrate of 160 kDa) is a Rab-GTPase activating protein (RabGAP) with several other functional domains, and its deficiency in mice or human patients lowers GLUT4 protein levels and causes severe insulin resistance. How its deficiency causes diminished GLUT4 proteins remains unknown. We found that the deletion of AS160 decreased GLUT4 levels in a cell/tissue-autonomous manner. Consequently, skeletal muscle-specific deletion of AS160 caused postprandial hyperglycemia and hyperinsulinemia. The pathogenic effects of AS160 deletion are mainly, if not exclusively, due to the loss of its RabGAP function since the RabGAP-inactive AS160R917K mutant mice phenocopied the AS160 knockout mice. The inactivation of RabGAP of AS160 promotes lysosomal degradation of GLUT4, and the inhibition of lysosome function could restore GLUT4 protein levels. Collectively, these findings demonstrate that the RabGAP activity of AS160 maintains GLUT4 protein levels in a cell/tissue-autonomous manner and its inactivation causes lysosomal degradation of GLUT4 and postprandial hyperglycemia and hyperinsulinemia.
Collapse
Affiliation(s)
- Bingxian Xie
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Qiaoli Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Liang Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Yang Sheng
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
| | - Hong Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
- Collaborative Innovation Center of Genetics and Development, Shanghai, China
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing, China
- Collaborative Innovation Center of Genetics and Development, Shanghai, China
| |
Collapse
|
76
|
Møller CL, Kjøbsted R, Enriori PJ, Jensen TE, Garcia-Rudaz C, Litwak SA, Raun K, Wojtaszewski J, Wulff BS, Cowley MA. α-MSH Stimulates Glucose Uptake in Mouse Muscle and Phosphorylates Rab-GTPase-Activating Protein TBC1D1 Independently of AMPK. PLoS One 2016; 11:e0157027. [PMID: 27467141 PMCID: PMC4965092 DOI: 10.1371/journal.pone.0157027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/24/2016] [Indexed: 12/21/2022] Open
Abstract
The melanocortin system includes five G-protein coupled receptors (family A) defined as MC1R-MC5R, which are stimulated by endogenous agonists derived from proopiomelanocortin (POMC). The melanocortin system has been intensely studied for its central actions in body weight and energy expenditure regulation, which are mainly mediated by MC4R. The pituitary gland is the source of various POMC-derived hormones released to the circulation, which raises the possibility that there may be actions of the melanocortins on peripheral energy homeostasis. In this study, we examined the molecular signaling pathway involved in α-MSH-stimulated glucose uptake in differentiated L6 myotubes and mouse muscle explants. In order to examine the involvement of AMPK, we investigate α-MSH stimulation in both wild type and AMPK deficient mice. We found that α-MSH significantly induces phosphorylation of TBC1 domain (TBC1D) family member 1 (S237 and T596), which is independent of upstream PKA and AMPK. We find no evidence to support that α-MSH-stimulated glucose uptake involves TBC1D4 phosphorylation (T642 and S704) or GLUT4 translocation.
Collapse
Affiliation(s)
| | - Rasmus Kjøbsted
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Pablo J. Enriori
- Monash Obesity & Diabetes Institute, Metabolic Neurophysiology Laboratory, Monash University, 3168 Clayton, Australia
| | - Thomas Elbenhardt Jensen
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Cecilia Garcia-Rudaz
- Department of Pediatrics, Centenary Hospital for Women, Youth and Children and Australian National University, 2605 Canberra, Australia
| | - Sara A. Litwak
- Monash Obesity & Diabetes Institute, Metabolic Neurophysiology Laboratory, Monash University, 3168 Clayton, Australia
| | - Kirsten Raun
- Incretin and Obesity Biology, Novo Nordisk A/S, 2760 Maaloev, Denmark
| | - Jørgen Wojtaszewski
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Michael A. Cowley
- Monash Obesity & Diabetes Institute, Metabolic Neurophysiology Laboratory, Monash University, 3168 Clayton, Australia
| |
Collapse
|
77
|
Lim GE, Piske M, Lulo JE, Ramshaw HS, Lopez AF, Johnson JD. Ywhaz/14-3-3ζ Deletion Improves Glucose Tolerance Through a GLP-1-Dependent Mechanism. Endocrinology 2016; 157:2649-59. [PMID: 27167773 DOI: 10.1210/en.2016-1016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple signaling pathways mediate the actions of metabolic hormones to control glucose homeostasis, but the proteins that coordinate such networks are poorly understood. We previously identified the molecular scaffold protein, 14-3-3ζ, as a critical regulator of in vitro β-cell survival and adipogenesis, but its metabolic roles in glucose homeostasis have not been studied in depth. Herein, we report that Ywhaz gene knockout mice (14-3-3ζKO) exhibited elevated fasting insulin levels while maintaining normal β-cell responsiveness to glucose when compared with wild-type littermate controls. In contrast with our observations after an ip glucose bolus, glucose tolerance was significantly improved in 14-3-3ζKO mice after an oral glucose gavage. This improvement in glucose tolerance was associated with significantly elevated fasting glucagon-like peptide-1 (GLP-1) levels. 14-3-3ζ knockdown in GLUTag L cells elevated GLP-1 synthesis and increased GLP-1 release. Systemic inhibition of the GLP-1 receptor attenuated the improvement in oral glucose tolerance that was seen in 14-3-3ζKO mice. When taken together these findings demonstrate novel roles of 14-3-3ζ in the regulation of glucose homeostasis and suggest that modulating 14-3-3ζ levels in intestinal L cells may have beneficial metabolic effects through GLP-1-dependent mechanisms.
Collapse
Affiliation(s)
- Gareth E Lim
- Department of Cellular and Physiological Sciences (G.E.L., M.P., J.D.J.), University of British Columbia, Vancouver, BC, Canada; ALPCO (J.E.L.), Salem, New Hampshire; and The Centre for Cancer Biology (H.S.R., A.F.L.), South Australia Pathology and University of South Australia, Adelaide, Australia
| | - Micah Piske
- Department of Cellular and Physiological Sciences (G.E.L., M.P., J.D.J.), University of British Columbia, Vancouver, BC, Canada; ALPCO (J.E.L.), Salem, New Hampshire; and The Centre for Cancer Biology (H.S.R., A.F.L.), South Australia Pathology and University of South Australia, Adelaide, Australia
| | - James E Lulo
- Department of Cellular and Physiological Sciences (G.E.L., M.P., J.D.J.), University of British Columbia, Vancouver, BC, Canada; ALPCO (J.E.L.), Salem, New Hampshire; and The Centre for Cancer Biology (H.S.R., A.F.L.), South Australia Pathology and University of South Australia, Adelaide, Australia
| | - Hayley S Ramshaw
- Department of Cellular and Physiological Sciences (G.E.L., M.P., J.D.J.), University of British Columbia, Vancouver, BC, Canada; ALPCO (J.E.L.), Salem, New Hampshire; and The Centre for Cancer Biology (H.S.R., A.F.L.), South Australia Pathology and University of South Australia, Adelaide, Australia
| | - Angel F Lopez
- Department of Cellular and Physiological Sciences (G.E.L., M.P., J.D.J.), University of British Columbia, Vancouver, BC, Canada; ALPCO (J.E.L.), Salem, New Hampshire; and The Centre for Cancer Biology (H.S.R., A.F.L.), South Australia Pathology and University of South Australia, Adelaide, Australia
| | - James D Johnson
- Department of Cellular and Physiological Sciences (G.E.L., M.P., J.D.J.), University of British Columbia, Vancouver, BC, Canada; ALPCO (J.E.L.), Salem, New Hampshire; and The Centre for Cancer Biology (H.S.R., A.F.L.), South Australia Pathology and University of South Australia, Adelaide, Australia
| |
Collapse
|
78
|
Bruno J, Brumfield A, Chaudhary N, Iaea D, McGraw TE. SEC16A is a RAB10 effector required for insulin-stimulated GLUT4 trafficking in adipocytes. J Cell Biol 2016; 214:61-76. [PMID: 27354378 PMCID: PMC4932369 DOI: 10.1083/jcb.201509052] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 06/08/2016] [Indexed: 12/19/2022] Open
Abstract
Sec16A is known to be required for COPII vesicle formation from the ER. Here, Bruno et al. show that, independent of its role at the ER, Sec16A is a RAB10 effector involved in the insulin-stimulated formation of specialized transport vesicles that ferry the GLUT4 glucose transporter to the plasma membrane of adipocytes. RAB10 is a regulator of insulin-stimulated translocation of the GLUT4 glucose transporter to the plasma membrane (PM) of adipocytes, which is essential for whole-body glucose homeostasis. We establish SEC16A as a novel RAB10 effector in this process. Colocalization of SEC16A with RAB10 is augmented by insulin stimulation, and SEC16A knockdown attenuates insulin-induced GLUT4 translocation, phenocopying RAB10 knockdown. We show that SEC16A and RAB10 promote insulin-stimulated mobilization of GLUT4 from a perinuclear recycling endosome/TGN compartment. We propose RAB10–SEC16A functions to accelerate formation of the vesicles that ferry GLUT4 to the PM during insulin stimulation. Because GLUT4 continually cycles between the PM and intracellular compartments, the maintenance of elevated cell-surface GLUT4 in the presence of insulin requires accelerated biogenesis of the specialized GLUT4 transport vesicles. The function of SEC16A in GLUT4 trafficking is independent of its previously characterized activity in ER exit site formation and therefore independent of canonical COPII-coated vesicle function. However, our data support a role for SEC23A, but not the other COPII components SEC13, SEC23B, and SEC31, in the insulin stimulation of GLUT4 trafficking, suggesting that vesicles derived from subcomplexes of COPII coat proteins have a role in the specialized trafficking of GLUT4.
Collapse
Affiliation(s)
- Joanne Bruno
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065 Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065
| | | | - Natasha Chaudhary
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - David Iaea
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065 Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
79
|
Gongpan P, Lu Y, Wang F, Xu Y, Xiong W. AS160 controls eukaryotic cell cycle and proliferation by regulating the CDK inhibitor p21. Cell Cycle 2016; 15:1733-41. [PMID: 27152871 DOI: 10.1080/15384101.2016.1183853] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AS160 (TBC1D4) has been implicated in multiple biological processes. However, the role and the mechanism of action of AS160 in the regulation of cell proliferation remain unclear. In this study, we demonstrated that AS160 knockdown led to blunted cell proliferation in multiple cell types, including fibroblasts and cancer cells. The results of cell cycle analysis showed that these cells were arrested in the G1 phase. Intriguingly, this inhibition of cell proliferation and the cell cycle arrest caused by AS160 depletion were glucose independent. Moreover, AS160 silencing led to a marked upregulation of the expression of the cyclin-dependent kinase inhibitor p21. Furthermore, whereas AS160 overexpression resulted in p21 downregulation and rescued the arrested cell cycle in AS160-depeleted cells, p21 silencing rescued the inhibited cell cycle and proliferation in the cells. Thus, our results demonstrated that AS160 regulates glucose-independent eukaryotic cell proliferation through p21-dependent control of the cell cycle, and thereby revealed a molecular mechanism of AS160 modulation of cell cycle and proliferation that is of general physiological significance.
Collapse
Affiliation(s)
- Pianchou Gongpan
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Yanting Lu
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Fang Wang
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Yuhui Xu
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China.,b Graduate University of Chinese Academy of Sciences , Beijing , P.R. China
| | - Wenyong Xiong
- a State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany , Chinese Academy of Sciences , Kunming , Yunnan , P.R. China
| |
Collapse
|
80
|
Lim GE, Johnson JD. 14-3-3ζ: A numbers game in adipocyte function? Adipocyte 2016; 5:232-7. [PMID: 27386155 PMCID: PMC4916895 DOI: 10.1080/21623945.2015.1120913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/05/2015] [Accepted: 11/10/2015] [Indexed: 12/22/2022] Open
Abstract
Molecular scaffolds are often viewed as passive signaling molecules that facilitate protein-protein interactions. However, new evidence gained from the use of loss-of-function or gain-of-function models is dispelling this notion. Our own recent discovery of 14-3-3ζ as an essential regulator of adipogenesis highlights the complex roles of this member of the 14-3-3 protein family. Depletion of the 14-3-3ζ isoform affected parallel pathways that drive adipocyte development, including pathways controlling the stability of key adipogenic transcription factors and cell cycle progression. Going beyond adipocyte differentiation, this study opens new avenues of research in the context of metabolism, as 14-3-3ζ binds to a variety of well-established metabolic proteins that harbor its canonical phosphorylation binding motifs. This suggests that 14-3-3ζ may contribute to key metabolic signaling pathways, such as those that facilitate glucose uptake and fatty acid metabolism. Herein, we discuss these novel areas of research, which will undoubtedly shed light onto novel roles of 14-3-3ζ, and perhaps its related family members, on glucose homeostasis.
Collapse
Affiliation(s)
- Gareth E. Lim
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
81
|
Hargett SR, Walker NN, Keller SR. Rab GAPs AS160 and Tbc1d1 play nonredundant roles in the regulation of glucose and energy homeostasis in mice. Am J Physiol Endocrinol Metab 2016; 310:E276-88. [PMID: 26625902 PMCID: PMC4888528 DOI: 10.1152/ajpendo.00342.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/29/2015] [Indexed: 11/22/2022]
Abstract
The related Rab GTPase-activating proteins (Rab GAPs) AS160 and Tbc1d1 regulate the trafficking of the glucose transporter GLUT4 that controls glucose uptake in muscle and fat cells and glucose homeostasis. AS160- and Tbc1d1-deficient mice exhibit different adipocyte- and skeletal muscle-specific defects in glucose uptake, GLUT4 expression and trafficking, and glucose homeostasis. A recent study analyzed male mice with simultaneous deletion of AS160 and Tbc1d1 (AS160(-/-)/Tbc1d1(-/-) mice). Herein, we describe abnormalities in male and female AS160(-/-)/Tbc1d1(-/-) mice on another strain background. We confirm the earlier observation that GLUT4 expression and glucose uptake defects of single-knockout mice join in AS160(-/-)/Tbc1d1(-/-) mice to affect all skeletal muscle and adipose tissues. In large mixed fiber-type skeletal muscles, changes in relative basal GLUT4 plasma membrane association in AS160(-/-) and Tbc1d1(-/-) mice also combine in AS160(-/-)/Tbc1d1(-/-) mice. However, we found different glucose uptake abnormalities in isolated skeletal muscles and adipocytes than reported previously, resulting in different interpretations of how AS160 and Tbc1d1 regulate GLUT4 translocation to the cell surface. In support of a larger role for AS160 in glucose homeostasis, in contrast with the previous study, we find similarly impaired glucose and insulin tolerance in AS160(-/-)/Tbc1d1(-/-) and AS160(-/-) mice. However, in vivo glucose uptake abnormalities in AS160(-/-)/Tbc1d1(-/-) skeletal muscles differ from those observed previously in AS160(-/-) mice, indicating additional defects due to Tbc1d1 deletion. Similar to AS160- and Tbc1d1-deficient mice, AS160(-/-)/Tbc1d1(-/-) mice show sex-specific abnormalities in glucose and energy homeostasis. In conclusion, our study supports nonredundant functions for AS160 and Tbc1d1.
Collapse
Affiliation(s)
- Stefan R Hargett
- Department of Medicine-Division of Endocrinology, University of Virginia, Charlottesville Virginia
| | - Natalie N Walker
- Department of Medicine-Division of Endocrinology, University of Virginia, Charlottesville Virginia
| | - Susanna R Keller
- Department of Medicine-Division of Endocrinology, University of Virginia, Charlottesville Virginia
| |
Collapse
|
82
|
Budi EH, Muthusamy BP, Derynck R. The insulin response integrates increased TGF-β signaling through Akt-induced enhancement of cell surface delivery of TGF-β receptors. Sci Signal 2015; 8:ra96. [PMID: 26420907 DOI: 10.1126/scisignal.aaa9432] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased activity of transforming growth factor-β (TGF-β), which binds to and stimulates cell surface receptors, contributes to cancer progression and fibrosis by driving epithelial cells toward a migratory mesenchymal phenotype and increasing the abundance of extracellular matrix proteins. The abundance of TGF-β receptors at the cell surface determines cellular responsiveness to TGF-β, which is often produced by the same cells that have the receptors, and thus serves as an autocrine signal. We found that Akt-mediated phosphorylation of AS160, a RabGAP [guanosine triphosphatase (GTPase)-activating protein], promoted the translocation of TGF-β receptors from intracellular stores to the plasma membrane of mouse embryonic fibroblasts and NMuMG epithelial cells. Consequently, insulin, which is commonly used to treat hyperglycemia and activates Akt signaling, increased the amount of TGF-β receptors at the cell surface, thereby enhancing TGF-β responsiveness. This insulin-induced increase in autocrine TGF-β signaling contributed to insulin-induced gene expression responses, attenuated the epithelial phenotype, and promoted the migration of NMuMG cells. Furthermore, the enhanced delivery of TGF-β receptors at the cell surface enabled insulin to increase TGF-β-induced gene responses. The enhancement of TGF-β responsiveness in response to Akt activation may help to explain the biological effects of insulin, the progression of cancers in which Akt is activated, and the increased incidence of fibroses in diabetes.
Collapse
Affiliation(s)
- Erine H Budi
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA
| | - Baby-Periyanayaki Muthusamy
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA
| | - Rik Derynck
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA.
| |
Collapse
|
83
|
Shen L, Haas M, Wang DQH, May A, Lo CC, Obici S, Tso P, Woods SC, Liu M. Ginsenoside Rb1 increases insulin sensitivity by activating AMP-activated protein kinase in male rats. Physiol Rep 2015; 3:3/9/e12543. [PMID: 26359241 PMCID: PMC4600387 DOI: 10.14814/phy2.12543] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Although ginseng has been reported to ameliorate hyperglycemia in animal models and clinical studies, the molecular mechanisms are largely unknown. We previously reported that chronic treatment with ginsenoside Rb1 (Rb1), a major component of ginseng, significantly reduced fasting glucose and improved glucose tolerance in high-fat diet (HFD)-induced obese rats. These effects were greater than those observed in pair-fed rats, suggesting a direct effect of Rb1 on glucose homeostasis, and this possibility was confirmed in the present study. In lean rats fed standard rodent chow, 5-day treatment with Rb1 significantly improved glucose tolerance and enhanced insulin sensitivity. Notably, those effects were not accompanied by reduced food intake or changed body weight. To elucidate the underlying molecular mechanisms, rats fed a HFD for 4 weeks were treated with Rb1 for 5 days. Subsequently, euglycemic-hyperinsulinemic clamp studies found that compared to vehicle, Rb1, while not changing food intake or body weight, significantly increased glucose infusion rate required to maintain euglycemia. Consistent with this, insulin-induced inhibition of hepatic gluconeogenesis was significantly enhanced and hepatic phosphoenolpyruvate carboxykinase and glucose-6-phosphatase gene expression was suppressed. Additionally, glucose uptake was significantly increased in skeletal muscle. While proximal insulin signaling was not changed after Rb1 treatment, increased phosphorylation of TBC1D4, a downstream target of AMPK signaling, appears to be a key part of the mechanism for Rb1-stimulated glucose uptake in skeletal muscle. These findings indicate that Rb1 has multiple effects on glucose homeostasis, and provide strong rationale for further evaluation of its potential therapeutic role.
Collapse
Affiliation(s)
- Ling Shen
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael Haas
- Department of Cancer & Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David Q-H Wang
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Aaron May
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Chunmin C Lo
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Silvana Obici
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
84
|
Wang H, Sharma N, Arias EB, Cartee GD. Insulin Signaling and Glucose Uptake in the Soleus Muscle of 30-Month-Old Rats After Calorie Restriction With or Without Acute Exercise. J Gerontol A Biol Sci Med Sci 2015; 71:323-32. [PMID: 26341783 DOI: 10.1093/gerona/glv142] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 07/16/2015] [Indexed: 12/16/2022] Open
Abstract
Exercise and calorie restriction (CR) can each improve insulin sensitivity in older individuals, but benefits of combining these treatments on skeletal muscle insulin signaling and glucose uptake are poorly understood, especially in predominantly slow-twitch muscles (eg, soleus). Accordingly, our purpose was to determine independent and combined effects of prior acute exercise and CR (beginning at 14 weeks old) on insulin signaling and glucose uptake in insulin-stimulated soleus muscles of 30-month-old rats. CR alone (but not exercise alone) versus ad libitum sedentary controls induced greater insulin-stimulated glucose uptake. There was a main effect of diet (CR > ad libitum) for insulin-stimulated Akt(Ser473) and Akt(Thr308) phosphorylation. CR alone versus ad libitum sedentary increased Akt substrate of 160 kDa (AS160) Ser(588) phosphorylation and TBC1D1 Thr(596), but not AS160 Thr(642) phosphorylation or abundance of GLUT4, GLUT1, or hexokinase II proteins. Combined CR and exercise versus CR alone did not further increase insulin-stimulated glucose uptake although phosphorylation of Akt(Ser473), Akt(Thr308), TBC1D1(Thr596), and AMPK(Thr172) for the combined group exceeded values for CR and/or exercise alone. These results revealed that although the soleus was highly responsive to a CR-induced enhancement of insulin-stimulated glucose uptake, the exercise protocol did not elevate insulin-stimulated glucose uptake, either alone or when combined with CR.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor. College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Naveen Sharma
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor. School of Health Sciences, Central Michigan University, Mount Pleasant
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor. Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor. Institute of Gerontology, University of Michigan, Ann Arbor.
| |
Collapse
|
85
|
Luiken JJFP, Glatz JFC, Neumann D. Cardiac contraction-induced GLUT4 translocation requires dual signaling input. Trends Endocrinol Metab 2015; 26:404-10. [PMID: 26138758 DOI: 10.1016/j.tem.2015.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/31/2015] [Accepted: 06/01/2015] [Indexed: 10/23/2022]
Abstract
Contraction-induced translocation of glucose transporter type-4 (GLUT4) to the sarcolemma is essential to stimulate cardiac glucose uptake during increased energy demand. As such, this process is a target for therapeutic strategies aiming at increasing glucose uptake in insulin-resistant and/or diabetic hearts. AMP-activated protein kinase (AMPK) and its upstream kinases form part of a signaling axis essential for contraction-induced GLUT4 translocation. Recently, activation of protein kinase-D1 (PKD1) was also shown to be as obligatory for contraction-induced GLUT4 translocation in cardiac muscle. However, contraction-induced PKD1 activation in this context occurs independently from AMPK signaling, suggesting that contraction-induced GLUT4 translocation requires the input of two separate signaling pathways. Necessity for dual input would more tightly couple GLUT4 translocation to stimuli that are inherent to cardiac contraction.
Collapse
Affiliation(s)
- Joost J F P Luiken
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, NL-6200 Maastricht MD, the Netherlands.
| | - Jan F C Glatz
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, NL-6200 Maastricht MD, the Netherlands
| | - Dietbert Neumann
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, NL-6200 Maastricht MD, the Netherlands
| |
Collapse
|
86
|
Tao W, Wu J, Xie BX, Zhao YY, Shen N, Jiang S, Wang XX, Xu N, Jiang C, Chen S, Gao X, Xue B, Li CJ. Lipid-induced Muscle Insulin Resistance Is Mediated by GGPPS via Modulation of the RhoA/Rho Kinase Signaling Pathway. J Biol Chem 2015; 290:20086-97. [PMID: 26112408 DOI: 10.1074/jbc.m115.657742] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Indexed: 12/14/2022] Open
Abstract
Elevated circulating free fatty acid levels are important contributors to insulin resistance in the muscle and liver, but the underlying mechanisms require further elucidation. Here, we show that geranylgeranyl diphosphate synthase 1 (GGPPS), which is a branch point enzyme in the mevalonic acid pathway, promotes lipid-induced muscle insulin resistance through activation of the RhoA/Rho kinase signaling pathway. We have found that metabolic perturbation would increase GGPPS expression in the skeletal muscles of db/db mice and high fat diet-fed mice. To address the metabolic effects of GGPPS activity in skeletal muscle, we generated mice with specific GGPPS deletions in their skeletal muscle tissue. Heterozygous knock-out of GGPPS in the skeletal muscle improved systemic insulin sensitivity and glucose homeostasis in mice fed both normal chow and high fat diets. These metabolic alterations were accompanied by activated PI3K/Akt signaling and enhanced glucose uptake in the skeletal muscle. Further investigation showed that the free fatty acid-stimulated GGPPS expression in the skeletal muscle was able to enhance the geranylgeranylation of RhoA, which further induced the inhibitory phosphorylation of IRS-1 (Ser-307) by increasing Rho kinase activity. These results implicate a crucial role of the GGPPS/RhoA/Rho kinase/IRS-1 pathway in skeletal muscle, in which it mediates lipid-induced systemic insulin resistance in obese mice. Therefore, skeletal muscle GGPPS may represent a potential pharmacological target for the prevention and treatment of obesity-related type 2 diabetes.
Collapse
Affiliation(s)
- Weiwei Tao
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Jing Wu
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Bing-Xian Xie
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Yuan-Yuan Zhao
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Ning Shen
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Shan Jiang
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Xiu-Xing Wang
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Na Xu
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Chen Jiang
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Shuai Chen
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Xiang Gao
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Bin Xue
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| | - Chao-Jun Li
- From the Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and the School of Medicine, Nanjing University, Nanjing 210061, China
| |
Collapse
|
87
|
Albers PH, Bojsen-Møller KN, Dirksen C, Serup AK, Kristensen DE, Frystyk J, Clausen TR, Kiens B, Richter EA, Madsbad S, Wojtaszewski JFP. Enhanced insulin signaling in human skeletal muscle and adipose tissue following gastric bypass surgery. Am J Physiol Regul Integr Comp Physiol 2015; 309:R510-24. [PMID: 26062634 DOI: 10.1152/ajpregu.00228.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/01/2015] [Indexed: 12/12/2022]
Abstract
Roux-en-Y gastric bypass (RYGB) leads to increased peripheral insulin sensitivity. The aim of this study was to investigate the effect of RYGB on expression and regulation of proteins involved in regulation of peripheral glucose metabolism. Skeletal muscle and adipose tissue biopsies from glucose-tolerant and type 2 diabetic subjects at fasting and during a hyperinsulinemic-euglycemic clamp before as well as 1 wk and 3 and 12 mo after RYGB were analyzed for relevant insulin effector proteins/signaling components. Improvement in peripheral insulin sensitivity mainly occurred at 12 mo postsurgery when major weight loss was evident and occurred concomitantly with alterations in plasma adiponectin and in protein expression/signaling in peripheral tissues. In skeletal muscle, protein expression of GLUT4, phosphorylated levels of TBC1D4, as well as insulin-induced changes in phosphorylation of Akt and glycogen synthase activity were enhanced 12 mo postsurgery. In adipose tissue, protein expression of GLUT4, Akt2, TBC1D4, and acetyl-CoA carboxylase (ACC), phosphorylated levels of AMP-activated protein kinase and ACC, as well as insulin-induced changes in phosphorylation of Akt and TBC1D4, were enhanced 12 mo postsurgery. Adipose tissue from glucose-tolerant subjects was the most responsive to RYGB compared with type 2 diabetic patients, whereas changes in skeletal muscle were largely similar in these two groups. In conclusion, an improved molecular insulin-sensitive phenotype of skeletal muscle and adipose tissue appears to contribute to the improved whole body insulin action following a substantial weight loss after RYGB.
Collapse
Affiliation(s)
- Peter H Albers
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark; Diabetes Research Unit, Novo Nordisk A/S, Maaloev, Denmark
| | - Kirstine N Bojsen-Møller
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark; Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; and
| | - Carsten Dirksen
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark; Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; and
| | - Annette K Serup
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Dorte E Kristensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | | | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark;
| |
Collapse
|
88
|
Affiliation(s)
- Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology; Department of Molecular & Integrative Physiology; and Institute of Gerontology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
89
|
Kjøbsted R, Treebak JT, Fentz J, Lantier L, Viollet B, Birk JB, Schjerling P, Björnholm M, Zierath JR, Wojtaszewski JFP. Prior AICAR stimulation increases insulin sensitivity in mouse skeletal muscle in an AMPK-dependent manner. Diabetes 2015; 64:2042-55. [PMID: 25552597 DOI: 10.2337/db14-1402] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/20/2014] [Indexed: 11/13/2022]
Abstract
An acute bout of exercise increases glucose uptake in skeletal muscle by an insulin-independent mechanism. In the period after exercise, insulin sensitivity to increased glucose uptake is enhanced. The molecular mechanisms underpinning this phenomenon are poorly understood but appear to involve an increased cell surface abundance of GLUT4. While increased proximal insulin signaling does not seem to mediate this effect, elevated phosphorylation of TBC1D4, a downstream target of both insulin (Akt) and exercise (AMPK) signaling, appears to play a role. The main purpose of this study was to determine whether AMPK activation increases skeletal muscle insulin sensitivity. We found that prior AICAR stimulation of wild-type mouse muscle increases insulin sensitivity to stimulate glucose uptake. However, this was not observed in mice with reduced or ablated AMPK activity in skeletal muscle. Furthermore, prior AICAR stimulation enhanced insulin-stimulated phosphorylation of TBC1D4 at Thr(649) and Ser(711) in wild-type muscle only. These phosphorylation events were positively correlated with glucose uptake. Our results provide evidence to support that AMPK activation is sufficient to increase skeletal muscle insulin sensitivity. Moreover, TBC1D4 phosphorylation may facilitate the effect of prior AMPK activation to enhance glucose uptake in response to insulin.
Collapse
Affiliation(s)
- Rasmus Kjøbsted
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Joachim Fentz
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Louise Lantier
- INSERM, U1016, Institut Cochin, Paris, France CNRS, UMR8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France CNRS, UMR8104, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jesper B Birk
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Peter Schjerling
- Institute of Sports Medicine, Department of Orthopedic Surgery, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Björnholm
- Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
90
|
Quan C, Xie B, Wang HY, Chen S. PKB-Mediated Thr649 Phosphorylation of AS160/TBC1D4 Regulates the R-Wave Amplitude in the Heart. PLoS One 2015; 10:e0124491. [PMID: 25923736 PMCID: PMC4414484 DOI: 10.1371/journal.pone.0124491] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/04/2015] [Indexed: 11/18/2022] Open
Abstract
The Rab GTPase activating protein (RabGAP), AS160/TBC1D4, is an important substrate of protein kinase B (PKB), and regulates insulin-stimulated trafficking of glucose transporter 4. Besides, AS160/TBC1D4 has also been shown to regulate trafficking of many other membrane proteins including FA translocase/CD36 in cardiomyocytes. However, it is not clear whether it plays any role in regulating heart functions in vivo. Here, we found that PKB-mediated phosphorylation of Thr649 on AS160/TBC1D4 represented one of the major PAS-binding signals in the heart in response to insulin. Mutation of Thr649 to a non-phosphorylatable alanine increased the R-wave amplitude in the AS160Thr649Ala knockin mice. However, this knockin mutation did not affect the heart functions under both normal and infarct conditions. Interestingly, myocardial infarction induced the expression of a related RabGAP, TBC1D1, in the infarct zone as well as in the border zone. Together, these data show that the Thr649 phosphorylation of AS160/TBC1D4 plays an important role in the heart’s electrical conduction system through regulating the R-wave amplitude.
Collapse
Affiliation(s)
- Chao Quan
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Bingxian Xie
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
| | - Hong Yu Wang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
- * E-mail: (HYW); ( (SC)
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210061, China
- * E-mail: (HYW); ( (SC)
| |
Collapse
|
91
|
Chadt A, Immisch A, de Wendt C, Springer C, Zhou Z, Stermann T, Holman GD, Loffing-Cueni D, Loffing J, Joost HG, Al-Hasani H. “Deletion of both Rab-GTPase–activating proteins TBC1D1 and TBC1D4 in mice eliminates insulin- and AICAR-stimulated glucose transport [corrected]. Diabetes 2015; 64:746-59. [PMID: 25249576 DOI: 10.2337/db14-0368] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Rab-GTPase–activating proteins TBC1D1 and TBC1D4 (AS160) were previously shown to regulate GLUT4 translocation in response to activation of AKT and AMP-dependent kinase [corrected]. However, knockout mice lacking either Tbc1d1 or Tbc1d4 displayed only partially impaired insulin-stimulated glucose uptake in fat and muscle tissue. The aim of this study was to determine the impact of the combined inactivation of Tbc1d1 and Tbc1d4 on glucose metabolism in double-deficient (D1/4KO) mice. D1/4KO mice displayed normal fasting glucose concentrations but had reduced tolerance to intraperitoneally administered glucose, insulin, and AICAR. D1/4KO mice showed reduced respiratory quotient, indicating increased use of lipids as fuel. These mice also consistently showed elevated fatty acid oxidation in isolated skeletal muscle, whereas insulin-stimulated glucose uptake in muscle and adipose cells was almost completely abolished. In skeletal muscle and white adipose tissue, the abundance of GLUT4 protein, but not GLUT4 mRNA, was substantially reduced. Cell surface labeling of GLUTs indicated that RabGAP deficiency impairs retention of GLUT4 in intracellular vesicles in the basal state. Our results show that TBC1D1 and TBC1D4 together play essential roles in insulin-stimulated glucose uptake and substrate preference in skeletal muscle and adipose cells.
Collapse
Affiliation(s)
- Alexandra Chadt
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, Düsseldorf, Germany German Center for Diabetes Research (DZD), Düsseldorf, Germany
| | - Anja Immisch
- German Institute for Human Nutrition, Potsdam, Germany
| | - Christian de Wendt
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christian Springer
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, Düsseldorf, Germany
| | - Zhou Zhou
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, Düsseldorf, Germany
| | - Torben Stermann
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, Düsseldorf, Germany
| | - Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath, Bath, U.K
| | | | | | - Hans-Georg Joost
- German Center for Diabetes Research (DZD), Düsseldorf, Germany German Institute for Human Nutrition, Potsdam, Germany
| | - Hadi Al-Hasani
- German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, Düsseldorf, Germany German Center for Diabetes Research (DZD), Düsseldorf, Germany
| |
Collapse
|
92
|
Cartee GD. Roles of TBC1D1 and TBC1D4 in insulin- and exercise-stimulated glucose transport of skeletal muscle. Diabetologia 2015; 58:19-30. [PMID: 25280670 PMCID: PMC4258142 DOI: 10.1007/s00125-014-3395-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/07/2014] [Indexed: 10/24/2022]
Abstract
This review focuses on two paralogue Rab GTPase activating proteins known as TBC1D1 Tre-2/BUB2/cdc 1 domain family (TBC1D) 1 and TBC1D4 (also called Akt Substrate of 160 kDa, AS160) and their roles in controlling skeletal muscle glucose transport in response to the independent and combined effects of insulin and exercise. Convincing evidence implicates Akt2-dependent TBC1D4 phosphorylation on T642 as a key part of the mechanism for insulin-stimulated glucose uptake by skeletal muscle. TBC1D1 phosphorylation on several insulin-responsive sites (including T596, a site corresponding to T642 in TBC1D4) does not appear to be essential for in vivo insulin-stimulated glucose uptake by skeletal muscle. In vivo exercise or ex vivo contraction of muscle result in greater TBC1D1 phosphorylation on S237 that is likely to be secondary to increased AMP-activated protein kinase activity and potentially important for contraction-stimulated glucose uptake. Several studies that evaluated both normal and insulin-resistant skeletal muscle stimulated with a physiological insulin concentration after a single exercise session found that greater post-exercise insulin-stimulated glucose uptake was accompanied by greater TBC1D4 phosphorylation on several sites. In contrast, enhanced post-exercise insulin sensitivity was not accompanied by greater insulin-stimulated TBC1D1 phosphorylation. The mechanism for greater TBC1D4 phosphorylation in insulin-stimulated muscles after acute exercise is uncertain, and a causal link between enhanced TBC1D4 phosphorylation and increased post-exercise insulin sensitivity has yet to be established. In summary, TBC1D1 and TBC1D4 have important, but distinct roles in regulating muscle glucose transport in response to insulin and exercise.
Collapse
Affiliation(s)
- Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, 401 Washtenaw Avenue, Ann Arbor, MI, 48109-2214, USA,
| |
Collapse
|
93
|
Iwabe M, Kawamoto E, Koshinaka K, Kawanaka K. Increased postexercise insulin sensitivity is accompanied by increased AS160 phosphorylation in slow-twitch soleus muscle. Physiol Rep 2014; 2:2/12/e12162. [PMID: 25501433 PMCID: PMC4332192 DOI: 10.14814/phy2.12162] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A single bout of exercise can enhance insulin‐stimulated glucose uptake in both
fast‐twitch (type II) and slow‐twitch (type I) skeletal muscle for several hours
postexercise. Akt substrate of 160 kDa (AS160) is most distal insulin signaling proteins that have
been proposed to contribute to the postexercise enhancement of insulin action in fast‐twitch
muscle. In this study, we examined whether the postexercise increase in insulin action of glucose
uptake in slow‐twitch muscle is accompanied by increased phosphorylation of AS160 and its
paralog TBC1D1. Male Wistar rats (~1‐month‐old) were exercised on a treadmill for 180
min (9 m/min). Insulin (50 μU/mL)‐stimulated glucose uptake was
increased at 2 h after cessation of exercise in soleus muscle composed of predominantly
slow‐twitch fibers. This postexercise increase in insulin action of glucose uptake was
accompanied by increased phosphorylation of AS160 (detected by phospho‐Thr642 and
phospho‐Ser588 antibody). On the other hand, prior exercise did not increase phosphorylation
of TBC1D1 (detected by phospho‐Thr590) at 2 h postexercise. These results suggest the
possibility that an enhancement in AS160 phosphorylation but not TBC1D1 phosphorylation is involved
with increased postexercise insulin action of glucose uptake in slow‐twitch muscle. In slow‐twitch soleus muscle, phosphorylation of AS160 Thr642 and Ser588 was increased
together with the enhanced insulin action of the glucose uptake at 2 h postexercise. The phosphosite
of TBC1D1 (Thr590), which is possibly important for insulin‐stimulated glucose uptake, did
not increase phosphorylation at 2 h postexercise. These results suggest that the increased
phosphorylation of AS160, but not TBC1D1, can account for the postexercise enhancement in the
insulin action of the glucose uptake in slow‐twitch muscle.
Collapse
Affiliation(s)
- Maiko Iwabe
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan
| | - Emi Kawamoto
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan
| | - Keiichi Koshinaka
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan
| | - Kentaro Kawanaka
- Department of Health and Nutrition, Niigata University of Health and Welfare, Niigata, Japan
| |
Collapse
|
94
|
Grahame Hardie D. AMP-activated protein kinase: a key regulator of energy balance with many roles in human disease. J Intern Med 2014; 276:543-59. [PMID: 24824502 PMCID: PMC5705060 DOI: 10.1111/joim.12268] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that regulates cellular and whole-body energy balance. A recently reported crystal structure has illuminated the complex regulatory mechanisms by which AMP and ADP cause activation of AMPK, involving phosphorylation by the upstream kinase LKB1. Once activated by falling cellular energy status, AMPK activates catabolic pathways that generate ATP whilst inhibiting anabolic pathways and other cellular processes that consume ATP. A role of AMPK is implicated in many human diseases. Mutations in the γ2 subunit cause heart disease due to excessive glycogen storage in cardiac myocytes, leading to ventricular pre-excitation. AMPK-activating drugs reverse many of the metabolic defects associated with insulin resistance, and recent findings suggest that the insulin-sensitizing effects of the widely used antidiabetic drug metformin are mediated by AMPK. The upstream kinase LKB1 is a tumour suppressor, and AMPK may exert many of its antitumour effects. AMPK activation promotes the oxidative metabolism typical of quiescent cells, rather than the aerobic glycolysis observed in tumour cells and cells involved in inflammation, explaining in part why AMPK activators have both antitumour and anti-inflammatory effects. Salicylate (the major in vivo metabolite of aspirin) activates AMPK, and this could be responsible for at least some of the anticancer and anti-inflammatory effects of aspirin. In addition to metformin and salicylates, novel drugs that modulate AMPK are likely to enter clinical trials soon. Finally, AMPK may be involved in viral infection: downregulation of AMPK during hepatitis C virus infection appears to be essential for efficient viral replication.
Collapse
Affiliation(s)
- D Grahame Hardie
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, Scotland, UK
| |
Collapse
|
95
|
Cheng KKY, Zhu W, Chen B, Wang Y, Wu D, Sweeney G, Wang B, Lam KSL, Xu A. The adaptor protein APPL2 inhibits insulin-stimulated glucose uptake by interacting with TBC1D1 in skeletal muscle. Diabetes 2014; 63:3748-58. [PMID: 24879834 DOI: 10.2337/db14-0337] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Insulin stimulates glucose uptake by promoting the trafficking of GLUT4 to the plasma membrane in muscle cells, and impairment of this insulin action contributes to hyperglycemia in type 2 diabetes. The adaptor protein APPL1 potentiates insulin-stimulated Akt activation and downstream actions. However, the physiological functions of APPL2, a close homolog of APPL1, in regulating glucose metabolism remain elusive. We show that insulin-evoked plasma membrane recruitment of GLUT4 and glucose uptake are impaired by APPL2 overexpression but enhanced by APPL2 knockdown. Likewise, conditional deletion of APPL2 in skeletal muscles enhances insulin sensitivity, leading to an improvement in glucose tolerance. We identified the Rab-GTPase-activating protein TBC1D1 as an interacting partner of APPL2. Insulin stimulates TBC1D1 phosphorylation on serine 235, leading to enhanced interaction with the BAR domain of APPL2, which in turn suppresses insulin-evoked TBC1D1 phosphorylation on threonine 596 in cultured myotubes and skeletal muscle. Substitution of serine 235 with alanine diminishes APPL2-mediated inhibition on insulin-dependent TBC1D1 phosphorylation on threonine 596 and the suppressive effects of TBC1D1 on insulin-induced glucose uptake and GLUT4 translocation to the plasma membrane in cultured myotubes. Therefore, the APPL2-TBC1D1 interaction is a key step to fine tune insulin-stimulated glucose uptake by regulating the membrane recruitment of GLUT4 in skeletal muscle.
Collapse
Affiliation(s)
- Kenneth K Y Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Department of Medicine, The University of Hong Kong, Hong Kong
| | - Weidong Zhu
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Bin Chen
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong
| | - Donghai Wu
- The Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Baile Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Department of Medicine, The University of Hong Kong, Hong Kong
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Department of Medicine, The University of Hong Kong, Hong Kong
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Department of Medicine, The University of Hong Kong, Hong Kong Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong
| |
Collapse
|
96
|
Saravanan M, Pandikumar P, Saravanan S, Toppo E, Pazhanivel N, Ignacimuthu S. Lipolytic and antiadipogenic effects of (3,3-dimethylallyl) halfordinol on 3T3-L1 adipocytes and high fat and fructose diet induced obese C57/BL6J mice. Eur J Pharmacol 2014; 740:714-21. [DOI: 10.1016/j.ejphar.2014.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 02/04/2023]
|
97
|
Chen Q, Quan C, Xie B, Chen L, Zhou S, Toth R, Campbell DG, Lu S, Shirakawa R, Horiuchi H, Li C, Yang Z, MacKintosh C, Wang HY, Chen S. GARNL1, a major RalGAP α subunit in skeletal muscle, regulates insulin-stimulated RalA activation and GLUT4 trafficking via interaction with 14-3-3 proteins. Cell Signal 2014; 26:1636-48. [DOI: 10.1016/j.cellsig.2014.04.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 04/11/2014] [Indexed: 11/16/2022]
|
98
|
Castorena CM, Arias EB, Sharma N, Cartee GD. Postexercise improvement in insulin-stimulated glucose uptake occurs concomitant with greater AS160 phosphorylation in muscle from normal and insulin-resistant rats. Diabetes 2014; 63:2297-308. [PMID: 24608437 PMCID: PMC4066340 DOI: 10.2337/db13-1686] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Earlier research on rats with normal insulin sensitivity demonstrated that acute exercise increased insulin-stimulated glucose uptake (GU) concomitant with greater phosphorylation of Akt substrate of 160 kDa (pAS160). Because mechanisms for exercise effects on GU in insulin-resistant muscle are unknown, our primary objective was to assess insulin-stimulated GU, proximal insulin signaling (insulin receptor [IR] tyrosine phosphorylation, IR substrate 1-phosphatidylinositol-3-kinase, and Akt phosphorylation and activity), and pAS160 in muscles from acutely exercised (one session) and sedentary rats fed either chow (low-fat diet [LFD]; normal insulin sensitivity) or a high-fat diet (HFD; for 2 weeks, insulin-resistant). At 3 h postexercise (3hPEX), isolated epitrochlearis muscles were used for insulin-stimulated GU and insulin signaling measurements. Although exercise did not enhance proximal signaling in either group, insulin-stimulated GU at 3hPEX exceeded respective sedentary control subjects (Sedentary) in both diet groups. Furthermore, insulin-stimulated GU for LFD-3hPEX was greater than HFD-3hPEX values. For HFD-3hPEX muscles, pAS160 exceeded HFD-Sedentary, but in muscle from LFD-3hPEX rats, pAS160 was greater still than HFD-3hPEX values. These results implicated pAS160 as a potential determinant of the exercise-induced elevation in insulin-stimulated GU for each diet group and also revealed pAS160 as a possible mediator of greater postexercise GU of insulin-stimulated muscles from the insulin-sensitive versus insulin-resistant group.
Collapse
Affiliation(s)
- Carlos M Castorena
- 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
| | - Naveen Sharma
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MISchool of Health Sciences, Central Michigan University, Mount Pleasant, MI
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MIDepartment of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MIInstitute of Gerontology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
99
|
Mackenzie RWA, Elliott BT. Akt/PKB activation and insulin signaling: a novel insulin signaling pathway in the treatment of type 2 diabetes. Diabetes Metab Syndr Obes 2014; 7:55-64. [PMID: 24611020 PMCID: PMC3928478 DOI: 10.2147/dmso.s48260] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes is a metabolic disease categorized primarily by reduced insulin sensitivity, β-cell dysfunction, and elevated hepatic glucose production. Treatments reducing hyperglycemia and the secondary complications that result from these dysfunctions are being sought after. Two distinct pathways encourage glucose transport activity in skeletal muscle, ie, the contraction-stimulated pathway reliant on Ca(2+)/5'-monophosphate-activated protein kinase (AMPK)-dependent mechanisms and an insulin-dependent pathway activated via upregulation of serine/threonine protein kinase Akt/PKB. Metformin is an established treatment for type 2 diabetes due to its ability to increase peripheral glucose uptake while reducing hepatic glucose production in an AMPK-dependent manner. Peripheral insulin action is reduced in type 2 diabetics whereas AMPK signaling remains largely intact. This paper firstly reviews AMPK and its role in glucose uptake and then focuses on a novel mechanism known to operate via an insulin-dependent pathway. Inositol hexakisphosphate (IP6) kinase 1 (IP6K1) produces a pyrophosphate group at the position of IP6 to generate a further inositol pyrophosphate, ie, diphosphoinositol pentakisphosphate (IP7). IP7 binds with Akt/PKB at its pleckstrin homology domain, preventing interaction with phosphatidylinositol 3,4,5-trisphosphate, and therefore reducing Akt/PKB membrane translocation and insulin-stimulated glucose uptake. Novel evidence suggesting a reduction in IP7 production via IP6K1 inhibition represents an exciting therapeutic avenue in the treatment of insulin resistance. Metformin-induced activation of AMPK is a key current intervention in the management of type 2 diabetes. However, this treatment does not seem to improve peripheral insulin resistance. In light of this evidence, we suggest that inhibition of IP6K1 may increase insulin sensitivity and provide a novel research direction in the treatment of insulin resistance.
Collapse
Affiliation(s)
- Richard WA Mackenzie
- Department of Human and Health Sciences, Facility of Science and Technology, University of Westminster, London, UK
- Correspondence: Richard WA Mackenzie, Department of Human and Health Sciences, Facility of Science and Technology, University of Westminster, 115 New Cavendish St, London W1W 6UW, UK, Tel +44 020 7911 5000 ext 3811, Email
| | - Bradley T Elliott
- Department of Human and Health Sciences, Facility of Science and Technology, University of Westminster, London, UK
| |
Collapse
|
100
|
Abstract
GLUT4 is regulated by its intracellular localization. In the absence of insulin, GLUT4 is efficiently retained intracellularly within storage compartments in muscle and fat cells. Upon insulin stimulation (and contraction in muscle), GLUT4 translocates from these compartments to the cell surface where it transports glucose from the extracellular milieu into the cell. Its implication in insulin-regulated glucose uptake makes GLUT4 not only a key player in normal glucose homeostasis but also an important element in insulin resistance and type 2 diabetes. Nevertheless, how GLUT4 is retained intracellularly and how insulin acts on this retention mechanism is largely unclear. In this review, the current knowledge regarding the various molecular processes that govern GLUT4 physiology is discussed as well as the questions that remain.
Collapse
|